1
|
Allard RL, Mayfield J, Barchiesi R, Salem NA, Mayfield RD. Toll-like receptor 7: A novel neuroimmune target to reduce excessive alcohol consumption. Neurobiol Stress 2024; 31:100639. [PMID: 38765062 PMCID: PMC11101708 DOI: 10.1016/j.ynstr.2024.100639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Toll-like receptors (TLRs) are a family of innate immune receptors that recognize molecular patterns in foreign pathogens and intrinsic danger/damage signals from cells. TLR7 is a nucleic acid sensing endosomal TLR that is activated by single-stranded RNAs from microbes or by small noncoding RNAs that act as endogenous ligands. TLR7 signals through the MyD88 adaptor protein and activates the transcription factor interferon regulatory factor 7 (IRF7). TLR7 is found throughout the brain and is highly expressed in microglia, the main immune cells of the brain that have also been implicated in alcohol drinking in mice. Upregulation of TLR7 mRNA and protein has been identified in postmortem hippocampus and cortex from AUD subjects that correlated positively with lifetime consumption of alcohol. Similarly, Tlr7 and downstream signaling genes were upregulated in rat hippocampal and cortical slice cultures after chronic alcohol exposure and in these regions after chronic binge-like alcohol treatment in mice. In addition, repeated administration of the synthetic TLR7 agonists imiquimod (R837) or resiquimod (R848) increased voluntary alcohol drinking in different rodent models and produced sustained upregulation of IRF7 in the brain. These findings suggest that chronic TLR7 activation may drive excessive alcohol drinking. In the brain, this could occur through increased levels of endogenous TLR7 activators, like microRNAs and Y RNAs. This review explores chronic TLR7 activation as a pathway of dysregulated neuroimmune signaling in AUD and the endogenous small RNA ligands in the brain that could perpetuate innate immune responses and escalate alcohol drinking.
Collapse
Affiliation(s)
- Ruth L. Allard
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Riccardo Barchiesi
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nihal A. Salem
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
2
|
Soleiman-Meigooni S, Yarahmadi A, Kheirkhah AH, Afkhami H. Recent advances in different interactions between toll-like receptors and hepatitis B infection: a review. Front Immunol 2024; 15:1363996. [PMID: 38545106 PMCID: PMC10965641 DOI: 10.3389/fimmu.2024.1363996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/26/2024] [Indexed: 04/17/2024] Open
Abstract
Hepatitis B virus (HBV) B infections remain a primary global health concern. The immunopathology of the infection, specifically the interactions between HBV and the host immune system, remains somewhat unknown. It has been discovered that innate immune reactions are vital in eliminating HBV. Toll-like receptors (TLRs) are an essential category of proteins that detect pathogen-associated molecular patterns (PAMPs). They begin pathways of intracellular signals to stimulate pro-inflammatory and anti-inflammatory cytokines, thus forming adaptive immune reactions. HBV TLRs include TLR2, TLR3, TLR4, TLR7 and TLR9. Each TLR has its particular molecule to recognize; various TLRs impact HBV and play distinct roles in the pathogenesis of the disease. TLR gene polymorphisms may have an advantageous or disadvantageous efficacy on HBV infection, and some single nucleotide polymorphisms (SNPs) can influence the progression or prognosis of infection. Additionally, it has been discovered that similar SNPs in TLR genes might have varied effects on distinct populations due to stress, diet, and external physical variables. In addition, activation of TLR-interceded signaling pathways could suppress HBV replication and increase HBV-particular T-cell and B-cell reactions. By identifying these associated polymorphisms, we can efficiently advance the immune efficacy of vaccines. Additionally, this will enhance our capability to forecast the danger of HBV infection or the threat of dependent liver disease development via several TLR SNPs, thus playing a role in the inhibition, monitoring, and even treatment guidance for HBV infection. This review will show TLR polymorphisms, their influence on TLR signaling, and their associations with HBV diseases.
Collapse
Affiliation(s)
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Amir-Hossein Kheirkhah
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
3
|
Huang Y, Nahar S, Alam MM, Hu S, McVicar DW, Yang D. Reactive Oxygen Species-Sensitive Biodegradable Mesoporous Silica Nanoparticles Harboring TheraVac Elicit Tumor-Specific Immunity for Colon Tumor Treatment. ACS NANO 2023; 17:19740-19752. [PMID: 37831945 DOI: 10.1021/acsnano.3c03195] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Immunotherapy has revolutionized the field of cancer treatment through invigorating robust antitumor immune response. Here, we report the development of a therapeutic vaccine [consisting of high mobility group nucleosome-binding protein 1 (HMGN1), resiquimod/R848, and anti-PD-L1 (αPD-L1)]-loaded reactive oxygen species (ROS)-responsive mesoporous silica nanoparticle (MSN@TheraVac) for curative therapy of colon cancer. In MSN@TheraVac, αPD-L1 conjugated onto the surface of MSNs via a diselenide bond, which can be rapidly released under the oxidative condition of the tumor microenvironment to avert immunosuppression and effector T cell exhaustion while coloaded HMGN1 and R848 would cooperatively trigger robust tumor-infiltrating dendritic cell (TiDC) maturation and elicitation of antitumor immune responses. Indeed, MSN@TheraVac induced the maturation and activation of dendritic cells (DCs) by promoting the surface expression of CD80, CD86, and CD103 as well as the production of pro-inflammatory cytokines, including TNFα, IL-12, and IL-1β. Importantly, treatment with intravenous MSN@TheraVac led to a complete cure of 100% of BALB/c mice bearing large colon tumors and induced the generation of tumor-specific protective memory without apparent toxicity. Thus, MSN@TheraVac provides a timely release of TheraVac for the curative treatment of colon tumors and holds potential for translation into a clinical therapy for patients with immunologically "cold" colorectal cancers. This ROS-responsive MSN platform may also be tailored for the selective delivery of other cancer vaccines for effective immunotherapy.
Collapse
Affiliation(s)
- Yue Huang
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick 21702, Maryland, United States
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Saifun Nahar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick 21702, Maryland, United States
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda 20892, Maryland, United States
| | - Md Masud Alam
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick 21702, Maryland, United States
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda 20892, Maryland, United States
| | - Shuo Hu
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick 21702, Maryland, United States
| | - De Yang
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick 21702, Maryland, United States
| |
Collapse
|
4
|
Lovelock DF, Liu W, Langston SE, Liu J, Van Voorhies K, Giffin KA, Vetreno RP, Crews FT, Besheer J. The Toll-like receptor 7 agonist imiquimod increases ethanol self-administration and induces expression of Toll-like receptor related genes. Addict Biol 2022; 27:e13176. [PMID: 35470561 PMCID: PMC9286850 DOI: 10.1111/adb.13176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/24/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022]
Abstract
There is growing evidence that immune signalling may be involved in both the causes and consequences of alcohol abuse. Toll-like receptor (TLR) expression is increased by alcohol consumption and is implicated in AUD, and specifically TLR7 may play an important role in ethanol consumption. We administered the TLR7-specific agonist imiquimod in male and female Long-Evans rats to determine (1) gene expression changes in brain regions involved in alcohol reinforcement, the nucleus accumbens core and anterior insular cortex, in rats with and without an alcohol history, and (2) whether TLR7 activation could modulate operant alcohol self-administration. Interferon regulatory factor 7 (IRF7) was dramatically increased in both sexes at both 2- and 24-h post-injection regardless of alcohol history and TLR3 and 7 gene expression was increased as well. The proinflammatory cytokine TNFα was increased 24-h post-injection in rats with an alcohol self-administration history, but this effect did not persist after four injections, suggesting molecular tolerance. Ethanol consumption was increased 24 h after imiquimod injections but did not occur until the third injection, suggesting adaptation to repeated TLR7 activation is necessary for increased drinking to occur. Notably, imiquimod reliably induced weight loss, indicating that sickness behaviour persisted across repeated injections. These findings show that TLR7 activation can modulate alcohol drinking in an operant self-administration paradigm and suggest that TLR7 and IRF7 signalling pathways may be a viable druggable target for treatment of AUD.
