1
|
Sadighi A, Aghamohammadpour Z, Sadeghpour Heravi F, Somi MH, Masnadi Shirazi Nezhad K, Hosseini S, Bahman Soufiani K, Ebrahimzadeh Leylabadlo H. The protective effects of Helicobacter pylori: A comprehensive review. JOURNAL OF RESEARCH IN CLINICAL MEDICINE 2024; 12:17. [DOI: 10.34172/jrcm.34509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2025] Open
Abstract
Previous reports have estimated that approximately half of the world’s population is infected with Helicobacter pylori, the most prevalent infectious agent responsible for gastrointestinal illnesses. Due to the life-threatening effects of H. pylori infections, numerous studies have focused on developing medical therapies for H. pylori infections, while the commensal relationship and positive impacts of this bacterium on overall human health have been largely overlooked. The inhibitory efficacy of H. pylori on the progression of several chronic inflammatory disorders and gastrointestinal diseases has recently raised concerns about whether this bacterium should be eradicated in affected individuals or maintained in an appropriate balance depending on the patient’s condition. This review investigates the beneficial effects of H. pylori in preventing various diseases and discusses the potential association of conditions such as inflammatory disorders with the absence of H. pylori.
Collapse
Affiliation(s)
- Ali Sadighi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Aghamohammadpour
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Samaneh Hosseini
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Katayoun Bahman Soufiani
- Department of Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | | |
Collapse
|
2
|
Yang M, Diaz F, Krause ART, Lei Y, Liu WS. Synergistic enhancement of the mouse Pramex1 and Pramel1 in repressing retinoic acid (RA) signaling during gametogenesis. Cell Biosci 2024; 14:28. [PMID: 38395975 PMCID: PMC10893636 DOI: 10.1186/s13578-024-01212-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND PRAME constitutes one of the largest multi-copy gene families in Eutherians, encoding cancer-testis antigens (CTAs) with leucine-rich repeats (LRR) domains, highly expressed in cancer cells and gametogenic germ cells. This study aims to elucidate genetic interactions between two members, Pramex1 and Pramel1, in the mouse Prame family during gametogenesis using a gene knockout approach. RESULT Single-gene knockout (sKO) of either Pramex1 or Pramel1 resulted in approximately 7% of abnormal seminiferous tubules, characterized by a Sertoli-cell only (SCO) phenotype, impacting sperm count and fecundity significantly. Remarkably, sKO female mice displayed normal reproductive functions. In contrast, Pramex1/Pramel1 double knockout (dKO) mice exhibited reduced fecundity in both sexes. In dKO females, ovarian primary follicle count decreased by 50% compared to sKO and WT mice, correlating with a 50% fecundity decrease. This suggested compensatory roles during oogenesis in Pramex1 or Pramel1 sKO females. Conversely, dKO males showed an 18% frequency of SCO tubules, increased apoptotic germ cells, and decreased undifferentiated spermatogonia compared to sKO and WT testes. Western blot analysis with PRAMEX1- or PRAMEL1-specific antibodies on sKO testes revealed compensatory upregulation of each protein (30-50%) in response to the other gene's deletion. Double KO males exhibited more severe defects in sperm count and litter size, surpassing Pramex1 and Pramel1 sKO accumulative effects, indicating a synergistic enhancement interaction during spermatogenesis. Additional experiments administering trans-retinoic acid (RA) and its inhibitor (WIN18,446) in sKO, dKO, and WT mice suggested that PRAMEX1 and PRAMEL1 synergistically repress the RA signaling pathway during spermatogenesis. CONCLUSION Data from sKO and dKO mice unveil a synergistic interaction via the RA signaling pathway between Pramex1 and Pramel1 genes during gametogenesis. This discovery sets the stage for investigating interactions among other members within the Prame family, advancing our understanding of multi-copy gene families involved in germ cell formation and function.
Collapse
Affiliation(s)
- Mingyao Yang
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Francisco Diaz
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Ana Rita T Krause
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Yuguo Lei
- Department of Biomedical Engineering, College of Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Wan-Sheng Liu
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA.
| |
Collapse
|
3
|
Rodrigues E-Lacerda R, Fang H, Robin N, Bhatwa A, Marko DM, Schertzer JD. Microbiota and Nod-like receptors balance inflammation and metabolism during obesity and diabetes. Biomed J 2023; 46:100610. [PMID: 37263539 PMCID: PMC10505681 DOI: 10.1016/j.bj.2023.100610] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/03/2023] Open
Abstract
Gut microbiota influence host immunity and metabolism during obesity. Bacterial sensors of the innate immune system relay signals from specific bacterial components (i.e., postbiotics) that can have opposing outcomes on host metabolic inflammation. NOD-like receptors (NLRs) such as Nod1 and Nod2 both recruit receptor-interacting protein kinase 2 (RIPK2) but have opposite effects on blood glucose control. Nod1 connects bacterial cell wall-derived signals to metabolic inflammation and insulin resistance, whereas Nod2 can promote immune tolerance, insulin sensitivity, and better blood glucose control during obesity. NLR family pyrin domain containing (NLRP) inflammasomes can also generate divergent metabolic outcomes. NLRP1 protects against obesity and metabolic inflammation potentially because of a bias toward IL-18 regulation, whereas NLRP3 appears to have a bias toward IL-1β-mediated metabolic inflammation and insulin resistance. Targeting specific postbiotics that improve immunometabolism is a key goal. The Nod2 ligand, muramyl dipeptide (MDP) is a short-acting insulin sensitizer during obesity or during inflammatory lipopolysaccharide (LPS) stress. LPS with underacylated lipid-A antagonizes TLR4 and counteracts the metabolic effects of inflammatory LPS. Providing underacylated LPS derived from Rhodobacter sphaeroides improved insulin sensitivity in obese mice. Therefore, certain types of LPS can generate metabolically beneficial metabolic endotoxemia. Engaging protective adaptive immunoglobulin immune responses can also improve blood glucose during obesity. A bacterial vaccine approach using an extract of the entire bacterial community in the upper gut promotes protective adaptive immune response and long-lasting improvements in blood glucose control. A key future goal is to identify and combine postbiotics that cooperate to improve blood glucose control.
Collapse
Affiliation(s)
- Rodrigo Rodrigues E-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Nazli Robin
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Arshpreet Bhatwa
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Daniel M Marko
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Guzelj S, Jakopin Ž. Nucleotide-Binding Oligomerization Domain 1/Toll-Like Receptor 4 Co-Engagement Promotes Non-Specific Immune Response Against K562 Cancer Cells. Front Pharmacol 2022; 13:920928. [PMID: 35935855 PMCID: PMC9354050 DOI: 10.3389/fphar.2022.920928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022] Open
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1) receptor and Toll-like receptor 4 (TLR4) belong to the family of pattern recognition receptors. Interactions between these receptors profoundly shape the innate immune responses. We previously demonstrated that co-stimulation of peripheral blood mononuclear cells (PBMCs) with D-glutamyl-meso-diaminopimelic acid (iE-DAP)-based NOD1 agonists and lipopolysaccharide (LPS), a TLR4 agonist, synergistically increased the cytokine production. Herein, we postulate that stimulation of NOD1 alone or a combined stimulation of NOD1 and TLR4 could also strengthen PBMC-mediated cytotoxicity against cancer cells. Initially, an in-house library of iE-DAP analogs was screened for NOD1 agonist activity to establish their potency in HEK-Blue NOD1 cells. Next, we showed that our most potent NOD1 agonist SZZ-38 markedly enhanced the LPS-induced cytokine secretion from PBMCs, in addition to PBMC- and natural killer (NK) cell-mediated killing of K562 cancer cells. Activation marker analysis revealed that the frequencies of CD69+, CD107a+, and IFN-γ+ NK cells are significantly upregulated following NOD1/TLR4 co-stimulation. Of note, SZZ-38 also enhanced the IFN-γ-induced PBMC cytotoxicity. Overall, our findings provide further insight into how co-engagement of two pathways boosts the non-specific immune response and attest to the importance of such interplay between NOD1 and TLR4.
Collapse
|
5
|
Jastrząb R, Graczyk D, Siedlecki P. Molecular and Cellular Mechanisms Influenced by Postbiotics. Int J Mol Sci 2021; 22:ijms222413475. [PMID: 34948270 PMCID: PMC8707144 DOI: 10.3390/ijms222413475] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, commensal bacteria colonizing the human body have been recognized as important determinants of health and multiple pathologic conditions. Among the most extensively studied commensal bacteria are the gut microbiota, which perform a plethora of functions, including the synthesis of bioactive products, metabolism of dietary compounds, and immunomodulation, both through attenuation and immunostimulation. An imbalance in the microbiota population, i.e., dysbiosis, has been linked to many human pathologies, including various cancer types and neurodegenerative diseases. Targeting gut microbiota and microbiome-host interactions resulting from probiotics, prebiotics, and postbiotics is a growing opportunity for the effective treatment of various diseases. As more research is being conducted, the microbiome field is shifting from simple descriptive analysis of commensal compositions to more molecular, cellular, and functional studies. Insight into these mechanisms is of paramount importance for understanding and modulating the effects that microbiota, probiotics, and their derivatives exert on host health.
Collapse
|
6
|
Gorskaya YF, Semenova EN, Nagurskaya EV, Bekhalo VA, Nesterenko VG. Simultaneous Administration of NOD-2 (MDP) and TLP-4 (LPS) Ligands to Bone Marrow Donors 24 h before Transplantation Increases the Content of Multipotent Stromal Cells (MSCs) in Bone Marrow Grafts in CBA Mice Compared to the Total Result of Their Isolated Administration. Bull Exp Biol Med 2021; 172:175-179. [PMID: 34853967 PMCID: PMC8635759 DOI: 10.1007/s10517-021-05358-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Indexed: 12/03/2022]
Abstract
In 3-month bone marrow transplants of CBA mice from bone marrow donors receiving single injections of TLR-4 ligand (LPS) or NOD-2 ligand (muramyl dipeptide, MDP) 24 h before transplantation, an increase in the total number of MSCs (by 2.6 and 1.9 times, respectively), as well as a slight increase in the number of nuclear cells and the mass of bone capsules (by 1.3 and 1.2 times) were observed. After combined administration of MDР and LPS to donors, the total content of MSCs in the grafts was higher by 1.6 times in comparison with the total result of their isolated administration (and by 7.2 times in comparison with the control). At the same time, the concentration of osteogenic MSCs in the grafts of all groups was almost the same and corresponded to the control level. The number of nuclear cells and the mass of bone capsules of the grafts after combined administration of LPS and MDP were close (~80%) to the sum of the results of their isolated administration. These findings suggest that activation of the stromal tissue and the success of bone marrow transplantation depend on the intensity of innate immune responses. These data can be useful for the development of optimal methods of tissue transplantation.
Collapse
Affiliation(s)
- Yu F Gorskaya
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - E N Semenova
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E V Nagurskaya
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V A Bekhalo
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V G Nesterenko
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
7
|
McDaniel MM, Meibers HE, Pasare C. Innate control of adaptive immunity and adaptive instruction of innate immunity: bi-directional flow of information. Curr Opin Immunol 2021; 73:25-33. [PMID: 34425435 PMCID: PMC8648974 DOI: 10.1016/j.coi.2021.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/14/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022]
Abstract
The ability of the innate and adaptive immune systems to communicate with each other is central to protective immune responses and maintenance of host health. Myeloid cells of the innate immune system are able to sense microbial ligands, perturbations in cellular homeostasis, and virulence factors, thereby allowing them to relay distinct pathogen-specific information to naïve T cells in the form of pathogen-derived peptides and a unique cytokine milieu. Once primed, effector T helper cells produce lineage-defining cytokines to help combat the original pathogen, and a subset of these cells persist as memory or effector-memory populations. These memory T cells then play a dual role in host protection by not only responding rapidly to reinfection, but by also directly instructing myeloid cells to express licensing cytokines. This means there is a bi-directional flow of information first from the innate to the adaptive immune system, and then from the adaptive back to innate immune system. Here, we focus on how signals, first from pathogens and then from primed effector and memory T cells, are integrated by myeloid cells and its consequences for protective immunity or systemic inflammation.