Collapse
Affiliation(s)
- Dennis F. Lovelock
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Wen Liu
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Sarah E. Langston
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Jiaqi Liu
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kalynn Van Voorhies
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kaitlin A. Giffin
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
5
|
Systemic Administration of the TLR7/8 Agonist Resiquimod (R848) to Mice Is Associated with Transient, In Vivo-Detectable Brain Swelling. BIOLOGY 2022; 11:biology11020274. [PMID: 35205140 PMCID: PMC8869423 DOI: 10.3390/biology11020274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022]
Abstract
Peripheral administration of the E. coli endotoxin lipopolysaccharide (LPS) to rats promotes secretion of pro-inflammatory cytokines and in previous studies was associated with transient enlargement of cortical volumes. Here, resiquimod (R848) was administered to mice to stimulate peripheral immune activation, and the effects on brain volumes and neurometabolites determined. After baseline scans, 24 male, wild-type C57BL mice were triaged into three groups including R848 at low (50 μg) and high (100 μg) doses and saline controls. Animals were scanned again at 3 h and 24 h following treatment. Sickness indices of elevated temperature and body weight loss were observed in all R848 animals. Animals that received 50 μg R848 exhibited decreases in hippocampal N-acetylaspartate and phosphocreatine at the 3 h time point that returned to baseline levels at 24 h. Animals that received the 100 μg R848 dose demonstrated transient, localized, volume expansion (~5%) detectable at 3 h in motor, somatosensory, and olfactory cortices; and pons. A metabolic response evident at the lower dose and a volumetric change at the higher dose suggests a temporal evolution of the effect wherein the neurochemical change is demonstrable earlier than neurostructural change. Transient volume expansion in response to peripheral immune stimulation corresponds with previous results and is consistent with brain swelling that may reflect CNS edema.
Collapse
|
6
|
Järver P, Dondalska A, Poux C, Sandberg A, Bergenstråhle J, Sköld AE, Dereuddre-Bosquet N, Martinon F, Pålsson S, Zaghloul E, Brodin D, Sander B, Lennox KA, Behlke MA, El-Andaloussi S, Lehtiö J, Lundeberg J, LeGrand R, Spetz AL. Single-Stranded Nucleic Acids Regulate TLR3/4/7 Activation through Interference with Clathrin-Mediated Endocytosis. Sci Rep 2018; 8:15841. [PMID: 30367171 PMCID: PMC6203749 DOI: 10.1038/s41598-018-33960-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022] Open
Abstract
Recognition of nucleic acids by endosomal Toll-like receptors (TLR) is essential to combat pathogens, but requires strict control to limit inflammatory responses. The mechanisms governing this tight regulation are unclear. We found that single-stranded oligonucleotides (ssON) inhibit endocytic pathways used by cargo destined for TLR3/4/7 signaling endosomes. Both ssDNA and ssRNA conferred the endocytic inhibition, it was concentration dependent, and required a certain ssON length. The ssON-mediated inhibition modulated signaling downstream of TLRs that localized within the affected endosomal pathway. We further show that injection of ssON dampens dsRNA-mediated inflammatory responses in the skin of non-human primates. These studies reveal a regulatory role for extracellular ssON in the endocytic uptake of TLR ligands and provide a mechanistic explanation of their immunomodulation. The identified ssON-mediated interference of endocytosis (SOMIE) is a regulatory process that temporarily dampens TLR3/4/7 signaling, thereby averting excessive immune responses.
Collapse
Affiliation(s)
- Peter Järver
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden.
| | - Aleksandra Dondalska
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Candice Poux
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - AnnSofi Sandberg
- Cancer Proteomics Mass Spectrometry, Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65, Stockholm, Sweden
| | - Joseph Bergenstråhle
- Department of Gene Technology, Science for Life Laboratory, Royal Institute of Technology, 171 65, Solna, Sweden
| | - Annette E Sköld
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Nathalie Dereuddre-Bosquet
- CEA, -Université Paris Sud-Inserm U1184, IDMIT Department, Institut de Biologie Francois Jacob (IBFJ), 922 60, Fontenay-aux-Roses, France
| | - Fréderic Martinon
- CEA, -Université Paris Sud-Inserm U1184, IDMIT Department, Institut de Biologie Francois Jacob (IBFJ), 922 60, Fontenay-aux-Roses, France
| | - Sandra Pålsson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Eman Zaghloul
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Stockholm, Sweden
| | - David Brodin
- Bioinformatics and Expression Analysis core facility, Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Stockholm, Sweden
| | - Birgitta Sander
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Kim A Lennox
- Integrated DNA Technologies Inc, Coralville, 52241, Iowa, USA
| | - Mark A Behlke
- Integrated DNA Technologies Inc, Coralville, 52241, Iowa, USA
| | - Samir El-Andaloussi
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Stockholm, Sweden.,Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3PT, Oxford, UK
| | - Janne Lehtiö
- Cancer Proteomics Mass Spectrometry, Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65, Stockholm, Sweden
| | - Joakim Lundeberg
- Department of Gene Technology, Science for Life Laboratory, Royal Institute of Technology, 171 65, Solna, Sweden
| | - Roger LeGrand
- CEA, -Université Paris Sud-Inserm U1184, IDMIT Department, Institut de Biologie Francois Jacob (IBFJ), 922 60, Fontenay-aux-Roses, France
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden.
| |
Collapse
|
7
|
Manavalan B, Shin TH, Kim MO, Lee G. PIP-EL: A New Ensemble Learning Method for Improved Proinflammatory Peptide Predictions. Front Immunol 2018; 9:1783. [PMID: 30108593 PMCID: PMC6079197 DOI: 10.3389/fimmu.2018.01783] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/19/2018] [Indexed: 02/03/2023] Open
Abstract
Proinflammatory cytokines have the capacity to increase inflammatory reaction and play a central role in first line of defence against invading pathogens. Proinflammatory inducing peptides (PIPs) have been used as an antineoplastic agent, an antibacterial agent and a vaccine in immunization therapies. Due to the advancement in sequence technologies that resulted an avalanche of protein sequence data. Therefore, it is necessary to develop an automated computational method to enable fast and accurate identification of novel PIPs within the vast number of candidate proteins and peptides. To address this, we proposed a new predictor, PIP-EL, for predicting PIPs using the strategy of ensemble learning (EL). Our benchmarking dataset is imbalanced. Thus, we applied a random under-sampling technique to generate 10 balanced models for each composition. Technically, PIP-EL is the fusion of 50 independent random forest (RF) models, where each of the five different compositions, including amino acid, dipeptide, composition-transition-distribution, physicochemical properties, and amino acid index contains 10 RF models. PIP-EL achieves the Matthews' correlation coefficient (MCC) of 0.435 in a 5-fold cross-validation test, which is ~2-5% higher than that of the individual classifiers and hybrid feature-based classifier. Furthermore, we evaluate the performance of PIP-EL on the independent dataset, showing that our method outperforms the existing method and two different machine learning methods developed in this study, with an MCC of 0.454. These results indicate that PIP-EL will be a useful tool for predicting PIPs and for researchers working in the field of peptide therapeutics and immunotherapy. The user-friendly web server, PIP-EL, is freely accessible.
Collapse
Affiliation(s)
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
8
|
Manavalan B, Govindaraj RG, Shin TH, Kim MO, Lee G. iBCE-EL: A New Ensemble Learning Framework for Improved Linear B-Cell Epitope Prediction. Front Immunol 2018; 9:1695. [PMID: 30100904 PMCID: PMC6072840 DOI: 10.3389/fimmu.2018.01695] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/10/2018] [Indexed: 11/13/2022] Open
Abstract
Identification of B-cell epitopes (BCEs) is a fundamental step for epitope-based vaccine development, antibody production, and disease prevention and diagnosis. Due to the avalanche of protein sequence data discovered in postgenomic age, it is essential to develop an automated computational method to enable fast and accurate identification of novel BCEs within vast number of candidate proteins and peptides. Although several computational methods have been developed, their accuracy is unreliable. Thus, developing a reliable model with significant prediction improvements is highly desirable. In this study, we first constructed a non-redundant data set of 5,550 experimentally validated BCEs and 6,893 non-BCEs from the Immune Epitope Database. We then developed a novel ensemble learning framework for improved linear BCE predictor called iBCE-EL, a fusion of two independent predictors, namely, extremely randomized tree (ERT) and gradient boosting (GB) classifiers, which, respectively, uses a combination of physicochemical properties (PCP) and amino acid composition and a combination of dipeptide and PCP as input features. Cross-validation analysis on a benchmarking data set showed that iBCE-EL performed better than individual classifiers (ERT and GB), with a Matthews correlation coefficient (MCC) of 0.454. Furthermore, we evaluated the performance of iBCE-EL on the independent data set. Results show that iBCE-EL significantly outperformed the state-of-the-art method with an MCC of 0.463. To the best of our knowledge, iBCE-EL is the first ensemble method for linear BCEs prediction. iBCE-EL was implemented in a web-based platform, which is available at http://thegleelab.org/iBCE-EL. iBCE-EL contains two prediction modes. The first one identifying peptide sequences as BCEs or non-BCEs, while later one is aimed at providing users with the option of mining potential BCEs from protein sequences.