Collapse
Affiliation(s)
- Margaret M McDaniel
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Hannah E Meibers
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, United States
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45220, United States.
| |
Collapse
|
8
|
Budikhina AS, Murugina NE, Maximchik PV, Dagil YA, Nikolaeva AM, Balyasova LS, Murugin VV, Selezneva EM, Pashchenkova YG, Chkadua GZ, Pinegin BV, Pashenkov MV. Interplay between NOD1 and TLR4 Receptors in Macrophages: Nonsynergistic Activation of Signaling Pathways Results in Synergistic Induction of Proinflammatory Gene Expression. THE JOURNAL OF IMMUNOLOGY 2021; 206:2206-2220. [PMID: 33846227 DOI: 10.4049/jimmunol.2000692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/21/2021] [Indexed: 11/19/2022]
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 IKKβ 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.
Collapse
Affiliation(s)
- Anna S Budikhina
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Nina E Murugina
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Polina V Maximchik
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Yulia A Dagil
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Anna M Nikolaeva
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia.,Biological Faculty, Lomonosov Moscow State University, Moscow, Russia; and
| | - Lyudmila S Balyasova
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Vladimir V Murugin
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | | | - Yulia G Pashchenkova
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Georgy Z Chkadua
- Laboratory of Experimental Diagnostics and Biotherapy of Tumors, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Boris V Pinegin
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Mikhail V Pashenkov
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia;
| |
Collapse
|
9
|
Ngou BPM, Ahn HK, Ding P, Jones JDG. Mutual potentiation of plant immunity by cell-surface and intracellular receptors. Nature 2021; 592:110-115. [PMID: 33692545 DOI: 10.1101/2020.04.10.034173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 02/01/2021] [Indexed: 05/25/2023]
Abstract
The plant immune system involves cell-surface receptors that detect intercellular pathogen-derived molecules, and intracellular receptors that activate immunity upon detection of pathogen-secreted effector proteins that act inside the plant cell. Immunity mediated by surface receptors has been extensively studied1, but that mediated by intracellular receptors has rarely been investigated in the absence of surface-receptor-mediated immunity. Furthermore, interactions between these two immune pathways are poorly understood. Here, by activating intracellular receptors without inducing surface-receptor-mediated immunity, we analyse interactions between these two distinct immune systems in Arabidopsis. Pathogen recognition by surface receptors activates multiple protein kinases and NADPH oxidases, and we find that intracellular receptors primarily potentiate the activation of these proteins by increasing their abundance through several mechanisms. Likewise, the hypersensitive response that depends on intracellular receptors is strongly enhanced by the activation of surface receptors. Activation of either immune system alone is insufficient to provide effective resistance against the bacterial pathogen Pseudomonas syringae. Thus, immune pathways activated by cell-surface and intracellular receptors in plants mutually potentiate to activate strong defences against pathogens. These findings reshape our understanding of plant immunity and have broad implications for crop improvement.
Collapse
Affiliation(s)
| | - Hee-Kyung Ahn
- The Sainsbury Laboratory, University of East Anglia, Norwich, UK
| | - Pingtao Ding
- The Sainsbury Laboratory, University of East Anglia, Norwich, UK.
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands.
| | | |
Collapse
|
10
|
Guo HW, Chang J, Wang P, Yin QQ, Liu CQ, Xu XX, Dang XW, Hu XF, Wang QL. Effects of compound probiotics and aflatoxin-degradation enzyme on alleviating aflatoxin-induced cytotoxicity in chicken embryo primary intestinal epithelium, liver and kidney cells. AMB Express 2021; 11:35. [PMID: 33646441 PMCID: PMC7921234 DOI: 10.1186/s13568-021-01196-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Aflatoxin B1 (AFB1) is one of the most dangerous mycotoxins for humans and animals. This study aimed to investigate the effects of compound probiotics (CP), CP supernatant (CPS), AFB1-degradation enzyme (ADE) on chicken embryo primary intestinal epithelium, liver and kidney cell viabilities, and to determine the functions of CP + ADE (CPADE) or CPS + ADE (CPSADE) for alleviating cytotoxicity induced by AFB1. The results showed that AFB1 decreased cell viabilities in dose-dependent and time-dependent manners. The optimal AFB1 concentrations and reactive time for establishing cell damage models were 200 µg/L AFB1 and 12 h for intestinal epithelium cells, 40 µg/L and 12 h for liver and kidney cells. Cell viabilities reached 231.58% (p < 0.05) for intestinal epithelium cells with CP addition, 105.29% and 115.84% (p < 0.05) for kidney and liver cells with CPS additions. The further results showed that intestinal epithelium, liver and kidney cell viabilities were significantly decreased to 87.12%, 88.7% and 84.19% (p < 0.05) when the cells were exposed to AFB1; however, they were increased to 93.49% by CPADE addition, 102.33% and 94.71% by CPSADE additions (p < 0.05). The relative mRNA abundances of IL-6, IL-8, TNF-α, iNOS, NF-κB, NOD1 (except liver cell) and TLR2 in three kinds of primary cells were significantly down-regulated by CPADE or CPSADE addition, compared with single AFB1 group (p < 0.05), indicating that CPADE or CPSADE addition could alleviate cell cytotoxicity and inflammation induced by AFB1 exposure through suppressing the activations of NF-κB, iNOS, NOD1 and TLR2 pathways.
Collapse
Affiliation(s)
- Hong-Wei Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Juan Chang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Ping Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Qing-Qiang Yin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Chao-Qi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xiao-Xiang Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xiao-Wei Dang
- Henan Delin Biological Product Co., Ltd, Xinxiang, 453000, China
| | - Xiao-Fei Hu
- Henan Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Quan-Liang Wang
- Henan Guangan Biotechnological Co., Ltd., Zhengzhou, 450001, China
| |
Collapse
|
11
|
Zuo ZT, Ma Y, Sun Y, Bai CQ, Ling CH, Yuan FL. The Protective Effects of Helicobacter pylori Infection on Allergic Asthma. Int Arch Allergy Immunol 2020; 182:53-64. [PMID: 33080611 DOI: 10.1159/000508330] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
As an ancient Gram-negative bacterium, Helicobacter pylori has settled in human stomach. Eradicating H. pylori increases the morbidities of asthma and other allergic diseases. Therefore, H. pylori might play a protective role against asthma. The "disappearing microbiota" hypothesis suggests that the absence of certain types of the ancestral microbiota could change the development of immunology, metabolism, and cognitive ability in our early life, contributing to the development of some diseases. And the Hygiene Hypothesis links early environmental and microbial exposure to the prevalence of atopic allergies and asthma. Exposure to the environment and microbes can influence the growing immune system and protect subsequent immune-mediated diseases. H. pylori can inhibit allergic asthma by regulating the ratio of helper T cells 1/2 (Th1/Th2), Th17/regulatory T cells (Tregs), etc. H. pylori can also target dendritic cells to promote immune tolerance and enhance the protective effect on allergic asthma, and this effect relies on highly suppressed Tregs. The remote regulation of lung immune function by H. pylori is consistent with the gut-lung axis theory. Perhaps, H. pylori also protects against asthma by altering levels of stomach hormones, affecting the autonomic nervous system and lowering the expression of heat shock protein 70. Therapeutic products from H. pylori may be used to prevent and treat asthma. This paper reviews the possible protective influence of H. pylori on allergic asthma and the possible application of H. pylori in treating asthma.
Collapse
Affiliation(s)
- Zhi Tong Zuo
- Department of Respiratory Disease, The Hospital Affiliated to Jiangnan University, Wuxi, China,
| | - Ya Ma
- Wuxi Medical College of Jiangnan University, Wuxi, China
| | - Yan Sun
- Department of Respiratory Disease, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Cui Qing Bai
- Department of Respiratory Disease, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Chun Hua Ling
- Department of Respiratory Disease, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Lai Yuan
- Department of Orthopaedics and Central Laboratory, The Hospital Affiliated to Jiangnan University, Wuxi, China
| |
Collapse
|
12
|
Maeyama JI, Kurata-Iesato Y, Isaka M, Komiya T, Sakurai S. Induction of antibody responses in mice immunized intranasally with Type I interferon as adjuvant and synergistic effect of chitosan. Microbiol Immunol 2020; 64:610-619. [PMID: 32662896 DOI: 10.1111/1348-0421.12832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/24/2020] [Accepted: 07/09/2020] [Indexed: 02/05/2023]
Abstract
Type I IFNs are a range of host-derived molecules with adjuvant potential; they have been used for many years in the treatment of cancer and viral hepatitis. Therefore, the safety of IFNs for human use has been established. In this study, we evaluated the mucosal adjuvanticity of IFN-β administered intranasally to mice with diphtheria toxoid, and suggested a method to improve its adjuvanticity. When IFN-β alone was used as a mucosal adjuvant, no clear results were obtained. However, simultaneous administration of IFN-β and chitosan resulted in an enhancement of the specific serum immunoglobulin G (IgG) and IgA antibody responses, the mucosal IgA antibody response, and antitoxin titers. Furthermore, the intranasal administration of IFN-α alone resulted in a greater increase in antibody titer than IFN-β, and a synergistic effect with chitosan was also observed. These findings suggest that intranasal administration of chitosan and Type I IFNs may display an effective synergistic mucosal adjuvant activity.
Collapse
Affiliation(s)
- Jun-Ichi Maeyama
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuko Kurata-Iesato
- Department of Pharmaceutical Quality Assurance, Toray Industries Inc., Mishima, Japan
| | - Masanori Isaka
- Department of Microbiology, Nagoya City University Medical School, Nagoya, Japan
| | - Takako Komiya
- Department of Bacterial Pathogenesis and Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shingou Sakurai
- Faculty of Pharmacy, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
13
|
Tukhvatulin AI, Dzharullaeva AS, Erokhova AS, Scheblyakov DV, Naroditsky BS, Gintsburg AL, Logunov DY. NOD1/2 and the C-Type Lectin Receptors Dectin-1 and Mincle Synergistically Enhance Proinflammatory Reactions Both In Vitro and In Vivo. J Inflamm Res 2020; 13:357-368. [PMID: 32801829 PMCID: PMC7383029 DOI: 10.2147/jir.s245638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 06/20/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose Pathogens consist of a wide variety of evolutionarily conserved molecular structures that are recognized by pattern recognition receptors (PRRs) of innate immunity. Reasonably assuming that no single PRR is ever likely to be the sole trigger of the immune response during infection, a great deal remains unknown about collaborative mechanisms and consequential crosstalk effects between multiple PRRs belonging to different families. Here, we aimed to investigate inflammatory response to combined stimulation of cytosolic nucleotide-binding oligomerization domain (NOD) receptors: NOD1, NOD2 and membrane-bound C-type lectin receptors (CLRs): Mincle and Dectin-1 in comparison to individual stimulation both in vitro and in vivo. Materials and Methods For in vitro studies, we used human monocytic THP-1 cells endogenously expressing NOD1,2, as well as Mincle and Dectin-1 receptors. Using reporter gene and immunoassay approaches, we measured activity of key proinflammatory transcription factors (NF-κB and AP-1) and cytokine production after addition of specific PRR agonists or their pairwise combinations. In vivo NF-κB activity (bioluminescent detection in NF-κB-Luc transgenic mice), as well as cytokine levels in mouse blood serum, was measured 3 hours after intramuscular injection of PRR agonists. Results We detected that combined stimulation of NOD1/2 and C-type lectin receptors (Dectin-1, Mincle) strongly potentiates NF-κB and AP-1 transcription factor activity in human monocytic THP-1 cells, as well as resulting in enhanced levels of IL-8 cytokine production. We demonstrated that RIP2- and Syk-dependent signaling pathways downstream of NOD1/2 and Dectin-1/Mincle, respectively, are essential for the potentiated proinflammatory cell response. Lastly, we confirmed that synergy between NOD and C-type lectin receptors resulting in potentiated levels of NF-κB activation and cytokine (IL-6, KC) production also occurs in vivo. Conclusion These findings originally indicate cooperation between NODs and CLRs, leading to potentiated levels of proinflammatory immune response both in vitro and in vivo.
Collapse
Affiliation(s)
- Amir I Tukhvatulin
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| | - Alina S Dzharullaeva
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| | - Alina S Erokhova
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| | - Dmitry V Scheblyakov
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| | - Boris S Naroditsky
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| | - Alexander L Gintsburg
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| | - Denis Y Logunov
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
14
|
Combined Administration of TLR4 (LPS) and TLR3 (Poly I:C) Ligands to CBA Mice Elevates the Content of Osteogenic MSC by 1.6 Times and Increases Content of Bone Marrow MSC to Intermediate Level between Values Attained by Their Individual Administration. Bull Exp Biol Med 2020; 168:767-772. [PMID: 32328942 DOI: 10.1007/s10517-020-04798-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Indexed: 12/22/2022]
Abstract
In 1 and 24 h after combined administration of TLR4 (LPS) and TLR3 (Poly I:C) ligands to CBA mice, the content of MSC in bone marrow increased to intermediate value between the levels attained by their individual injections. The content of osteogenic MSC assessed in 24 h postinjection corresponded to the control level in Poly I:C group, decreased in LPS group by 2.5 times relatively to the control, and increased by 1.6 times (relatively to control) after combined administration of the ligands. In 3 h after combined addition of LPS and Poly I:C in vitro to 12-day-old primary culture of mouse bone marrow stromal cells, the concentration of TNFα in culture medium was intermediate between the levels attained by their individual application. The data revealed dependence of activation of stromal tissue on intensity of innate immunity reactions; they also attested to marked elevation of osteogenicity of MSC pool after costimulation with Poly I:C and LPS, which can underlie augmented calcification of the tissues during combined viral and bacterial infections.
Collapse
|
15
|
Kasimsetty SG, Hawkes A, Barekatain K, Soo E, Welch AK, McKay DB. TLR2 and NODs1 and 2 cooperate in inflammatory responses associated with renal ischemia reperfusion injury. Transpl Immunol 2020; 58:101260. [PMID: 31760144 PMCID: PMC7041897 DOI: 10.1016/j.trim.2019.101260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/06/2019] [Accepted: 11/19/2019] [Indexed: 11/21/2022]
Abstract
Pattern recognition receptors (PRRs) are potent triggers of tissue injury following renal ischemia/reperfusion injury (IRI). Specific PRRs, such as the toll-like receptor 2 (TLR2) and the nucleotide-binding oligomerization domain-like receptors (NLRs) NOD1 and NOD2 are promising targets to abrogate inflammatory injury associated with renal IRI. Several recent reports have shown there is crosstalk between TLRs and NODs, which might boost inflammatory responses to tissue injury. This study examined the relative roles of TLR2 and NODs 1 and 2 in activation of myeloid cells that contribute to inflammation after renal IRI. We found that TLR2 and NOD1 and 2 signaling induces neutrophil, macrophage and dendritic cell migration in vitro, however their blockade only decreases neutrophil infiltration into ischemic kidneys. The results of this study suggest that future therapies targeted to innate immune blockade should consider that either TLR2 or NOD1/2 blockade could decrease neutrophil inflammation following an ischemic insult to the kidney, however blockade of these PRRs would not likely impact infiltration of dendritic cells or macrophages. Developing rational approaches that target innate immunity in IRI-induced acute kidney injury requires an understanding of the relative role of PRRs in directing inflammation in the kidney.
Collapse
Affiliation(s)
- Sashi G Kasimsetty
- Department of Immunology and Microbial Sciences, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America
| | - Alana Hawkes
- Department of Immunology and Microbial Sciences, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America; Scripps Clinic and Green Hospital, Division of Transplantation, 10660 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America
| | - Kayvan Barekatain
- University of California San Diego, Department of Medicine, Division of Nephrology and Hypertension, 9500 Gilman Drive, La Jolla, CA 92093, United States of America
| | - Elizabeth Soo
- Department of Immunology and Microbial Sciences, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America; Scripps Clinic and Green Hospital, Division of Transplantation, 10660 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America
| | - Alexander K Welch
- Department of Immunology and Microbial Sciences, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America; University of California San Diego, Department of Medicine, Division of Nephrology and Hypertension, 9500 Gilman Drive, La Jolla, CA 92093, United States of America
| | - Dianne B McKay
- Department of Immunology and Microbial Sciences, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America; Scripps Clinic and Green Hospital, Division of Transplantation, 10660 N. Torrey Pines Rd, La Jolla, CA 92037, United States of America.
| |
Collapse
|
16
|
Guzelj S, Gobec M, Urbančič D, Mlinarič-Raščan I, Corsini E, Jakopin Ž. Structural features and functional activities of benzimidazoles as NOD2 antagonists. Eur J Med Chem 2020; 190:112089. [PMID: 32014680 DOI: 10.1016/j.ejmech.2020.112089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
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.
Collapse
Affiliation(s)
- Samo Guzelj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Martina Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Dunja Urbančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Irena Mlinarič-Raščan
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Environmental Science and Policy, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
17
|
Jakopin Ž, Corsini E. THP-1 Cells and Pro-inflammatory Cytokine Production: An in Vitro Tool for Functional Characterization of NOD1/NOD2 Antagonists. Int J Mol Sci 2019; 20:ijms20174265. [PMID: 31480368 PMCID: PMC6747088 DOI: 10.3390/ijms20174265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023] Open
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.
Collapse
Affiliation(s)
- Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Pharmacological Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
18
|
Haile PA, Casillas LN, Votta BJ, Wang GZ, Charnley AK, Dong X, Bury MJ, Romano JJ, Mehlmann JF, King BW, Erhard KF, Hanning CR, Lipshutz DB, Desai BM, Capriotti CA, Schaeffer MC, Berger SB, Mahajan MK, Reilly MA, Nagilla R, Rivera EJ, Sun HH, Kenna JK, Beal AM, Ouellette MT, Kelly M, Stemp G, Convery MA, Vossenkämper A, MacDonald TT, Gough PJ, Bertin J, Marquis RW. Discovery of a First-in-Class Receptor Interacting Protein 2 (RIP2) Kinase Specific Clinical Candidate, 2-((4-(Benzo[ d]thiazol-5-ylamino)-6-( tert-butylsulfonyl)quinazolin-7-yl)oxy)ethyl Dihydrogen Phosphate, for the Treatment of Inflammatory Diseases. J Med Chem 2019; 62:6482-6494. [PMID: 31265286 DOI: 10.1021/acs.jmedchem.9b00575] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
RIP2 kinase has been identified as a key signal transduction partner in the NOD2 pathway contributing to a variety of human pathologies, including immune-mediated inflammatory diseases. Small-molecule inhibitors of RIP2 kinase or its signaling partners on the NOD2 pathway that are suitable for advancement into the clinic have yet to be described. Herein, we report our discovery and profile of the prodrug clinical compound, inhibitor 3, currently in phase 1 clinical studies. Compound 3 potently binds to RIP2 kinase with good kinase specificity and has excellent activity in blocking many proinflammatory cytokine responses in vivo and in human IBD explant samples. The highly favorable physicochemical and ADMET properties of 3 combined with high potency led to a predicted low oral dose in humans.
Collapse
Affiliation(s)
- Pamela A Haile
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Linda N Casillas
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Bartholomew J Votta
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Gren Z Wang
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Adam K Charnley
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Xiaoyang Dong
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Michael J Bury
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Joseph J Romano
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - John F Mehlmann
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Bryan W King
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Karl F Erhard
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Charles R Hanning
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - David B Lipshutz
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Biva M Desai
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Carol A Capriotti
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Michelle C Schaeffer
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Scott B Berger
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Mukesh K Mahajan
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Michael A Reilly
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Rakesh Nagilla
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Elizabeth J Rivera
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Helen H Sun
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - John K Kenna
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Allison M Beal
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Michael T Ouellette
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Mike Kelly
- GlaxoSmithKline , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Gillian Stemp
- GlaxoSmithKline , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Máire A Convery
- GlaxoSmithKline , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Anna Vossenkämper
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry , Queen Mary University of London , London E1 2AD , U.K
| | - Thomas T MacDonald
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry , Queen Mary University of London , London E1 2AD , U.K
| | - Peter J Gough
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - John Bertin
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Robert W Marquis
- GlaxoSmithKline , Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| |
Collapse
|
19
|
Zom GG, Willems MMJHP, Meeuwenoord NJ, Reintjens NRM, Tondini E, Khan S, Overkleeft HS, van der Marel GA, Codee JDC, Ossendorp F, Filippov DV. Dual Synthetic Peptide Conjugate Vaccine Simultaneously Triggers TLR2 and NOD2 and Activates Human Dendritic Cells. Bioconjug Chem 2019; 30:1150-1161. [PMID: 30865430 DOI: 10.1021/acs.bioconjchem.9b00087] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Simultaneous triggering of Toll-like receptors (TLRs) and NOD-like receptors (NLRs) has previously been shown to synergistically activate monocytes, dendritic cells, and macrophages. We applied these properties in a T-cell vaccine setting by conjugating the NOD2-ligand muramyl-dipeptide (MDP) and TLR2-ligand Pam3CSK4 to a synthetic peptide derived from a model antigen. Stimulation of human DCs with the MDP-peptide-Pam3CSK4 conjugate led to a strongly increased secretion of pro-inflammatory and Th1-type cytokines and chemokines. We further show that the conjugated ligands retain their ability to trigger their respective receptors, while even improving NOD2-triggering. Also, activation of murine DCs was enhanced by the dual triggering, ultimately leading to effective induction of vaccine-specific T cells expressing IFNγ, IL-2, and TNFα. Together, these data indicate that the dual MDP-SLP-Pam3CSK4 conjugate constitutes a chemically well-defined vaccine approach that holds promise for the use in the treatment of virus infections and cancer.