Collapse
Affiliation(s)
| | - Rajiv Gandhi Govindaraj
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea.,Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea.,Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
9
|
Manavalan B, Shin TH, Kim MO, Lee G. AIPpred: Sequence-Based Prediction of Anti-inflammatory Peptides Using Random Forest. Front Pharmacol 2018; 9:276. [PMID: 29636690 PMCID: PMC5881105 DOI: 10.3389/fphar.2018.00276] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/12/2018] [Indexed: 12/31/2022] Open
Abstract
The use of therapeutic peptides in various inflammatory diseases and autoimmune disorders has received considerable attention; however, the identification of anti-inflammatory peptides (AIPs) through wet-lab experimentation is expensive and often time consuming. Therefore, the development of novel computational methods is needed to identify potential AIP candidates prior to in vitro experimentation. In this study, we proposed a random forest (RF)-based method for predicting AIPs, called AIPpred (AIP predictor in primary amino acid sequences), which was trained with 354 optimal features. First, we systematically studied the contribution of individual composition [amino acid-, dipeptide composition (DPC), amino acid index, chain-transition-distribution, and physicochemical properties] in AIP prediction. Since the performance of the DPC-based model is significantly better than that of other composition-based models, we applied a feature selection protocol on this model and identified the optimal features. AIPpred achieved an area under the curve (AUC) value of 0.801 in a 5-fold cross-validation test, which was ∼2% higher than that of the control RF predictor trained with all DPC composition features, indicating the efficiency of the feature selection protocol. Furthermore, we evaluated the performance of AIPpred on an independent dataset, with results showing that our method outperformed an existing method, as well as 3 different machine learning methods developed in this study, with an AUC value of 0.814. These results indicated that AIPpred will be a useful tool for predicting AIPs and might efficiently assist the development of AIP therapeutics and biomedical research. AIPpred is freely accessible at www.thegleelab.org/AIPpred.
Collapse
Affiliation(s)
| | - Tae H Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea.,Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Myeong O Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea.,Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
10
|
Manavalan B, Shin TH, Lee G. PVP-SVM: Sequence-Based Prediction of Phage Virion Proteins Using a Support Vector Machine. Front Microbiol 2018; 9:476. [PMID: 29616000 PMCID: PMC5864850 DOI: 10.3389/fmicb.2018.00476] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/28/2018] [Indexed: 12/29/2022] Open
Abstract
Accurately identifying bacteriophage virion proteins from uncharacterized sequences is important to understand interactions between the phage and its host bacteria in order to develop new antibacterial drugs. However, identification of such proteins using experimental techniques is expensive and often time consuming; hence, development of an efficient computational algorithm for the prediction of phage virion proteins (PVPs) prior to in vitro experimentation is needed. Here, we describe a support vector machine (SVM)-based PVP predictor, called PVP-SVM, which was trained with 136 optimal features. A feature selection protocol was employed to identify the optimal features from a large set that included amino acid composition, dipeptide composition, atomic composition, physicochemical properties, and chain-transition-distribution. PVP-SVM achieved an accuracy of 0.870 during leave-one-out cross-validation, which was 6% higher than control SVM predictors trained with all features, indicating the efficiency of the feature selection method. Furthermore, PVP-SVM displayed superior performance compared to the currently available method, PVPred, and two other machine-learning methods developed in this study when objectively evaluated with an independent dataset. For the convenience of the scientific community, a user-friendly and publicly accessible web server has been established at www.thegleelab.org/PVP-SVM/PVP-SVM.html.
Collapse
Affiliation(s)
| | - Tae H Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea.,Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea.,Institute of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
11
|
Manavalan B, Shin TH, Lee G. DHSpred: support-vector-machine-based human DNase I hypersensitive sites prediction using the optimal features selected by random forest. Oncotarget 2018; 9:1944-1956. [PMID: 29416743 PMCID: PMC5788611 DOI: 10.18632/oncotarget.23099] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
DNase I hypersensitive sites (DHSs) are genomic regions that provide important information regarding the presence of transcriptional regulatory elements and the state of chromatin. Therefore, identifying DHSs in uncharacterized DNA sequences is crucial for understanding their biological functions and mechanisms. Although many experimental methods have been proposed to identify DHSs, they have proven to be expensive for genome-wide application. Therefore, it is necessary to develop computational methods for DHS prediction. In this study, we proposed a support vector machine (SVM)-based method for predicting DHSs, called DHSpred (DNase I Hypersensitive Site predictor in human DNA sequences), which was trained with 174 optimal features. The optimal combination of features was identified from a large set that included nucleotide composition and di- and trinucleotide physicochemical properties, using a random forest algorithm. DHSpred achieved a Matthews correlation coefficient and accuracy of 0.660 and 0.871, respectively, which were 3% higher than those of control SVM predictors trained with non-optimized features, indicating the efficiency of the feature selection method. Furthermore, the performance of DHSpred was superior to that of state-of-the-art predictors. An online prediction server has been developed to assist the scientific community, and is freely available at: http://www.thegleelab.org/DHSpred.html.
Collapse
Affiliation(s)
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
12
|
Han C, Li Q, Zhang Z, Huang J. Characterization, expression, and evolutionary analysis of new TLR3 and TLR5M genes cloned from the spiny eel Mastacembelus armatus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 77:174-187. [PMID: 28821419 DOI: 10.1016/j.dci.2017.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/12/2017] [Accepted: 08/13/2017] [Indexed: 06/07/2023]
Abstract
Toll-like receptors (TLRs) play an important role in innate and adaptive immunity. Here, we identify two new TLRs from the spiny eel Mastacembelus armatus (TLR3 and membrane TLR5M). Both MaTLR3 and MaTLR5M were expressed in all tested tissues; expression was highest in liver and spleen, respectively. After infection with Vibrio parahaemolyticus, expression of both TLRs fluctuated and differed significantly from controls at several time points. The predicted three-dimensional model of the MaTLR3 and MaTLR5M proteins indicates that most sites under positive selection were located in the extracellular domains of TLRs. Evolutionary analysis detected positively selected sites in the ancestral lineages of vertebrates, amphibians and reptiles. Multiple ML methods recovered 10 positively selected sites in teleost TLR3 and 24 in TLR5M, and most sites were located in leucine-rich repeat domain, possibly related to an "arms-race" co-evolution with pathogens.
Collapse
Affiliation(s)
- Chong Han
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Qiang Li
- School of Life Sciences, Guangzhou University, Guangzhou, PR China
| | - Zhipeng Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Jianrong Huang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
13
|
Manavalan B, Basith S, Shin TH, Choi S, Kim MO, Lee G. MLACP: machine-learning-based prediction of anticancer peptides. Oncotarget 2017; 8:77121-77136. [PMID: 29100375 PMCID: PMC5652333 DOI: 10.18632/oncotarget.20365] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/13/2017] [Indexed: 01/25/2023] Open
Abstract
Cancer is the second leading cause of death globally, and use of therapeutic peptides to target and kill cancer cells has received considerable attention in recent years. Identification of anticancer peptides (ACPs) through wet-lab experimentation is expensive and often time consuming; therefore, development of an efficient computational method is essential to identify potential ACP candidates prior to in vitro experimentation. In this study, we developed support vector machine- and random forest-based machine-learning methods for the prediction of ACPs using the features calculated from the amino acid sequence, including amino acid composition, dipeptide composition, atomic composition, and physicochemical properties. We trained our methods using the Tyagi-B dataset and determined the machine parameters by 10-fold cross-validation. Furthermore, we evaluated the performance of our methods on two benchmarking datasets, with our results showing that the random forest-based method outperformed the existing methods with an average accuracy and Matthews correlation coefficient value of 88.7% and 0.78, respectively. To assist the scientific community, we also developed a publicly accessible web server at www.thegleelab.org/MLACP.html.