Collapse
Affiliation(s)
- Gijs G Zom
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Marian M J H P Willems
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Nico J Meeuwenoord
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Niels R M Reintjens
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Elena Tondini
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Selina Khan
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Herman S Overkleeft
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Gijsbert A van der Marel
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Jeroen D C Codee
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Dmitri V Filippov
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| |
Collapse
|
20
|
Ligands of NOD2 (Muramyl Dipeptide) and TLR4 (LPS) in 24 h after Combined In Vivo Administration Produce a Synergistic Increase in the Content of Multipotent Stromal Cells in the Bone Marrow and Peritoneal Exudate of CBA Mice. Bull Exp Biol Med 2019; 166:473-476. [PMID: 30788737 DOI: 10.1007/s10517-019-04375-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Indexed: 10/27/2022]
Abstract
In 24 h after combined administration of ligands of NOD2 (muramyl dipeptide) and TLR4 (LPS) receptors to CBA mice, a synergistic increase (by 10 times compared to the intact control) in cloning efficiency and content of multipotent stromal cells was observed in the bone marrow in comparison with the total effects of their individual administration (by 2.1 and 4.1 times, respectively). A similar effect was also observed in the peritoneal exudate. When ligands were administered simultaneously, the concentration of osteogenic multipotent stromal cells in the bone marrow decreased to a greater extent than in case of individual injections of the ligands, but did not drop below 7% of the control, which is apparently indicative of a decline threshold. In 3 h after simultaneous addition of the ligands in vitro to 12-day primary cultures of mouse bone marrow stromal cells, a synergistic increase in TNFα concentration was observed (32-fold increase from the level of intact control), while IL-10 concentration did not differ from the control, which is indicative of the proinflammatory nature of the process and the absence of immunosuppressive effect. These results suggest that activation of the stromal tissue depends on the intensity of innate immunity reactions.
Collapse
|
21
|
Pashenkov MV, Murugina NE, Budikhina AS, Pinegin BV. Synergistic interactions between NOD receptors and TLRs: Mechanisms and clinical implications. J Leukoc Biol 2018; 105:669-680. [PMID: 30517768 DOI: 10.1002/jlb.2ru0718-290r] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/23/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Interactions between pattern recognition receptors (PRRs) shape innate immune responses to particular classes of pathogens. Here, we review interactions between TLRs and nucleotide-binding oligomerization domain 1 and 2 (NOD1 and NOD2) receptors, two major groups of PRRs involved in innate recognition of bacteria. Most of experimental data both in vitro and in vivo suggest that NODs and TLRs synergize with each other at inducing the production of cytokines and antimicrobial peptides. Molecular mechanisms of this synergy remain poorly understood, although several scenarios can be proposed: (i) direct interactions of signaling pathways downstream of NODs and TLRs; (ii) mutual transcriptional regulation of unique components of NOD-dependent and TLR-dependent signaling pathways; and (iii) interactions at the post-transcriptional level. Potential practical implications of NOD-TLR synergy are dual. In sepsis, where synergistic effects probably contribute to excessive proinflammatory cytokine production, blockade of NOD1, and/or NOD2 in addition to TLR4 blockade may be required to achieve therapeutic benefit. On the other hand, synergistic combinations of relatively small doses of NOD and TLR agonists administered before infection could be used to boost innate resistance against bacterial pathogens.
Collapse
Affiliation(s)
- Mikhail V Pashenkov
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Nina E Murugina
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Anna S Budikhina
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Boris V Pinegin
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology, Federal Medical-Biological Agency of Russia, Moscow, Russia
| |
Collapse
|
22
|
Haile PA, Casillas LN, Bury MJ, Mehlmann JF, Singhaus R, Charnley AK, Hughes TV, DeMartino MP, Wang GZ, Romano JJ, Dong X, Plotnikov NV, Lakdawala AS, Convery MA, Votta BJ, Lipshutz DB, Desai BM, Swift B, Capriotti CA, Berger SB, Mahajan MK, Reilly MA, Rivera EJ, Sun HH, Nagilla R, LePage C, Ouellette MT, Totoritis RD, Donovan BT, Brown BS, Chaudhary KW, Gough PJ, Bertin J, Marquis RW. Identification of Quinoline-Based RIP2 Kinase Inhibitors with an Improved Therapeutic Index to the hERG Ion Channel. ACS Med Chem Lett 2018; 9:1039-1044. [PMID: 30344914 DOI: 10.1021/acsmedchemlett.8b00344] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/11/2018] [Indexed: 12/25/2022] Open
Abstract
RIP2 kinase was recently identified as a therapeutic target for a variety of autoimmune diseases. We have reported previously a selective 4-aminoquinoline-based RIP2 inhibitor GSK583 and demonstrated its effectiveness in blocking downstream NOD2 signaling in cellular models, rodent in vivo models, and human ex vivo disease models. While this tool compound was valuable in validating the biological pathway, it suffered from activity at the hERG ion channel and a poor PK/PD profile thereby limiting progression of this analog. Herein, we detail our efforts to improve both this off-target liability as well as the PK/PD profile of this series of inhibitors through modulation of lipophilicity and strengthening hinge binding ability. These efforts have led to inhibitor 7, which possesses high binding affinity for the ATP pocket of RIP2 (IC50 = 1 nM) and inhibition of downstream cytokine production in human whole blood (IC50 = 10 nM) with reduced hERG activity (14 μM).
Collapse
Affiliation(s)
- Pamela A. Haile
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Linda N. Casillas
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Michael J. Bury
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - John F. Mehlmann
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Robert Singhaus
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Adam K. Charnley
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Terry V. Hughes
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Michael P. DeMartino
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Gren Z. Wang
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Joseph J. Romano
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Xiaoyang Dong
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Nikolay V. Plotnikov
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Ami S. Lakdawala
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Maire A. Convery
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| | - Bartholomew J. Votta
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - David B. Lipshutz
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Biva M. Desai
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Barbara Swift
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Carol A. Capriotti
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Scott B. Berger
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Mukesh K. Mahajan
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Michael A. Reilly
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Elizabeth J. Rivera
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Helen H. Sun
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Rakesh Nagilla
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Carol LePage
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Michael T. Ouellette
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Rachel D. Totoritis
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Brian T. Donovan
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Barry S. Brown
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Khuram W. Chaudhary
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Peter J. Gough
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - John Bertin
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Robert W. Marquis
- GlaxoSmithKline, Collegeville Road, Collegeville, Pennsylvania 19426, United States
| |
Collapse
|
23
|
Abstract
The innate immune system recognizes microbial products using germline-encoded receptors that initiate inflammatory responses to infection. The bacterial cell wall component peptidoglycan is a prime example of a conserved pathogen-associated molecular pattern (PAMP) for which the innate immune system has evolved sensing mechanisms. Peptidoglycan is a direct target for innate immune receptors and also regulates the accessibility of other PAMPs to additional innate immune receptors. Subtle structural modifications to peptidoglycan can influence the ability of the innate immune system to detect bacteria and can allow bacteria to evade or alter host defences. This Review focuses on the mechanisms of peptidoglycan recognition that are used by mammalian cells and discusses new insights into the role of peptidoglycan recognition in inflammation, metabolism, immune homeostasis and disease.
Collapse
Affiliation(s)
- Andrea J Wolf
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center
| | - David M Underhill
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center.,Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, California 90048, USA
| |
Collapse
|
24
|
NOD1 and NOD2: Molecular targets in prevention and treatment of infectious diseases. Int Immunopharmacol 2017; 54:385-400. [PMID: 29207344 DOI: 10.1016/j.intimp.2017.11.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
Nucleotide-binding oligomerization domain (NOD) 1 and NOD2 are pattern-recognition receptors responsible for sensing fragments of bacterial peptidoglycan known as muropeptides. Stimulation of innate immunity by systemic or local administration of NOD1 and NOD2 agonists is an attractive means to prevent and treat infectious diseases. In this review, we discuss novel data concerning structural features of selective and non-selective (dual) NOD1 and NOD2 agonists, main signaling pathways and biological effects induced by NOD1 and NOD2 stimulation, including induction of pro-inflammatory cytokines, type I interferons and antimicrobial peptides, induction of autophagy, alterations of metabolism. We also discuss interactions between NOD1/NOD2 and Toll-like receptor agonists in terms of synergy and cross-tolerance. Finally, we review available animal data on the role of NOD1 and NOD2 in protection against infections, and discuss how these data could be applied in human infectious diseases.
Collapse
|
25
|
Tartey S, Takeuchi O. Pathogen recognition and Toll-like receptor targeted therapeutics in innate immune cells. Int Rev Immunol 2017; 36:57-73. [PMID: 28060562 DOI: 10.1080/08830185.2016.1261318] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The innate immune system deploys a variety of pattern-recognition receptors (PRRs) which include Toll-like receptors (TLRs), RIG-I-like receptors, NOD-like receptors, and C-type lectin receptors to detect the invasion of pathogens and initiate protective responses. The intercellular and intracellular orchestration of signals from different PRRs, their endogenous or microbial ligands and accessory molecules determine the stimulatory or inhibitory responses. Progressing over the last two decades, considerable research on the molecular mechanisms underlying host-pathogen interactions has led to a paradigm shift of our understanding of TLR signaling in the innate immune system. Given that a significant amount of evidence implicates TLRs in the pathogenesis of immune diseases and cancer, and their activation occurs early in the inflammatory cascade, they are attractive targets for novel therapeutic agents. In this review, we discuss the recent advances in TLR signaling cross talks and the mechanism of pathogen recognition with special emphasis on the role of TLRs in tumor immunity and TLR-targeted therapeutics.
Collapse
Affiliation(s)
- Sarang Tartey
- a Laboratory of Infection and Prevention, Institute for Virus Research, Kyoto University , Kawara-Cho, Sakyo-Ku, Kyoto , Japan.,b AMED-CREST, Japan Agency for Medical Research and Development , Kyoto , Japan
| | - Osamu Takeuchi
- a Laboratory of Infection and Prevention, Institute for Virus Research, Kyoto University , Kawara-Cho, Sakyo-Ku, Kyoto , Japan.,b AMED-CREST, Japan Agency for Medical Research and Development , Kyoto , Japan
| |
Collapse
|
26
|
Philip NH, DeLaney A, Peterson LW, Santos-Marrero M, Grier JT, Sun Y, Wynosky-Dolfi MA, Zwack EE, Hu B, Olsen TM, Rongvaux A, Pope SD, López CB, Oberst A, Beiting DP, Henao-Mejia J, Brodsky IE. Activity of Uncleaved Caspase-8 Controls Anti-bacterial Immune Defense and TLR-Induced Cytokine Production Independent of Cell Death. PLoS Pathog 2016; 12:e1005910. [PMID: 27737018 PMCID: PMC5063320 DOI: 10.1371/journal.ppat.1005910] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/01/2016] [Indexed: 12/29/2022] Open
Abstract
Caspases regulate cell death programs in response to environmental stresses, including infection and inflammation, and are therefore critical for the proper operation of the mammalian immune system. Caspase-8 is necessary for optimal production of inflammatory cytokines and host defense against infection by multiple pathogens including Yersinia, but whether this is due to death of infected cells or an intrinsic role of caspase-8 in TLR-induced gene expression is unknown. Caspase-8 activation at death signaling complexes results in its autoprocessing and subsequent cleavage and activation of its downstream apoptotic targets. Whether caspase-8 activity is also important for inflammatory gene expression during bacterial infection has not been investigated. Here, we report that caspase-8 plays an essential cell-intrinsic role in innate inflammatory cytokine production in vivo during Yersinia infection. Unexpectedly, we found that caspase-8 enzymatic activity regulates gene expression in response to bacterial infection as well as TLR signaling independently of apoptosis. Using newly-generated mice in which caspase-8 autoprocessing is ablated (Casp8DA/DA), we now demonstrate that caspase-8 enzymatic activity, but not autoprocessing, mediates induction of inflammatory cytokines by bacterial infection and a wide variety of TLR stimuli. Because unprocessed caspase-8 functions in an enzymatic complex with its homolog cFLIP, our findings implicate the caspase-8/cFLIP heterodimer in control of inflammatory cytokines during microbial infection, and provide new insight into regulation of antibacterial immune defense. TLR signaling induces expression of key inflammatory cytokines and pro-survival factors that facilitate control of microbial infection. TLR signaling can also engage cell death pathways through activation of enzymes known as caspases. Caspase-8 activates apoptosis in response to infection by pathogens that interfere with NF-κB signaling, including Yersinia, but has also recently been linked to control of inflammatory gene expression. Pathogenic Yersinia can cause severe disease ranging from gastroenteritis to plague. While caspase-8 mediates cell death in response to Yersinia infection as well as other signals, its precise role in gene expression and host defense during in vivo infection is unknown. Here, we show that caspase-8 activity promotes cell-intrinsic cytokine expression, independent of its role in cell death in response to Yersinia infection. Our studies further demonstrate that caspase-8 enzymatic activity plays a previously undescribed role in ensuring optimal TLR-induced gene expression by innate cells during bacterial infection. This work sheds new light on mechanisms that regulate essential innate anti-bacterial immune defense.
Collapse
Affiliation(s)
- Naomi H. Philip
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
| | - Alexandra DeLaney
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
| | - Lance W. Peterson
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
| | - Melanie Santos-Marrero
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
| | - Jennifer T. Grier
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
| | - Yan Sun
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
| | - Meghan A. Wynosky-Dolfi
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
| | - Erin E. Zwack
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
| | - Baofeng Hu
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
| | - Tayla M. Olsen
- University of Washington, Department of Immunology, Seattle, Washington, United States of America
| | - Anthony Rongvaux
- Fred Hutchinson Cancer Research Center, Clinical Research Division and Program in Immunology, Seattle, Washington, United States of America
| | - Scott D. Pope
- Yale University School of Medicine, Department of Immunobiology, New Haven, Connecticut, United States of America
| | - Carolina B. López
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
| | - Andrew Oberst
- University of Washington, Department of Immunology, Seattle, Washington, United States of America
| | - Daniel P. Beiting
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
| | - Jorge Henao-Mejia
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania and Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Igor E. Brodsky
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Institute for Immunology, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ag85A DNA Vaccine Delivery by Nanoparticles: Influence of the Formulation Characteristics on Immune Responses. Vaccines (Basel) 2016; 4:vaccines4030032. [PMID: 27626449 PMCID: PMC5041026 DOI: 10.3390/vaccines4030032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/16/2016] [Accepted: 09/05/2016] [Indexed: 11/16/2022] Open
Abstract
The influence of DNA vaccine formulations on immune responses in combination with adjuvants was investigated with the aim to increase cell-mediated immunity against plasmid DNA (pDNA) encoding Mycobacterium tuberculosis antigen 85A. Different ratios of pDNA with cationic trimethyl chitosan (TMC) nanoparticles were characterized for their morphology and physicochemical characteristics (size, zeta potential, loading efficiency and pDNA release profile) applied in vitro for cellular uptake studies and in vivo, to determine the dose-dependent effects of pDNA on immune responses. A selected pDNA/TMC nanoparticle formulation was optimized by the incorporation of muramyl dipeptide (MDP) as an immunostimulatory agent. Cellular uptake investigations in vitro showed saturation to a maximum level upon the increase in the pDNA/TMC nanoparticle ratio, correlating with increasing Th1-related antibody responses up to a definite pDNA dose applied. Moreover, TMC nanoparticles induced clear polarization towards a Th1 response, indicated by IgG2c/IgG1 ratios above unity and enhanced numbers of antigen-specific IFN-γ producing T-cells in the spleen. Remarkably, the incorporation of MDP in TMC nanoparticles provoked a significant additional increase in T-cell-mediated responses induced by pDNA. In conclusion, pDNA-loaded TMC nanoparticles are capable of provoking strong Th1-type cellular and humoral immune responses, with the potential to be further optimized by the incorporation of MDP.
Collapse
|
28
|
Tukhvatulin AI, Dzharullaeva AS, Tukhvatulina NM, Shcheblyakov DV, Shmarov MM, Dolzhikova IV, Stanhope-Baker P, Naroditsky BS, Gudkov AV, Logunov DY, Gintsburg AL. Powerful Complex Immunoadjuvant Based on Synergistic Effect of Combined TLR4 and NOD2 Activation Significantly Enhances Magnitude of Humoral and Cellular Adaptive Immune Responses. PLoS One 2016; 11:e0155650. [PMID: 27187797 PMCID: PMC4871337 DOI: 10.1371/journal.pone.0155650] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/02/2016] [Indexed: 12/28/2022] Open
Abstract
Binding of pattern recognition receptors (PRRs) by pathogen-associated molecular patterns (PAMPs) activates innate immune responses and contributes to development of adaptive immunity. Simultaneous stimulation of different types of PRRs can have synergistic immunostimulatory effects resulting in enhanced production of molecules that mediate innate immunity such as inflammatory cytokines, antimicrobial peptides, etc. Here, we evaluated the impact of combined stimulation of PRRs from different families on adaptive immunity by generating alum-based vaccine formulations with ovalbumin as a model antigen and the Toll-like receptor 4 (TLR4) agonist MPLA and the Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) agonist MDP adsorbed individually or together on the alum-ovalbumin particles. Multiple in vitro and in vivo readouts of immune system activation all showed that while individual PRR agonists increased the immunogenicity of vaccines compared to alum alone, the combination of both PRR agonists was significantly more effective. Combined stimulation of TLR4 and NOD2 results in a stronger and broader transcriptional response in THP-1 cells compared to individual PRR stimulation. Immunostimulatory composition containing both PRR agonists (MPLA and MDP) in the context of the alum-based ovalbumin vaccine also enhanced uptake of vaccine particles by bone marrow derived dendritic cells (BMDCs) and promoted maturation (up-regulation of expression of CD80, CD86, MHCII) and activation (production of cytokines) of BMDCs. Finally, immunization of mice with vaccine particles containing both PRR agonists resulted in enhanced cellular immunity as indicated by increased proliferation and activation (IFN-γ production) of splenic CD4+ and CD8+ T cells following in vitro restimulation with ovalbumin and enhanced humoral immunity as indicated by higher titers of ovalbumin-specific IgG antibodies. These results indicate that combined stimulation of TLR4 and NOD2 receptors dramatically enhances activation of both the humoral and cellular branches of adaptive immunity and suggests that inclusion of agonists of these receptors in standard alum-based adjuvants could be used to improve the effectiveness of vaccination.
Collapse
Affiliation(s)
- Amir I. Tukhvatulin
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | - Alina S. Dzharullaeva
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | - Natalia M. Tukhvatulina
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | - Dmitry V. Shcheblyakov
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | - Maxim M. Shmarov
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | - Inna V. Dolzhikova
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | | | - Boris S. Naroditsky
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| | - Andrei V. Gudkov
- Cleveland BioLabs, Inc., Buffalo, New York, United States of America
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton str., 14263 Buffalo, New York, United States of America
| | - Denis Y. Logunov
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
- * E-mail:
| | - Alexander L. Gintsburg
- N. F. Gamaleya Research Institute for Epidemiology and Microbiology, Gamaleya str.18, 123098 Moscow, Russia
| |
Collapse
|
29
|
Ghosh S, Dass JFP. Study of pathway cross-talk interactions with NF-κB leading to its activation via ubiquitination or phosphorylation: A brief review. Gene 2016; 584:97-109. [PMID: 26968890 DOI: 10.1016/j.gene.2016.03.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/05/2016] [Accepted: 03/02/2016] [Indexed: 12/23/2022]
Abstract
NFκB has been known to be a necessary transcription factor for the functioning of nearly all cells in a living organism. For its proper functioning, it talks to several other molecular cofactors and interacts with their functionalities resulting in a convoluted cross talking mesh of signalling networks. To completely understand the working of nuclear factor-kappa B protein, one needs to understand the interactions that occur during its lifecycle, with cofactors from various biological processes. This study attempts to elaborate and bridge the gaps on the cross-talk interactions that NFkB is a part of, during its activation pathway. For this Cytoscape and its various plugins (Cytocopter, Allegro, AgilentLitSearch and Styles) are employed. Other related pathways were also collated and analysed for cross-talk between NfκB and interacting molecules. NFκB was found to mainly interact with E3 ubiquitin ligase, NIK, RIP, TCR, IRAK-1, TLR, TRAF-6, NLR and IL-1, details of which are discussed as a part of this study.
Collapse
Affiliation(s)
- Sayantan Ghosh
- Bioinformatics Division, School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - J Febin Prabhu Dass
- Bioinformatics Division, School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
30
|
Wei LJ, Tan X, Fan GJ, Jiang YN, Shah QA. Role of the NOD1/NF-κB pathway on bovine neutrophil responses to crude lipopolysaccharide. Vet J 2016; 214:24-31. [PMID: 27387722 DOI: 10.1016/j.tvjl.2016.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/02/2016] [Accepted: 02/13/2016] [Indexed: 11/27/2022]
Abstract
Cytosolic nucleotide oligomerisation domain (NOD)-like receptors play an important role in host defence against infection. Reduced NOD1 expression has been observed in dysfunctional neutrophils derived from periparturient cattle known to be most susceptible to coliform mastitis. However, whether impairment of NOD1 suppresses the immune responses of bovine neutrophils during bacterial infections remains unknown. Crude (phenol extracted) lipopolysaccharide (cLPS), which often contains other immunostimulatory molecules, including NOD1 agonist, is known to induce almost the whole bacterial response. This study was conducted to explore the role of NOD1/nuclear factor (NF)-κB pathway in the cytokine and functional responses of bovine neutrophils challenged with Escherichia coli-derived cLPS. Freshly isolated blood neutrophils from healthy heifers were pre-incubated for 2 h with ML130, a selective inhibitor of NOD1/NF-κB pathway. Cells were then exposed to cLPS for additional 4 h. Inhibition of the NOD1/NF-κB pathway resulted in a decrease in cLPS-induced phosphorylation of the inhibitor of NF-κBα (IκBα) in neutrophils. Impairment of the NOD1/NF-κB pathway tended to down-regulate mRNA levels of pro-inflammatory cytokines interleukin (IL)-1β and tumour necrosis factor (TNF)-α, chemokines IL-8 and C-X-C motif ligand 2 (CXCL2), and adhesion molecules CD11b and CD62L, in cLPS-challenged cells. Functional analyses showed that blocking the NOD1/NF-κB pathway inhibited neutrophil migration and phagocytic killing capacity, and promoted neutrophil death upon cLPS stimulation. The data presented here demonstrate that activation of NOD1/NF-κB pathway contributes to the functional responses of neutrophils to cLPS.
Collapse
Affiliation(s)
- Liang-Jun Wei
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, 866 Yuhangtang Rd., Hangzhou 310058, China
| | - Xun Tan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, 866 Yuhangtang Rd., Hangzhou 310058, China.
| | - Guo-Juan Fan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, 866 Yuhangtang Rd., Hangzhou 310058, China
| | - Ya-Nan Jiang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, 866 Yuhangtang Rd., Hangzhou 310058, China
| | - Qurban A Shah
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, 866 Yuhangtang Rd., Hangzhou 310058, China
| |
Collapse
|
31
|
Liu Q, Ding JL. The molecular mechanisms of TLR-signaling cooperation in cytokine regulation. Immunol Cell Biol 2016; 94:538-42. [PMID: 26860369 DOI: 10.1038/icb.2016.18] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/02/2016] [Accepted: 02/07/2016] [Indexed: 12/19/2022]
Abstract
Innate immune cells recognize pathogens through pattern recognition receptors (PRRs), and activation of PRRs induces downstream signaling pathways to mount appropriate immune responses. Pathogens usually carry multiple ligands, which can simultaneously activate multiple PRRs. The cooperation of multiple PRRs and consequential crosstalk between their downstream pathways could enhance cytokine expression, which is required for effective immune responses. On the other hand, immune over-activation could also harm the host if immune homeostasis is not restored. Therefore, it is important to understand the mechanisms of PRR cooperation during an infection. As the best characterized PRRs, Toll-like receptors (TLRs) have an important role in pathogen recognition, and crosstalk among TLRs is common. In this review, we provide an update on the recent findings on the mechanisms of TLR cooperation. We summarize the known mechanisms and provide a future perspective on TLR crosstalk study, with a caution against the use of multiple TLR ligands as adjuvants in therapeutic strategies.