Collapse
Affiliation(s)
| | - Shaherin Basith
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sun Choi
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
14
|
Investigating toxicity specific to adjuvanted vaccines. Regul Toxicol Pharmacol 2017; 91:29-38. [PMID: 28888957 DOI: 10.1016/j.yrtph.2017.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/14/2017] [Accepted: 09/05/2017] [Indexed: 11/22/2022]
Abstract
In an attempt to understand the unique toxicity of adjuvanted vaccines, we studied how toxicity develops over time following vaccine administration. In addition to on- and off-target toxicity typically observed with general pharmaceuticals, we observed toxicity associated with both the generation and the broad action of effectors (antibodies and/or cytotoxic T lymphocytes, CTLs). The impact on effector generation appears to be related to local tolerance specific to the adjuvant. The vaccine immune response by effectors serves to demonstrate species relevance as outlined in the recent WHO guideline on the nonclinical evaluation of adjuvanted vaccines. When regarded as pharmaceuticals that function at sites of local administration, adjuvants have inherent on- and off-target toxicity. On-target toxicity of the adjuvant is typically associated with effector generation, and could vary depending on animal species. Therefore, the use of species with sensitivity to adjuvants described in the WHO guidelines is required to evaluate the toxicity of the vaccine associated with effector generation. Changes in safety pharmacology endpoints would be considered off-target and further studies are conducted only if changes in these endpoints are observed in nonclinical or clinical studies. Thus our decision tree does not recommend the routine conduct of stand-alone safety pharmacology studies.
Collapse
|
15
|
Durai P, Shin HJ, Achek A, Kwon HK, Govindaraj RG, Panneerselvam S, Yesudhas D, Choi J, No KT, Choi S. Toll-like receptor 2 antagonists identified through virtual screening and experimental validation. FEBS J 2017; 284:2264-2283. [PMID: 28570013 DOI: 10.1111/febs.14124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 12/23/2022]
Abstract
Toll-like receptor 2 (TLR2) antagonists are key therapeutic targets because they inhibit several inflammatory diseases caused by surplus TLR2 activation. In this study, we identified two novel nonpeptide TLR2 antagonists, C11 and C13, through pharmacophore-based virtual screening. At 10 μm, the level of interleukin (IL)-8 inhibition by C13 and C11 in human embryonic kidney TLR2 overexpressing cells was comparable to the commercially available TLR2 inhibitor CU-CPT22. In addition, C11 and C13 acted in mouse macrophage-like RAW 264.7 cells as TLR2-specific inhibitors and did not suppress the tumor necrosis factor-α induction by TLR3 and TLR4 activators. Moreover, the two identified compounds bound directly to the human recombinant TLR2 ectodomain, during surface plasmon resonance analysis, and did not affect cell viability in a 3-(4,5-dimethylthiazol-2-yl)-5(3-carboxymethonyphenol)-2-(4-sulfophenyl)-2H-tetrazolium assay. In total, two virtually screened molecules, C11 and C13, were experimentally proven to be effective as TLR2 antagonists, and thus will provide new insights into the structure of TLR2 antagonists, and pave the way for the development of TLR2-targeted drug molecules.
Collapse
Affiliation(s)
| | - Hyeon-Jun Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Asma Achek
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Hyuk-Kwon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | | | | | - Dhanusha Yesudhas
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Jiwon Choi
- Bioinformatics and Molecular Design Research Center, Seoul, Korea
| | - Kyoung Tai No
- Bioinformatics and Molecular Design Research Center, Seoul, Korea.,Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| |
Collapse
|
16
|
Chai HH, Lim D, Suk JE, Choi BH, Cho YM. Design of anti-BVDV drug based on common chemical features, their interaction, and scaffolds of TLR8 agonists. Int J Biol Macromol 2016; 92:1095-1112. [DOI: 10.1016/j.ijbiomac.2016.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 11/16/2022]
|
17
|
Monk BJ, Facciabene A, Brady WE, Aghajanian CA, Fracasso PM, Walker JL, Lankes HA, Manjarrez KL, Danet-Desnoyers GÄH, Bell-McGuinn KM, McCourt CK, Malykhin A, Hershberg RM, Coukos G. Integrative Development of a TLR8 Agonist for Ovarian Cancer Chemoimmunotherapy. Clin Cancer Res 2016; 23:1955-1966. [PMID: 27702821 DOI: 10.1158/1078-0432.ccr-16-1453] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/19/2016] [Accepted: 09/08/2016] [Indexed: 01/04/2023]
Abstract
Purpose: Immunotherapy is an emerging paradigm for the treatment of cancer, but the potential efficacy of many drugs cannot be sufficiently tested in the mouse. We sought to develop a rational combination of motolimod-a novel Toll-like receptor 8 (TLR8) agonist that stimulates robust innate immune responses in humans but diminished responses in mice-with pegylated liposomal doxorubicin (PLD), a chemotherapeutic that induces immunogenic cell death.Experimental Design: We followed an integrative pharmacologic approach including healthy human volunteers, non-human primates, NSG-HIS ("humanized immune system") mice reconstituted with human CD34+ cells, and patients with cancer to test the effects of motolimod and to assess the combination of motolimod with PLD for the treatment of ovarian cancer.Results: The pharmacodynamic effects of motolimod monotherapy in NSG-HIS mice closely mimicked those in non-human primates and healthy human subjects, whereas the effects of the motolimod/PLD combination in tumor-bearing NSG-HIS mice closely mimicked those in patients with ovarian cancer treated in a phase Ib trial (NCT01294293). The NSG-HIS mouse helped elucidate the mechanism of action of the combination and revealed a positive interaction between the two drugs in vivo The combination produced no dose-limiting toxicities in patients with ovarian cancer. Two subjects (15%) had complete responses and 7 subjects (53%) had disease stabilization. A phase II study was consequently initiated.Conclusions: These results are the first to demonstrate the value of pharmacologic approaches integrating the NSG-HIS mouse, non-human primates, and patients with cancer for the development of novel immunomodulatory anticancer agents with human specificity. Clin Cancer Res; 23(8); 1955-66. ©2016 AACR.
Collapse
Affiliation(s)
- Bradley J Monk
- Arizona Oncology (US Oncology Network), University of Arizona College of Medicine-Phoenix, Creighton University School of Medicine at St. Joseph's Hospital, Phoenix, Arizona.
| | | | - William E Brady
- NRG Oncology Statistics and Data Management Center, Roswell Park Cancer Institute, Buffalo, New York
| | | | | | - Joan L Walker
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Heather A Lankes
- NRG Oncology Statistics and Data Management Center, Roswell Park Cancer Institute, Buffalo, New York
| | | | | | | | | | | | | | - George Coukos
- University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Schad J, Voigt CC. Adaptive evolution of virus-sensing toll-like receptor 8 in bats. Immunogenetics 2016; 68:783-795. [PMID: 27502317 PMCID: PMC7079948 DOI: 10.1007/s00251-016-0940-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 07/12/2016] [Indexed: 11/29/2022]
Abstract
Recently, bats have gained attention as potential reservoir hosts for emerging zoonotic single-stranded (ssRNA) viruses that may prove fatal for humans and other mammals. It has been hypothesized that some features of their innate immune system may enable bats to trigger an efficient early immune response. Toll-like receptors (TLRs) represent a first line defense within the innate immune system and lie directly at the host–pathogen interface in targeting specific microbe-molecular patterns. However, the direction and strength of selection acting on TLRs are largely unknown for bats. Here, we studied the selection on viral ssRNA sensing TLR8 based on sequence data of 21 bat species. The major part (63 %) of the TLR8 gene evolved under purifying selection, likely due to functional constraints. We also found evidence for persistent positive selection acting on specific amino acid sites (7 %), especially when compared to viral TLR evolution of other mammals. All of these putatively positively selected codons were located in the ligand-binding ectodomain, some coincidenced or were in close proximity to functional sites, as suggested by the crystallographic structure of the human TLR8. This might contribute to the inter-species variation in the ability to recognize molecular patterns of viruses. TLR8 evolution within bats revealed that branches leading to ancestral and recent lineages evolved under episodic positive selection, indicating selective selection pressures in restricted bat lineages. Altogether, we found that the TLR8 displays extensive sequence variation within bats and that unique features separate them from humans and other mammals.