Collapse
Affiliation(s)
- Qian Liu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Jeak Ling Ding
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Thaiss CA, Levy M, Itav S, Elinav E. Integration of Innate Immune Signaling. Trends Immunol 2016; 37:84-101. [PMID: 26755064 DOI: 10.1016/j.it.2015.12.003] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/07/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022]
Abstract
The last decades of research in innate immunology have revealed a multitude of sensing receptors that evaluate the presence of microorganisms or cellular damage in tissues. In the context of a complex tissue, many such sensing events occur simultaneously. Thus, the downstream pathways need to be integrated to launch an appropriate cellular response, to tailor the magnitude of the reaction to the inciting event, and to terminate it in a manner that avoids immunopathology. Here, we provide a conceptual overview of the crosstalk between innate immune receptors in the initiation of a concerted immune reaction to microbial and endogenous triggers. We classify the known interactions into categories of communication and provide examples of their importance in pathogenic infection.
Collapse
Affiliation(s)
| | - Maayan Levy
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Shlomik Itav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
33
|
Wojdak-Maksymiec K, Mikołajczyk K, Prüffer K. Association of <i>TLR4</i> and <i>CARD15/NOD2</i> polymorphisms with SCC in Holstein–Friesian cattle. Arch Anim Breed 2015. [DOI: 10.5194/aab-58-293-2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Abstract. Mastitis is one of the most important dairy cattle diseases which results in economic losses in dairy production. Mastitis cases can be classified as subclinical or clinical. All forms of mastitis lead to changes in milk composition and induce an increase in somatic cell count (SCC). SCC is a very important and basic indicator of udder health. An increase in SCC is usually caused by the immune response to the invasion of pathogens contributing to mastitis. The aim of this study was to investigate associations between the polymorphisms of selected genes (TLR4 and CARD15/NOD2) whose products are involved in the identification of pathogen-associated molecular patterns (PAMPs) during the innate immune response to infection, and immunity to mastitis expressed as SCC. The genes under study were also examined for epistatic effects as well as effects of interactions with parity and stages of lactation. In all the studied classes, allele G of TLR4 had a favourable additive effect with negative values, contributing to a lower lnSCC. Allele A of CARD15/NOD2 had a desirable additive effect which varied with time and the changing internal environment during lactation. With regard to the dominance effect, allele A of CARD15/NOD2 was found to be significantly associated with a higher SCC in milk in the first lactation and in the third stage of each single lactation. Moreover, statistically significant epistatic effects were found, in particular additive–additive and dominance–additive interactions were favourably associated with SCC which was lower than expected in the case of no epistasis.
Collapse
|
34
|
Subramanian N, Torabi-Parizi P, Gottschalk RA, Germain RN, Dutta B. Network representations of immune system complexity. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:13-38. [PMID: 25625853 PMCID: PMC4339634 DOI: 10.1002/wsbm.1288] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 12/25/2022]
Abstract
The mammalian immune system is a dynamic multiscale system composed of a hierarchically organized set of molecular, cellular, and organismal networks that act in concert to promote effective host defense. These networks range from those involving gene regulatory and protein–protein interactions underlying intracellular signaling pathways and single‐cell responses to increasingly complex networks of in vivo cellular interaction, positioning, and migration that determine the overall immune response of an organism. Immunity is thus not the product of simple signaling events but rather nonlinear behaviors arising from dynamic, feedback‐regulated interactions among many components. One of the major goals of systems immunology is to quantitatively measure these complex multiscale spatial and temporal interactions, permitting development of computational models that can be used to predict responses to perturbation. Recent technological advances permit collection of comprehensive datasets at multiple molecular and cellular levels, while advances in network biology support representation of the relationships of components at each level as physical or functional interaction networks. The latter facilitate effective visualization of patterns and recognition of emergent properties arising from the many interactions of genes, molecules, and cells of the immune system. We illustrate the power of integrating ‘omics’ and network modeling approaches for unbiased reconstruction of signaling and transcriptional networks with a focus on applications involving the innate immune system. We further discuss future possibilities for reconstruction of increasingly complex cellular‐ and organism‐level networks and development of sophisticated computational tools for prediction of emergent immune behavior arising from the concerted action of these networks. WIREs Syst Biol Med 2015, 7:13–38. doi: 10.1002/wsbm.1288 This article is categorized under:
Analytical and Computational Methods > Computational Methods Laboratory Methods and Technologies > Macromolecular Interactions, Methods
Collapse
Affiliation(s)
- Naeha Subramanian
- Institute for Systems Biology, Seattle, WA, USA; Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
35
|
Collaborative action of Toll-like and NOD-like receptors as modulators of the inflammatory response to pathogenic bacteria. Mediators Inflamm 2014; 2014:432785. [PMID: 25525300 PMCID: PMC4267164 DOI: 10.1155/2014/432785] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/11/2014] [Accepted: 06/27/2014] [Indexed: 01/20/2023] Open
Abstract
Early sensing of pathogenic bacteria by the host immune system is important to develop effective mechanisms to kill the invader. Microbial recognition, activation of signaling pathways, and effector mechanisms are sequential events that must be highly controlled to successfully eliminate the pathogen. Host recognizes pathogens through pattern-recognition receptors (PRRs) that sense pathogen-associated molecular patterns (PAMPs). Some of these PRRs include Toll-like receptors (TLRs), nucleotide-binding oligomerization domain-like receptors (NLRs), retinoic acid-inducible gene-I- (RIG-I-) like receptors (RLRs), and C-type lectin receptors (CLRs). TLRs and NLRs are PRRs that play a key role in recognition of extracellular and intracellular bacteria and control the inflammatory response. The activation of TLRs and NLRs by their respective ligands activates downstream signaling pathways that converge on activation of transcription factors, such as nuclear factor-kappaB (NF-κB), activator protein-1 (AP-1) or interferon regulatory factors (IRFs), leading to expression of inflammatory cytokines and antimicrobial molecules. The goal of this review is to discuss how the TLRs and NRLs signaling pathways collaborate in a cooperative or synergistic manner to counteract the infectious agents. A deep knowledge of the biochemical events initiated by each of these receptors will undoubtedly have a high impact in the design of more effective strategies to control inflammation.
Collapse
|
36
|
Juárez E, Carranza C, Hernández-Sánchez F, Loyola E, Escobedo D, León-Contreras JC, Hernández-Pando R, Torres M, Sada E. Nucleotide-oligomerizing domain-1 (NOD1) receptor activation induces pro-inflammatory responses and autophagy in human alveolar macrophages. BMC Pulm Med 2014; 14:152. [PMID: 25253572 PMCID: PMC4190423 DOI: 10.1186/1471-2466-14-152] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/19/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Nucleotide-binding oligomerizing domain-1 (NOD1) is a cytoplasmic receptor involved in recognizing bacterial peptidoglycan fragments that localize to the cytosol. NOD1 activation triggers inflammation, antimicrobial mechanisms and autophagy in both epithelial cells and murine macrophages. NOD1 mediates intracellular pathogen clearance in the lungs of mice; however, little is known about NOD1's role in human alveolar macrophages (AMs) or its involvement in Mycobacterium tuberculosis (Mtb) infection. METHODS AMs, monocytes (MNs), and monocyte-derived macrophages (MDMs) from healthy subjects were assayed for NOD1 expression. Cells were stimulated with the NOD1 ligand Tri-DAP and cytokine production and autophagy were assessed. Cells were infected with Mtb and treated with Tri-DAP post-infection. CFUs counting determined growth control, and autophagy protein recruitment to pathogen localization sites was analyzed by immunoelectron microscopy. RESULTS NOD1 was expressed in AMs, MDMs and to a lesser extent MNs. Tri-DAP stimulation induced NOD1 up-regulation and a significant production of IL1β, IL6, IL8, and TNFα in AMs and MDMs; however, the level of NOD1-dependent response in MNs was limited. Autophagy activity determined by expression of proteins Atg9, LC3, IRGM and p62 degradation was induced in a NOD1-dependent manner in AMs and MDMs but not in MNs. Infected AMs could be activated by stimulation with Tri-DAP to control the intracellular growth of Mtb. In addition, recruitment of NOD1 and the autophagy proteins IRGM and LC3 to the Mtb localization site was observed in infected AMs after treatment with Tri-DAP. CONCLUSIONS NOD1 is involved in AM and MDM innate responses, which include proinflammatory cytokines and autophagy, with potential implications in the killing of Mtb in humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Eduardo Sada
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City, México.
| |
Collapse
|
37
|
Ghaderi H, Kiany F, Razmkhah M, Dadras S, Chenari N, Hosseini A, Younesi V, Ghaderi A. mRNA expression of pattern recognition receptors and their signaling mediators in healthy and diseased gingival tissues. J Indian Soc Periodontol 2014; 18:150-4. [PMID: 24872620 PMCID: PMC4033878 DOI: 10.4103/0972-124x.131309] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 10/07/2013] [Indexed: 01/19/2023] Open
Abstract
Background: Gingivitis and periodontitis are initiated by inflammation caused by microorganisms. Pathogen-associated molecular patterns (PAMPs) from these microorganisms are recognized through various toll-like receptors (TLRs) and NOD-like receptors (NLRs). In this study, we have chosen five TLRs and two NLRs as representatives taking part in the recognition and inflammation process, along with a few of their signaling mediators including CD14, MYD88, and TRIF to compare their mRNA expression levels between healthy and diseased gingival tissues. This will provide deeper insight into the mechanisms underlying gingivitis and periodontitis. Understanding the mechanisms involved in the onset and progression of the periodontal diseases could greatly help in establishing effective ways for prevention and treatment of these diseases besides decreasing the risk factor for relevant systemic disorders. Materials and Methods: Gingival tissue samples for mRNA extraction and cDNA synthesis were taken from patients with gingivitis and periodontitis and from healthy control subjects. Messenger RNA expression of all genes was assessed using real-time polymerase chain reaction (PCR). Results: Among the genes studied in different groups, only MYD88 mRNA expression was significantly higher in the periodontitis group compared to that of the controls. The expression level of this molecule was also significantly higher in patients with severe periodontitis compared to other patients and also compared to healthy individuals. In different tissues, positive significant correlations were observed between the mRNA expression levels of some genes. Conclusions: Elevated mRNA levels of MYD88 in periodontitis might have a key role in the pathogenesis of this disease. Therefore, MYD88 may be a useful target for the therapy of this disease.
Collapse
Affiliation(s)
- Hamid Ghaderi
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran ; Department of Periodontology, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farin Kiany
- Department of Periodontology, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Dadras
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Noushafarin Chenari
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Hosseini
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Younesi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran ; Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Chai L, Wu S, Liu G, Wang Z, Tian W, Ma Y. OCILRP2 signaling synergizes with LPS to induce the maturation and differentiation of murine dendritic cells. Biochem Biophys Res Commun 2014; 446:836-42. [PMID: 24631687 DOI: 10.1016/j.bbrc.2014.02.145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 02/25/2014] [Indexed: 11/29/2022]
Abstract
Osteoclast Inhibitory Lectin-related Protein 2 (OCILRP2) is a typical type II transmembrane protein and belongs to C-type lectin-related protein family. It is preferentially expressed in dendritic cells (DC), B lymphocytes, and activated T lymphocytes. Upon binding to its ligand, OCILRP2 can promote CD28-mediated co-stimulation and enhance T cell activation. However, the role of OCILRP2 in DC development and activation is unclear. In this report, we present evidence that recombinant protein OCILRP2-Fc inhibits the generation and LPS-induced maturation of murine bone marrow-derived dendritic cells (BMDCs) by downregulating the expression of CD11c, MHC-II, and co-stimulators CD80 and CD86. OCILRP2-Fc also reduces the capacity of BMDCs to take up antigens, activates T cells, and secret inflammatory cytokines such as IL-6, IL-12, and TNF-α. Additionally, we show that OCILRP2-Fc may cause the aforementioned effects through inhibiting NF-κB activation. Therefore, OCILRP2 is a new regulator of DC maturation and differentiation following TLR4 activation.
Collapse
Affiliation(s)
- Lihui Chai
- Department of Microbiology and Immunology, Zhengzhou University School of Medicine, 100 Kexue Road, Zhengzhou 450001, People's Republic of China; Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Suxia Wu
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Guangchao Liu
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Zhanzheng Wang
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Wenzhi Tian
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China.
| | - Yuanfang Ma
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China.
| |
Collapse
|
39
|
Hepcidin induction by pathogens and pathogen-derived molecules is strongly dependent on interleukin-6. Infect Immun 2013; 82:745-52. [PMID: 24478088 DOI: 10.1128/iai.00983-13] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepcidin, the iron-regulatory hormone, is increased during infection or inflammation, causing hypoferremia. This response is thought to be a host defense mechanism that restricts iron availability to invading pathogens. It is not known if hepcidin is differentially induced by bacterial versus viral infections, whether the stimulation of pattern recognition receptors directly regulates hepcidin transcription, or which of the proposed signaling pathways are essential for hepcidin increase during infection. We analyzed hepcidin induction and its dependence on interleukin-6 (IL-6) in response to common bacterial or viral infections in mice or in response to a panel of pathogen-derived molecules (PAMPs) in mice and human primary hepatocytes. In wild-type (WT) mice, hepcidin mRNA was induced several hundred-fold both by a bacterial (Streptococcus pneumoniae) and a viral infection (influenza virus PR8) within 2 to 5 days. Treatment of mice and human primary hepatocytes with most Toll-like receptor ligands increased hepcidin mRNA within 6 h. Hepcidin induction by microbial stimuli was IL-6 dependent. IL-6 knockout mice failed to increase hepcidin in response to S. pneumoniae or influenza infection and had greatly diminished hepcidin response to PAMPs. In vitro, hepcidin induction by PAMPs in primary human hepatocytes was abolished by the addition of neutralizing IL-6 antibodies. Our results support the key role of IL-6 in hepcidin regulation in response to a variety of infectious and inflammatory stimuli.
Collapse
|
40
|
Abstract
Kupffer cells are a critical component of the mononuclear phagocytic system and are central to both the hepatic and systemic response to pathogens. Kupffer cells are reemerging as critical mediators of both liver injury and repair. Kupffer cells exhibit a tremendous plasticity; depending on the local metabolic and immune environment, then can express a range of polarized phenotypes, from the proinflammatory M1 phenotype to the alternative/M2 phenotype. Multiple M2 phenotypes can be distinguished, each involved in the resolution of inflammation and wound healing. Here, we have provided an update on recent research that has contributed to the developing delineation of the contribution of Kupffer cells to different types of liver injury, with an emphasis on alcoholic and nonalcoholic liver diseases. These recent advances in our understanding of Kupffer cell function and regulation will likely provide new insights into the potential for therapeutic manipulation of Kupffer cells to promote the resolution of inflammation and enhance wound healing in liver disease.
Collapse
Affiliation(s)
- Laura J Dixon
- Liver Disease Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
41
|
Moreno L, Gatheral T. Therapeutic targeting of NOD1 receptors. Br J Pharmacol 2013; 170:475-85. [PMID: 23848281 PMCID: PMC3791987 DOI: 10.1111/bph.12300] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/30/2013] [Accepted: 07/04/2013] [Indexed: 12/11/2022] Open
Abstract
The nucleotide-binding oligomerization domain 1 (NOD1) protein is an intracellular receptor for breakdown products of peptidoglycan (PGN), an essential bacterial cell wall component. NOD1 responds to γ-D-glutamyl-meso-diaminopimelic acid, which is an epitope unique to PGN structures from all Gram-negative bacteria and certain Gram-positive bacteria. Upon ligand recognition, NOD1 undergoes conformational changes and self-oligomerization mediated by the nucleotide-binding NACHT domains, followed by the recruitment and activation of the serine threonine kinase receptor-interacting protein 2 leading to the activation of NF-κB and MAPK pathways and induction of inflammatory genes. Much of our knowledge is derived from seminal studies using mice deficient in NOD1 and confirming an essential role for NOD1 in the host immune response against gastrointestinal and respiratory pathogens. In addition, recent studies have revealed a role for intracellular NOD1 receptors in the regulation of vascular inflammation and metabolism. This review will discuss our current understanding of intracellular NOD1 receptors in host immunity and chronic inflammatory disorders with a focus on cardiovascular diseases. Although therapeutic advances may have to wait until the complex interplay with pathogens, danger signals, other pattern recognition receptors and overlapping metabolic pathways is further unravelled, the steadily growing body of knowledge suggest that NOD1 antagonism might represent attractive candidate to reduce excessive inflammation associated to intestinal, cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- L Moreno
- Ciber de Enfermedades Respiratorias (CIBERES), Bunyola, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | | |
Collapse
|
42
|
Xue LY, Ouyang Q, Zhou XG, Huang ZH, Chen W, Chen M, Yu LM. Bacterial immune interaction in experimental colitis. J Dig Dis 2013; 14:526-35. [PMID: 23734583 DOI: 10.1111/1751-2980.12079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This study aimed to analyze the effects of 5-aminosalicylic acid (5-ASA) on intestinal microbiota and immune regulation in inflammatory bowel disease (IBD) and to investigate the correlation between intestinal microbiota and immune factors. METHODS Colitis in mice was induced by oxazolone. The community composition of luminal and mucosal microbiota was analyzed by a terminal restriction fragment length polymorphism. The expression of occludin, toll-like receptor (TLR)-2, TLR-4 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 proteins were measured by immunohistochemistry and Western blot. Linear correlation between intestinal microbial community and the severity of the colitis or intestinal microbial community and expressions of immune factors were determined. RESULTS Protective bacteria decreased while aggressive bacteria increased in the colitis group. The richness and diversity of both luminal and mucosal microbiota decreased in the colitis group the decrease was enhanced in the 5-ASA-treated group. The diversity of mucosal microbiota significantly correlated with the extent of the colitis. Expressions of occludin, TLR-2, TLR-4, tumor necrosis factor-α and NF-κB p65 were significantly correlated with the diversity of mucosal microbiota. CONCLUSIONS Mucosal microbiota are important in the pathogenesis of IBD. 5-ASA increases protective bacteria but decreases aggressive bacteria, thus inducing the new intestinal microbial homeostasis.
Collapse
Affiliation(s)
- Lin Yun Xue
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Jakopin Ž, Gobec M, Kodela J, Hazdovac T, Mlinarič-Raščan I, Sollner Dolenc M. Synthesis of conformationally constrained γ-D-glutamyl-meso-diaminopimelic acid derivatives as ligands of nucleotide-binding oligomerization domain protein 1 (Nod1). Eur J Med Chem 2013; 69:232-43. [PMID: 24044936 DOI: 10.1016/j.ejmech.2013.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/07/2013] [Accepted: 08/12/2013] [Indexed: 11/26/2022]
Abstract
Nod1, an important member of the pattern recognition receptor family, remains a virtually unexploited target. Harnessing its innate immune stimulatory properties still remains an unfulfilled goal of medicinal chemistry. Nucleotide-binding oligomerization domain protein 1 (Nod1) agonists have been shown to boost the inflammatory responses against pathogenic microbes and could thus constitute a new class of broad spectrum antimicrobial agents. To gain additional insight into the structure/activity relationships of Nod1 agonistic compounds, a series of novel, conformationally constrained γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) analogs have been designed and synthesized. Ramos-Blue cells expressing Nod1 were used to screen and validate our compounds for their Nod1-agonist activity. Their immunomodulatory properties were subsequently determined in vitro, by evaluating their capacity to induce pro-inflammatory cytokine and chemokine production from human peripheral blood mononuclear cells (PBMC), by themselves and in synergy with lipopolysaccharide (LPS), a Toll-like receptor 4 (TLR4) ligand. The synthesized iE-DAP analogs were shown to possess immuno-enhancing properties as a result of their potent and specific Nod1-agonistic effect. The activity of the compound exhibiting the greatest capacity to induce pro-inflammatory cytokine release from PBMC surpassed that of lauroyl-γ-D-glutamyl-meso-diaminopimelic acid (C12-iE-DAP).
Collapse
Affiliation(s)
- Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | | | | | | | | | | |
Collapse
|
44
|
Combined stimulation of Toll-like receptor 5 and NOD1 strongly potentiates activity of NF-κB, resulting in enhanced innate immune reactions and resistance to Salmonella enterica serovar Typhimurium infection. Infect Immun 2013; 81:3855-64. [PMID: 23897616 DOI: 10.1128/iai.00525-13] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pathogen recognition receptors (PRRs) are essential components of host innate immune systems that detect specific conserved pathogen-associated molecular patterns (PAMPs) presented by microorganisms. Members of two families of PRRs, transmembrane Toll-like receptors (TLRs 1, 2, 4, 5, and 6) and cytosolic NOD receptors (NOD1 and NOD2), are stimulated upon recognition of various bacterial PAMPs. Such stimulation leads to induction of a number of immune defense reactions, mainly triggered via activation of the transcription factor NF-κB. While coordination of responses initiated via different PRRs sensing multiple PAMPS present during an infection makes clear biological sense for the host, such interactions have not been fully characterized. Here, we demonstrate that combined stimulation of NOD1 and TLR5 (as well as other NOD and TLR family members) strongly potentiates activity of NF-κB and induces enhanced levels of innate immune reactions (e.g., cytokine production) both in vitro and in vivo. Moreover, we show that an increased level of NF-κB activity plays a critical role in formation of downstream responses. In live mice, synergy between these receptors resulting in potentiation of NF-κB activity was organ specific, being most prominent in the gastrointestinal tract. Coordinated activity of NOD1 and TLR5 significantly increased protection of mice against enteroinvasive Salmonella infection. Obtained results suggest that cooperation of NOD and TLR receptors is important for effective responses to microbial infection in vivo.