Collapse
Affiliation(s)
- Julia Schad
- Department of Evolutionary Ecology, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, 10315, Berlin, Germany.
| | - Christian C Voigt
- Department of Evolutionary Ecology, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, 10315, Berlin, Germany
| |
Collapse
|
19
|
Yu D, Wu Y, Xu L, Fan Y, Peng L, Xu M, Yao YG. Identification and characterization of toll-like receptors (TLRs) in the Chinese tree shrew (Tupaia belangeri chinensis). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 60:127-138. [PMID: 26923770 DOI: 10.1016/j.dci.2016.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 02/23/2016] [Indexed: 06/05/2023]
Abstract
In mammals, the toll-like receptors (TLRs) play a major role in initiating innate immune responses against pathogens. Comparison of the TLRs in different mammals may help in understanding the TLR-mediated responses and developing of animal models and efficient therapeutic measures for infectious diseases. The Chinese tree shrew (Tupaia belangeri chinensis), a small mammal with a close relationship to primates, is a viable experimental animal for studying viral and bacterial infections. In this study, we characterized the TLRs genes (tTLRs) in the Chinese tree shrew and identified 13 putative TLRs, which are orthologs of mammalian TLR1-TLR9 and TLR11-TLR13, and TLR10 was a pseudogene in tree shrew. Positive selection analyses using the Maximum likelihood (ML) method showed that tTLR8 and tTLR9 were under positive selection, which might be associated with the adaptation to the pathogen challenge. The mRNA expression levels of tTLRs presented an overall low and tissue-specific pattern, and were significantly upregulated upon Hepatitis C virus (HCV) infection. tTLR4 and tTLR9 underwent alternative splicing, which leads to different transcripts. Phylogenetic analysis and TLR structure prediction indicated that tTLRs were evolutionarily conserved, which might reflect an ancient mechanism and structure in the innate immune response system. Taken together, TLRs had both conserved and unique features in the Chinese tree shrew.
Collapse
Affiliation(s)
- Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Li Peng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
20
|
Monjazeb AM, Kent MS, Grossenbacher SK, Mall C, Zamora AE, Mirsoian A, Chen M, Kol A, Shiao SL, Reddy A, Perks JR, T N Culp W, Sparger EE, Canter RJ, Sckisel GD, Murphy WJ. Blocking Indolamine-2,3-Dioxygenase Rebound Immune Suppression Boosts Antitumor Effects of Radio-Immunotherapy in Murine Models and Spontaneous Canine Malignancies. Clin Cancer Res 2016; 22:4328-40. [PMID: 26979392 DOI: 10.1158/1078-0432.ccr-15-3026] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/28/2016] [Indexed: 01/23/2023]
Abstract
PURPOSE Previous studies demonstrate that intratumoral CpG immunotherapy in combination with radiotherapy acts as an in-situ vaccine inducing antitumor immune responses capable of eradicating systemic disease. Unfortunately, most patients fail to respond. We hypothesized that immunotherapy can paradoxically upregulate immunosuppressive pathways, a phenomenon we term "rebound immune suppression," limiting clinical responses. We further hypothesized that the immunosuppressive enzyme indolamine-2,3-dioxygenase (IDO) is a mechanism of rebound immune suppression and that IDO blockade would improve immunotherapy efficacy. EXPERIMENTAL DESIGN We examined the efficacy and immunologic effects of a novel triple therapy consisting of local radiotherapy, intratumoral CpG, and systemic IDO blockade in murine models and a pilot canine clinical trial. RESULTS In murine models, we observed marked increase in intratumoral IDO expression after treatment with radiotherapy, CpG, or other immunotherapies. The addition of IDO blockade to radiotherapy + CpG decreased IDO activity, reduced tumor growth, and reduced immunosuppressive factors, such as regulatory T cells in the tumor microenvironment. This triple combination induced systemic antitumor effects, decreasing metastases, and improving survival in a CD8(+) T-cell-dependent manner. We evaluated this novel triple therapy in a canine clinical trial, because spontaneous canine malignancies closely reflect human cancer. Mirroring our mouse studies, the therapy was well tolerated, reduced intratumoral immunosuppression, and induced robust systemic antitumor effects. CONCLUSIONS These results suggest that IDO maintains immune suppression in the tumor after therapy, and IDO blockade promotes a local antitumor immune response with systemic consequences. The efficacy and limited toxicity of this strategy are attractive for clinical translation. Clin Cancer Res; 22(17); 4328-40. ©2016 AACR.
Collapse
Affiliation(s)
- Arta M Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California.
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, UC Davis School of Veterinary Medicine, Davis, California
| | | | - Christine Mall
- Department of Dermatology, UC Davis Health Sciences, Sacramento, California
| | - Anthony E Zamora
- Department of Dermatology, UC Davis Health Sciences, Sacramento, California
| | - Annie Mirsoian
- Department of Dermatology, UC Davis Health Sciences, Sacramento, California
| | - Mingyi Chen
- Department of Pathology, UC Davis Health Sciences, Sacramento, California
| | - Amir Kol
- Department of Pathology, Microbiology, and Immunology, UC Davis School of Veterinary Medicine, Davis, California
| | - Stephen L Shiao
- Departments of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Abhinav Reddy
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - Julian R Perks
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - William T N Culp
- Department of Surgical and Radiological Sciences, UC Davis School of Veterinary Medicine, Davis, California
| | - Ellen E Sparger
- Department of Surgical and Radiological Sciences, UC Davis School of Veterinary Medicine, Davis, California
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - Gail D Sckisel
- Department of Dermatology, UC Davis Health Sciences, Sacramento, California
| | - William J Murphy
- Department of Dermatology, UC Davis Health Sciences, Sacramento, California. Division of Hematology and Oncology, Department of Internal Medicine, UC Davis Comprehensive Cancer Center, Sacramento, California
| |
Collapse
|
21
|
Ma Y, Han F, Liang J, Yang J, Shi J, Xue J, Yang L, Li Y, Luo M, Wang Y, Wei J, Liu X. A species-specific activation of Toll-like receptor signaling in bovine and sheep bronchial epithelial cells triggered by Mycobacterial infections. Mol Immunol 2016; 71:23-33. [DOI: 10.1016/j.molimm.2016.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/03/2016] [Accepted: 01/04/2016] [Indexed: 01/29/2023]
|
22
|
Abe Y, Sakai-Kato K, Goda Y. Cell Type-Specific Responses of Peripheral Blood CD14-Positive Monocytes to Liposome-Encapsulated Immunostimulatory siRNA. Biol Pharm Bull 2016; 39:1859-1867. [DOI: 10.1248/bpb.b16-00450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yasuhiro Abe
- Division of Drugs, National Institute of Health Sciences
| | | | - Yukihiro Goda
- Division of Drugs, National Institute of Health Sciences
| |
Collapse
|
23
|
Ganapathi L, Van Haren S, Dowling DJ, Bergelson I, Shukla NM, Malladi SS, Balakrishna R, Tanji H, Ohto U, Shimizu T, David SA, Levy O. The Imidazoquinoline Toll-Like Receptor-7/8 Agonist Hybrid-2 Potently Induces Cytokine Production by Human Newborn and Adult Leukocytes. PLoS One 2015; 10:e0134640. [PMID: 26274907 PMCID: PMC4537157 DOI: 10.1371/journal.pone.0134640] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/11/2015] [Indexed: 11/30/2022] Open
Abstract
Background Newborns and young infants are at higher risk for infections than adults, and manifest suboptimal vaccine responses, motivating a search for novel immunomodulators and/or vaccine adjuvants effective in early life. In contrast to most TLR agonists (TLRA), TLR8 agonists such as imidazoquinolines (IMQs) induce adult-level Th1-polarizing cytokine production from human neonatal cord blood monocytes and are candidate early life adjuvants. We assessed whether TLR8-activating IMQ congeners may differ in potency and efficacy in inducing neonatal cytokine production in vitro, comparing the novel TLR7/8-activating IMQ analogues Hybrid-2, Meta-amine, and Para-amine to the benchmark IMQ resiquimod (R848). Methods TLRA-induced NF-κB activation was measured in TLR-transfected HEK cells. Cytokine production in human newborn cord and adult peripheral blood and in monocyte-derived dendritic cell cultures were measured by ELISA and multiplex assays. X-ray crystallography characterized the interaction of human TLR8 with Hybrid-2. Results Hybrid-2 selectively activated both TLR7 and 8 and was more potent than R848 in inducing adult-like levels of TNF-α, and IL-1β. Consistent with its relatively high in vitro activity, crystallographic studies suggest that absence in Hybrid-2 of an ether oxygen of the C2-ethoxymethyl substituent, which can engage in unfavorable electrostatic and/or dipolar interactions with the carbonyl oxygen of Gly572 in human TLR8, may confer greater efficacy and potency compared to R848. Conclusions Hybrid-2 is a selective and potent TLR7/8 agonist that is a candidate adjuvant for early life immunization.