Collapse
|
45
|
Shi G, Vistica BP, Nugent LF, Tan C, Wawrousek EF, Klinman DM, Gery I. Differential involvement of Th1 and Th17 in pathogenic autoimmune processes triggered by different TLR ligands. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:415-23. [PMID: 23720812 PMCID: PMC3749791 DOI: 10.4049/jimmunol.1201732] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interaction between TLRs and their cognate ligands triggers both the innate and adaptive immune systems, and thus can play a pivotal role in the defense against pathogen invasion. This work investigates the differentiation of naive CD4 cells into Th1 or Th17 phenotypes in mice treated with different TLR ligands. We use a model system in which naive transgenic cells specific to hen egg lysozyme are adoptively transferred into recipients that express hen egg lysozyme in the lens of the eye. The transferred naive T cells induce ocular inflammation only in recipients treated with TLR ligands. Treatment with LPS preferentially stimulated IL-17 production, whereas CpG oligodeoxynucleotide and polyinosinic:polycytidylic acid primarily stimulated Th1 cells. Peptidoglycan stimulated the two Th subpopulations equally. The preferential induction of Th1 or Th17 by the four ligands was detected in the spleen (where a major portion of the adoptively transferred cells homed) and in the eyes, where activated Th cells initiate inflammation. Analysis of the cytokines present in recipient mice suggests that Th1 induction is elicited by IL-12 and/or IFN-α, whereas Th17 generation is preferentially mediated by IL-6. Importantly, we show in this article that treatment with LPS selectively promoted in the recipient mice the generation of IL-6-producing activated B cells. An inverse correlation was found between the level of regulatory T cells and severity of inflammation induced by the donor cells. Taken together, our data show that specific TLR ligands differentially activate the immune system as evidenced by the generation of distinct Th phenotypes from naive CD4 cells.
Collapse
Affiliation(s)
- Guangpu Shi
- Laboratory of Immunology, NIH, Bethesda, MD 20892
| | | | | | - Cuiyan Tan
- Laboratory of Immunology, NIH, Bethesda, MD 20892
| | - Eric F. Wawrousek
- Laboratory of Molecular and Developmental Biology of the National Eye Institute, NIH, Bethesda, MD 20892
| | - Dennis M. Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21702
| | - Igal Gery
- Laboratory of Immunology, NIH, Bethesda, MD 20892
| |
Collapse
|
46
|
Myles A, Rahman MT, Aggarwal A. Membrane-bound toll-like receptors are overexpressed in peripheral blood and synovial fluid mononuclear cells of enthesitis-related arthritis category of juvenile idiopathic arthritis (JIA–ERA) patients and lead to secretion of inflammatory mediators. J Clin Immunol 2012; 32:488-96. [PMID: 22302567 DOI: 10.1007/s10875-011-9640-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 12/20/2011] [Indexed: 01/04/2023]
Abstract
We examined expression and function of TLRs in enthesitis-related arthritis (ERA) patients. RNA levels of TLR1, TLR3, and TLRs 5–8 were measured in 24 ERA peripheral blood mononuclear cells (PBMC), 18 synovial fluid mononuclear cells (SFMC), and IRAK1, IRAK4, TRIF, TRAF3, and TRAF6 in 18 PBMC and 10 SFMC. IL-6 and IL-8 were measured in supernatants from ERA PBMC (n=7), SFMC (n=3), and healthy PBMC (n=5) cultured with ligands for TLR1/2 (Pam 3-cys), TLR3 (polyI:C), TLR5 (flagellin), and TLR2/6 (zymosan). TLRs 1, 3, 5, and 6 were measured in whole blood (n=20 ERA, seven healthy) and SFMC (n=2) by flow cytometry. ERA PBMC compared to healthy PBMC and SFMC compared to ERA PBMC had higher RNA expression of TLR1, TLR3, TLR5, TLR6, IRAK1, IRAK4, TRIF, TRAF3, and TRAF6. TLR7 and TLR8 RNA expression was similar in all study groups. IL-6 and IL-8 levels were higher in stimulated ERA SFMC compared to ERA PBMC and in ERA PBMC compared to control PBMC. TLRs 1, 3, and 6 were also overexpressed at the protein level.
Collapse
Affiliation(s)
- Arpita Myles
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | | | | |
Collapse
|
47
|
Ahn MY, Yoon HE, Park JH, Lee J, Min SK, Ahn SG, Yoon JH. Characterization of NODs and TLRs in innate immune response of human cementoblast cells. Oral Dis 2012; 19:374-80. [PMID: 22957853 DOI: 10.1111/odi.12012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/18/2012] [Accepted: 08/02/2012] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Microbial Pattern-recognition receptors (PRRs), such as Toll-like receptors (TLRs) and the nucleotide-binding oligomerization domains (NODs), are essential for mammalian innate immune response. In this study, we examined the characterization of NODs and TLRs on innate immune responses in human cementoblast (HCEM) cells. MATERIALS AND METHODS The gene expression of NODs and TLRs was examined by RT-PCR. Interleukin-6 (IL-6) and Interleukin-8 (IL-8) productions in culture supernatants were measured by ELISA. Western blot analysis was performed to determine the degradation of IκB-α and Mitogen activated protein kinase (MAPK) activation in response to their agonist. RESULTS The levels of NODs and TLRs were apparently expressed in HCEM cells. Although a few gene levels were weak in intact cells, the stimulation by their agonists increased the gene expression of TLRs. NODs and TLRs led to the production of IL-6 or IL-8 and the degradation of IκB-α and MAPK activation in HCEM cells. Combination treatment of NOD1 or NOD2 agonists with TLRs agonists did not influence the production of IL-6 and IL-8 in HCEM cells. CONCLUSIONS Our results indicate that NODs and TLRs are functionally expressed in HCEM cells and can trigger innate immune responses. However, NOD1 and NOD2 may not be cooperated with TLRs to elicit an immune response in HCEM cells.
Collapse
Affiliation(s)
- M Y Ahn
- Department of Oral & Maxillofacial Pathology, College of Dentistry, Wonkwang Bone Regeneration Institute, Daejeon Dental Hospital, Wonkwang University, Daejeon, Korea
| | | | | | | | | | | | | |
Collapse
|
48
|
Kang MJ, Heo SK, Song EJ, Kim DJ, Han SY, Han JH, Kim BY, Park JH. Activation of Nod1 and Nod2 induces innate immune responses of prostate epithelial cells. Prostate 2012; 72:1351-8. [PMID: 22228081 DOI: 10.1002/pros.22483] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/12/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND Nod1 and Nod2 are cytosolic receptors which are responsible for sensing bacterial peptidoglycan derivatives. In this study, we determined whether Nod1 and Nod2 are involved in the innate immune responses of prostate epithelial cells. METHODS The expression of Nod1 and Nod2 was examined by RT-PCR and immunohistochemistry. ELISA was performed to determine the production of cytokines/chemokines. Activation of NF-κB and MAPK was examined using western blot analysis. RESULTS The Nod1 gene was distinctly expressed in all tested cells including DU145, PC3, and TRAMP-C2 cells, whereas Nod2 expression was weak. Both Nod1 and Nod2 proteins were expressed in normal mouse prostate epithelia with difference of expression levels. Tri-DAP (Nod1 agonist), but not MDP (Nod2), increased the production of IL-8 (or KC) and IL-6 in prostate epithelial cells. Tri-DAP and MDP could upregulate the gene expression of COX-2 and activate NF-κB and MAPK. In addition, Tri-DAP and MDP synergized with TLR agonists to induce the production of IL-8/KC or IL-6 in PC3 and TRAMP-C2 cells. We finally showed that Nod1 and Nod2 were also expressed in a wide range of prostate lesions including prostate intraepithelial neoplasm (PIN), phyllodes-like tumor, and adenocarcinoma in TRAMP (transgenic adenocarcinoma of the mouse prostate) mice, even though the expression level of Nod1 and Nod2 was different. CONCLUSION These results indicate that Nod1 and Nod2 may play important roles in the innate immune response of prostate epithelial cells and the development and progression of prostate cancer.
Collapse
Affiliation(s)
- Min-Jung Kang
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 302-711, Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Gatheral T, Reed DM, Moreno L, Gough PJ, Votta BJ, Sehon CA, Rickard DJ, Bertin J, Lim E, Nicholson AG, Mitchell JA. A key role for the endothelium in NOD1 mediated vascular inflammation: comparison to TLR4 responses. PLoS One 2012; 7:e42386. [PMID: 22870324 PMCID: PMC3411636 DOI: 10.1371/journal.pone.0042386] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/04/2012] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanisms by which pathogens induce vascular inflammation and dysfunction may reveal novel therapeutic targets in sepsis and related conditions. The intracellular receptor NOD1 recognises peptidoglycan which features in the cell wall of gram negative and some gram positive bacteria. NOD1 engagement generates an inflammatory response via activation of NFκB and MAPK pathways. We have previously shown that stimulation of NOD1 directly activates blood vessels and causes experimental shock in vivo. In this study we have used an ex vivo vessel-organ culture model to characterise the relative contribution of the endothelium in the response of blood vessels to NOD1 agonists. In addition we present the novel finding that NOD1 directly activates human blood vessels. Using human cultured cells we confirm that endothelial cells respond more avidly to NOD1 agonists than vascular smooth muscle cells. Accordingly we have sought to pharmacologically differentiate NOD1 and TLR4 mediated signalling pathways in human endothelial cells, focussing on TAK1, NFκB and p38 MAPK. In addition we profile novel inhibitors of RIP2 and NOD1 itself, which specifically inhibit NOD1 ligand induced inflammatory signalling in the vasculature. This paper is the first to demonstrate activation of whole human artery by NOD1 stimulation and the relative importance of the endothelium in the sensing of NOD1 ligands by vessels. This data supports the potential utility of NOD1 and RIP2 as therapeutic targets in human disease where vascular inflammation is a clinical feature, such as in sepsis and septic shock.
Collapse
Affiliation(s)
- Timothy Gatheral
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Daniel M. Reed
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Laura Moreno
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, United Kingdom
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Peter J. Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Philadelphia, Pennsylvania, United States of America
| | - Bart J. Votta
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Philadelphia, Pennsylvania, United States of America
| | - Clark A. Sehon
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Philadelphia, Pennsylvania, United States of America
| | - David J. Rickard
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Philadelphia, Pennsylvania, United States of America
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Philadelphia, Pennsylvania, United States of America
| | - Eric Lim
- Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Andrew G. Nicholson
- Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Jane A. Mitchell
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, United Kingdom
- Institute of Cardiovascular Medicine and Science (ICMS), London, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Jeong YJ, Kim CH, Song EJ, Kang MJ, Kim JC, Oh SM, Lee KB, Park JH. Nucleotide-binding oligomerization domain 2 (Nod2) is dispensable for the innate immune responses of macrophages against Yersinia enterocolitica. J Microbiol 2012; 50:489-95. [PMID: 22752913 DOI: 10.1007/s12275-012-1534-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/13/2012] [Indexed: 01/10/2023]
Abstract
Nucleotide-binding oligomerization domain 2 (Nod2) is a cytosolic sensor for muramyl dipeptide, a component of bacterial peptidoglycan. In this study, we have examined whether Nod2 mediates the immune response of macrophages against Yersinia enterocolitica. Bone-marrow-derived macrophages (BMDMs) were isolated from WT and Nod2-deficient mice and were infected with various strains of Y. enterocolitica. ELISA showed that the production of IL-6 and TNF-α in BMDMs infected with Y. enterocolitica was not affected by the Nod2 deficiency. iNOS mRNA expression was induced in both WT and Nod2-deficienct BMDMs in response to Y. enterocolitica, beginning 2 h after infection. Nitric oxide (NO) production by Y. enterocolitica did not differ between WT and Nod2-deficient BMDMs. Western blot analysis revealed that Y. enterocolitica induces activation of NF-κB, p38, and ERK MAPK through a Nod2-independent pathway. Neither LDH release by Y. enterocolitica nor the phagocytic activity of the macrophages was altered by Nod2 deficiency. An in vivo experiment showed that bacterial clearance ability and production of IL-6 and KC in serum were comparable in WT and Nod2-deficient mice infected with Y. enterocolitica. These findings suggest that Nod2 may not be critical for initiating the innate immune response of macrophages against Yersinia infection.
Collapse
Affiliation(s)
- Yu-Jin Jeong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 302-711, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|