Collapse
Affiliation(s)
- Lakshmi Ganapathi
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Simon Van Haren
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - David J. Dowling
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Ilana Bergelson
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
| | - Nikunj M. Shukla
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Subbalakshmi S. Malladi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Rajalakshmi Balakrishna
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Hiromi Tanji
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Umeharu Ohto
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Sunil A. David
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Ofer Levy
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
24
|
Darfour-Oduro KA, Megens HJ, Roca AL, Groenen MAM, Schook LB. Adaptive Evolution of Toll-Like Receptors (TLRs) in the Family Suidae. PLoS One 2015; 10:e0124069. [PMID: 25894218 PMCID: PMC4404360 DOI: 10.1371/journal.pone.0124069] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/25/2015] [Indexed: 12/16/2022] Open
Abstract
Members of the family Suidae have diverged over extended evolutionary periods in diverse environments, suggesting that adaptation in response to endemic infectious agents may have occurred. Toll-like receptors (TLRs) comprise a multigene family that acts as the first line of defense against infectious microbes at the host-environment interface. We hypothesized that across the Suidae, positive selection mediated by infectious agents has contributed to the evolution of TLR diversity. Thus, we analyzed Sus scrofa, Sus barbatus, Sus verrucosus, Sus celebensis, Sus scebifrons, Babyrousa babyrussa, Potamochoerus larvatus, Potamochoerus porcus and Phacochoerus africanus genomes. Specifically, analyses were performed to identify evidence of positive selection using Maximum likelihood (ML) methods within a phylogenetic framework for bacterial and viral sensing Suidae TLR extracellular domains. Our analyses did not reveal evidence of positive selection for TLR3 and TLR7, suggesting strong functional conservation among these two genes for members of the Suidae. Positive selection was inferred for Suidae TLR1, TLR2, TLR6 and TLR8 evolution. ML methods identified amino acid sites of the bacterial sensing TLR1, TLR2, TLR6 and the viral sensing TLR8 to be under persistent positive selection. Some of these sites are in close proximity to functionally relevant sites, further strengthening the case for pathogen mediated selection for these sites. The branch leading to the genus Sus demonstrated evidence of episodic positive selection for TLR1, indicating selection mediated by infectious agents encountered within the specific geographic origin of the Sus. These results indicate that species of the Suidae have positively selected residues within functional domains of TLRs reflective of prior infections. Thus, TLR genes represent candidates for experimental validation to determine their functional role in antibacterial and antiviral activity within members of the Suidae.
Collapse
Affiliation(s)
- Kwame A. Darfour-Oduro
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Hendrik-Jan Megens
- Animal Breeding and Genomics Centre, Wageningen University, Wageningen, The Netherlands
| | - Alfred L. Roca
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Martien A. M. Groenen
- Animal Breeding and Genomics Centre, Wageningen University, Wageningen, The Netherlands
| | - Lawrence B. Schook
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois, United States of America
- * E-mail:
| |
Collapse
|
25
|
Khan AR, Amu S, Saunders SP, Hams E, Blackshields G, Leonard MO, Weaver CT, Sparwasser T, Sheils O, Fallon PG. Ligation of TLR7 on CD19(+) CD1d(hi) B cells suppresses allergic lung inflammation via regulatory T cells. Eur J Immunol 2015; 45:1842-54. [PMID: 25763771 DOI: 10.1002/eji.201445211] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/27/2015] [Accepted: 03/10/2015] [Indexed: 01/10/2023]
Abstract
B cells have been described as having the capacity to regulate cellular immune responses and suppress inflammatory processes. One such regulatory B-cell population is defined as IL-10-producing CD19(+) CD1d(hi) cells. Previous work has identified an expansion of these cells in mice infected with the helminth, Schistosoma mansoni. Here, microarray analysis of CD19(+) CD1d(hi) B cells from mice infected with S. mansoni demonstrated significantly increased Tlr7 expression, while CD19(+) CD1d(hi) B cells from uninfected mice also demonstrated elevated Tlr7 expression. Using IL-10 reporter, Il10(-/-) and Tlr7(-/-) mice, we formally demonstrate that TLR7 ligation of CD19(+) CD1d(hi) B cells increases their capacity to produce IL-10. In a mouse model of allergic lung inflammation, the adoptive transfer of TLR7-elicited CD19(+) CD1d(hi) B cells reduced airway inflammation and associated airway hyperresponsiveness. Using DEREG mice to deplete FoxP3(+) T regulatory cells in allergen-sensitized mice, we show that that TLR7-elicited CD19(+) CD1d(hi) B cells suppress airway hyperresponsiveness via a T regulatory cell dependent mechanism. These studies identify that TLR7 stimulation leads to the expansion of IL-10-producing CD19(+) CD1d(hi) B cells, which can suppress allergic lung inflammation via T regulatory cells.
Collapse
Affiliation(s)
- Adnan R Khan
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sylvie Amu
- Institute of Molecular Medicine, School of Medicine, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Sean P Saunders
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Emily Hams
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Gordon Blackshields
- Department of Histopathology, Trinity College Dublin, Sir Patrick Duns Research Laboratory, St. James's Hospital, Dublin, Ireland
| | - Martin O Leonard
- School of Medicine and Medical Sciences, The Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Casey T Weaver
- Department of Pathology, University of Alabama, Birmingham, AL, USA
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hanover, Germany
| | - Orla Sheils
- Department of Histopathology, Trinity College Dublin, Sir Patrick Duns Research Laboratory, St. James's Hospital, Dublin, Ireland
| | - Padraic G Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Institute of Molecular Medicine, School of Medicine, St James's Hospital, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
26
|
Bryant CE, Gay NJ, Heymans S, Sacre S, Schaefer L, Midwood KS. Advances in Toll-like receptor biology: Modes of activation by diverse stimuli. Crit Rev Biochem Mol Biol 2015; 50:359-79. [DOI: 10.3109/10409238.2015.1033511] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Nick J. Gay
- Department of Biochemistry, University of Cambridge, Cambridge, UK,
| | - Stephane Heymans
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium,
- ICIN – Netherlands Heart Institute, Utrecht, The Netherlands,
| | - Sandra Sacre
- Brighton & Sussex Medical School, University of Sussex, Brighton, UK,
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany, and
| | - Kim S. Midwood
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
27
|
Chuang TH, Lai CY, Tseng PH, Yuan CJ, Hsu LC. Development of CpG-oligodeoxynucleotides for effective activation of rabbit TLR9 mediated immune responses. PLoS One 2014; 9:e108808. [PMID: 25269083 PMCID: PMC4182578 DOI: 10.1371/journal.pone.0108808] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 09/01/2014] [Indexed: 12/12/2022] Open
Abstract
CpG-oligodeoxynucleotides (CpG-ODN) are potent immune stimuli being developed for use as adjuvants in different species. Toll-like receptor 9 (TLR9) is the cellular receptor for CpG-ODN in mammalian cells. The CpG-ODN with 18–24 deoxynucleotides that are in current use for human and mouse cells, however, have low activity with rabbit TLR9. Using a cell-based activation assay, we developed a type of CpG-ODN containing a GACGTT or AACGTT motif in 12 phosphorothioate-modified deoxynucleotides with potent stimulatory activity for rabbit TLR9. The developed CpG-ODN have higher activities than other developed CpG-ODN in eliciting antigen-nonspecific immune responses in rabbit splenocytes. When mixed with an NJ85 peptide derived from rabbit hemorrhagic disease virus, they had potent activities to boost an antigen-specific T cell activation and antibody production in rabbits. Compared to Freund’s adjuvant, the developed CpG-ODN are capable of boosting a potent and less toxic antibody response. The results of this study suggest that both the choice of CpG-motif and its length are important factors for CpG-ODN to effectively activate rabbit TLR9 mediated immune responses.
Collapse
Affiliation(s)
- Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan; Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chao-Yang Lai
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Ping-Hui Tseng
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Chiun-Jye Yuan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Li-Chung Hsu
- Institute of Molecular Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Lai CY, Liu YL, Yu GY, Maa MC, Leu TH, Xu C, Luo Y, Xiang R, Chuang TH. TLR7/8 agonists activate a mild immune response in rabbits through TLR8 but not TLR7. Vaccine 2014; 32:5593-9. [PMID: 25131730 DOI: 10.1016/j.vaccine.2014.07.104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/18/2014] [Accepted: 07/31/2014] [Indexed: 01/04/2023]
Abstract
Toll-like receptors 7 (TLR7) and 8 (TLR8) recognize viral single-stranded RNA and small molecular weight agonists to activate anti-viral immune responses. TLR8s from different species have distinct ligand recognitions. For example, human TLR8 is responsive to ligand stimulation, but mouse and rat TLR8 are activated by small molecular weight agonists only in the presence of polyT-oligodeoxynucleotides. TLR7 and TLR8 have been reported to be absent and pseudogenized, respectively, in rabbit (Oryctolagus cuniculus). In this study, we detected the expression of rabbit (rab)TLR8 in immune-cell-associated tissues. Cell proliferation and cytokine expressions in rabbit splenocytes were induced by the TLR7/8 ligand but not by the TLR7 ligands, suggesting that rabTLR8 is functional but rabTLR7 is not. In rabbits, CL075, a TLR7/8 ligand, activated an antigen-specific antibody response, although one not as potent as aluminum salt or Freund's adjuvant. Nevertheless, CL075, alone or in combination with aluminum salt, generates fewer adverse effects than Freund's adjuvant at the injection sites. To further investigate the activation of rabTLR8, we cloned its cDNA. In cell-based assay, this rabTLR8 is activated by TLR7/8 ligand but not activated by TLR7 ligand. Upon stimulation the rabTLR8 had a lower activation compared to the activation of TLR8 from other species, except the mouse and rat TLR8s. Using different deletion and human-rabbit chimeric TLR8 expressing constructs, we showed that an extra peptide in the undefined region results in reduced activity of rabTLR8. These results provide a molecular basis for the mild activities of TLR7/8 ligands in rabbits, and suggest TLR7/8 agonists may provide safer immune stimuli in rabbits than in other non-rodent species.
Collapse
Affiliation(s)
- Chao-Yang Lai
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yi-Ling Liu
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Ming-Chei Maa
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| | - Tzeng-Horng Leu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Congfeng Xu
- Shanghai Institute of Immunology, Institutes of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Yunping Luo
- Department of Immunology, School of Basic Medicine, Peking Union Medical College, Beijing 100005, PR China
| | - Rong Xiang
- School of Medicine, University of Nankai, Tianjin 300071, PR China
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
29
|
Colak E, Leslie A, Zausmer K, Khatamzas E, Kubarenko AV, Pichulik T, Klimosch SN, Mayer A, Siggs O, Hector A, Fischer R, Klesser B, Rautanen A, Frank M, Hill AVS, Manoury B, Beutler B, Hartl D, Simmons A, Weber ANR. RNA and imidazoquinolines are sensed by distinct TLR7/8 ectodomain sites resulting in functionally disparate signaling events. THE JOURNAL OF IMMUNOLOGY 2014; 192:5963-73. [PMID: 24813206 DOI: 10.4049/jimmunol.1303058] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
TLRs 7 and 8 are pattern recognition receptors controlling antiviral host defense or autoimmune diseases. Apart from foreign and host RNA, synthetic RNA oligoribonucleotides (ORN) or small molecules of the imidazoquinoline family activate TLR7 and 8 and are being developed as therapeutic agonists. The structure-function relationships for RNA ORN and imidazoquinoline sensing and consequent downstream signaling by human TLR7 and TLR8 are unknown. Proteome- and genome-wide analyses in primary human monocyte-derived dendritic cells here showed that TLR8 sensing of RNA ORN versus imidazoquinoline translates to ligand-specific differential phosphorylation and transcriptional events. In addition, TLR7 and 8 ectodomains were found to discriminate between RNA ORN and imidazoquinolines by overlapping and nonoverlapping recognition sites to which murine loss-of-function mutations and human naturally occurring hyporesponsive polymorphisms map. Our data suggest TLR7 and TLR8 can signal in two different "modes" depending on the class of ligand. Considering RNA ORN and imidazoquinolines have been regarded as functionally interchangeable, our study highlights important functional incongruities whose understanding will be important for developing TLR7 or 8 therapeutics with desirable effector and safety profiles for in vivo application.
Collapse
Affiliation(s)
- Elif Colak
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Alasdair Leslie
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom;
| | - Kieran Zausmer
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Elham Khatamzas
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Andriy V Kubarenko
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Tica Pichulik
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Sascha N Klimosch
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Alice Mayer
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Owen Siggs
- Department of Genetics, The Scripps Research Institute, La Jolla, CA 92037
| | - Andreas Hector
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Roman Fischer
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Benedikt Klesser
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Anna Rautanen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, United Kingdom
| | - Martin Frank
- Core Facility Molecular Structure Analysis, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Adrian V S Hill
- Wellcome Trust Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, United Kingdom
| | - Bénédicte Manoury
- Institut National de la Santé et de la Recherché Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, 75015 Paris, France; and
| | - Bruce Beutler
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Dominik Hartl
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Alison Simmons
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom;
| | - Alexander N R Weber
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany; Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
30
|
Maharana J, Patra MC, De BC, Sahoo BR, Behera BK, De S, Pradhan SK. Structural insights into the MDP binding and CARD-CARD interaction in zebrafish (Danio rerio) NOD2: a molecular dynamics approach. J Mol Recognit 2014; 27:260-75. [DOI: 10.1002/jmr.2357] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 12/20/2013] [Accepted: 12/20/2013] [Indexed: 01/01/2023]
Affiliation(s)
- Jitendra Maharana
- Biotechnology Laboratory; Central Inland Fisheries Research Institute; Kolkata 700120 West Bengal India
| | - Mahesh Chandra Patra
- BIF-Centre, Department of Bioinformatics; Orissa University of Agriculture and Technology; Bhubaneswar 751003 Odisha India
- Animal Genomics Laboratory, Animal Biotechnology Centre; National Dairy Research Institute; Karnal 132001 Haryana India
| | - Bidhan Chandra De
- Biotechnology Laboratory; Central Inland Fisheries Research Institute; Kolkata 700120 West Bengal India
| | - Bikash Ranjan Sahoo
- BIF-Centre, Department of Bioinformatics; Orissa University of Agriculture and Technology; Bhubaneswar 751003 Odisha India
- Laboratory of Molecular Biophysics, Institute of Protein Research; Osaka University; Osaka Prefecture 5650871 Japan
| | - Bijay Kumar Behera
- Biotechnology Laboratory; Central Inland Fisheries Research Institute; Kolkata 700120 West Bengal India
| | - Sachinandan De
- Animal Genomics Laboratory, Animal Biotechnology Centre; National Dairy Research Institute; Karnal 132001 Haryana India
| | - Sukanta Kumar Pradhan
- BIF-Centre, Department of Bioinformatics; Orissa University of Agriculture and Technology; Bhubaneswar 751003 Odisha India
| |
Collapse
|
31
|
Hipp MM, Shepherd D, Gileadi U, Aichinger MC, Kessler BM, Edelmann MJ, Essalmani R, Seidah NG, Reis e Sousa C, Cerundolo V. Processing of human toll-like receptor 7 by furin-like proprotein convertases is required for its accumulation and activity in endosomes. Immunity 2013; 39:711-21. [PMID: 24138882 PMCID: PMC4839496 DOI: 10.1016/j.immuni.2013.09.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 07/25/2013] [Indexed: 11/18/2022]
Abstract
Toll-like receptor 7 (TLR7) triggers antiviral immune responses by recognizing viral single-stranded RNA in endosomes, but the biosynthetic pathway of human TLR7 (hTLR7) remains unclear. Here, we show that hTLR7 is proteolytically processed and that the C-terminal fragment selectively accumulates in endocytic compartments. hTLR7 processing occurred at neutral pH and was dependent on furin-like proprotein convertases (PCs). Furthermore, TLR7 processing was required for its functional response to TLR7 agonists such as R837 or influenza virus. Notably, proinflammatory and differentiation stimuli increased the expression of furin-like PCs in immune cells, suggesting a positive feedback mechanism for TLR7 processing during infection. Because self-RNA can under certain conditions activate TLR7 and trigger autoimmunity, our results identify furin-like PCs as a possible target to attenuate TLR7-dependent autoimmunity and other immune pathologies.
Collapse
Affiliation(s)
- Madeleine M Hipp
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DU, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Durai P, Govindaraj RG, Choi S. Structure and dynamic behavior of Toll-like receptor 2 subfamily triggered by malarial glycosylphosphatidylinositols of Plasmodium falciparum. FEBS J 2013; 280:6196-212. [PMID: 24090058 PMCID: PMC4163636 DOI: 10.1111/febs.12541] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/15/2013] [Accepted: 09/17/2013] [Indexed: 12/31/2022]
Abstract
Proinflammatory responses by Toll‐like receptors (TLRs) to malaria infection are considered to be a significant factor in suppressing pathogen growth and in disease control. The key protozoan parasite Plasmodium falciparum causes malaria through glycosylphosphatidylinositols (GPIs), which induce the host immune response mainly via TLR2 signalling. Experimental studies have suggested that malarial GPIs from P. falciparum are recognized by the TLR2 subfamily. However, the interaction site and their involvement in the activation mechanism are still unknown. A better understanding of the detailed structure of the TLR–GPI interaction is important for the design of more effective anti‐malarial therapeutics. We used a molecular docking method to predict the binding regions of malarial GPIs with the TLR2 subfamily members. We also employed molecular dynamics simulations and principal component analysis to understand ligand‐induced conformational changes of the TLR2 subfamily. We observed the expected structural changes upon ligand binding, and significant movements were found in loop regions located in the ligand‐binding site of the TLR2 subfamily. We further propose that the binding modes of malarial GPIs are similar to lipopeptides, and that the lipid portions of the ligands could play an essential role in selective dimerization of the TLR2 subfamily.
Collapse
|
33
|
Contrasted evolutionary histories of two Toll-like receptors (Tlr4 and Tlr7) in wild rodents (MURINAE). BMC Evol Biol 2013; 13:194. [PMID: 24028551 PMCID: PMC3848458 DOI: 10.1186/1471-2148-13-194] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/06/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In vertebrates, it has been repeatedly demonstrated that genes encoding proteins involved in pathogen-recognition by adaptive immunity (e.g. MHC) are subject to intensive diversifying selection. On the other hand, the role and the type of selection processes shaping the evolution of innate-immunity genes are currently far less clear. In this study we analysed the natural variation and the evolutionary processes acting on two genes involved in the innate-immunity recognition of Microbe-Associated Molecular Patterns (MAMPs). RESULTS We sequenced genes encoding Toll-like receptor 4 (Tlr4) and 7 (Tlr7), two of the key bacterial- and viral-sensing receptors of innate immunity, across 23 species within the subfamily Murinae. Although we have shown that the phylogeny of both Tlr genes is largely congruent with the phylogeny of rodents based on a comparably sized non-immune sequence dataset, we also identified several potentially important discrepancies. The sequence analyses revealed that major parts of both Tlrs are evolving under strong purifying selection, likely due to functional constraints. Yet, also several signatures of positive selection have been found in both genes, with more intense signal in the bacterial-sensing Tlr4 than in the viral-sensing Tlr7. 92% and 100% of sites evolving under positive selection in Tlr4 and Tlr7, respectively, were located in the extracellular domain. Directly in the Ligand-Binding Region (LBR) of TLR4 we identified two rapidly evolving amino acid residues and one site under positive selection, all three likely involved in species-specific recognition of lipopolysaccharide of gram-negative bacteria. In contrast, all putative sites of LBRTLR7 involved in the detection of viral nucleic acids were highly conserved across rodents. Interspecific differences in the predicted 3D-structure of the LBR of both Tlrs were not related to phylogenetic history, while analyses of protein charges clearly discriminated Rattini and Murini clades. CONCLUSIONS In consequence of the constraints given by the receptor protein function purifying selection has been a dominant force in evolution of Tlrs. Nevertheless, our results show that episodic diversifying parasite-mediated selection has shaped the present species-specific variability in rodent Tlrs. The intensity of diversifying selection was higher in Tlr4 than in Tlr7, presumably due to structural properties of their ligands.
Collapse
|
34
|
Piccoli S, Suku E, Garonzi M, Giorgetti A. Genome-wide Membrane Protein Structure Prediction. Curr Genomics 2013; 14:324-9. [PMID: 24403851 PMCID: PMC3763683 DOI: 10.2174/13892029113149990009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 01/25/2023] Open
Abstract
Transmembrane proteins allow cells to extensively communicate with the external world in a very accurate and specific way. They form principal nodes in several signaling pathways and attract large interest in therapeutic intervention, as the majority pharmaceutical compounds target membrane proteins. Thus, according to the current genome annotation methods, a detailed structural/functional characterization at the protein level of each of the elements codified in the genome is also required. The extreme difficulty in obtaining high-resolution three-dimensional structures, calls for computational approaches. Here we review to which extent the efforts made in the last few years, combining the structural characterization of membrane proteins with protein bioinformatics techniques, could help describing membrane proteins at a genome-wide scale. In particular we analyze the use of comparative modeling techniques as a way of overcoming the lack of high-resolution three-dimensional structures in the human membrane proteome.
Collapse
Affiliation(s)
- Stefano Piccoli
- Applied Bioinformatics Group, Dept. of Biotechnology, University of Verona, strada Le grazie 15, 37134, Verona,
Italy
| | - Eda Suku
- Applied Bioinformatics Group, Dept. of Biotechnology, University of Verona, strada Le grazie 15, 37134, Verona,
Italy
| | - Marianna Garonzi
- Applied Bioinformatics Group, Dept. of Biotechnology, University of Verona, strada Le grazie 15, 37134, Verona,
Italy
| | - Alejandro Giorgetti
- Applied Bioinformatics Group, Dept. of Biotechnology, University of Verona, strada Le grazie 15, 37134, Verona,
Italy
- German Research School for Simulation Sciences, Juelich, Germany
- Center for Biomedical Computing (CBMC), University of Verona, strada Le grazie 8, 37134, Verona, Italy
| |
Collapse
|
35
|
Bryant CE, Monie TP. Mice, men and the relatives: cross-species studies underpin innate immunity. Open Biol 2013; 2:120015. [PMID: 22724060 PMCID: PMC3376732 DOI: 10.1098/rsob.120015] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 03/16/2012] [Indexed: 12/18/2022] Open
Abstract
The innate immune response is the first line of defence against infection. Germ-line-encoded receptors recognize conserved molecular motifs from both exogenous and endogenous sources. Receptor activation results in the initiation of a pro-inflammatory immune response that enables the resolution of infection. Understanding the inner workings of the innate immune system is a fundamental requirement in the search to understand the basis of health and disease. The development of new vaccinations, the treatment of pathogenic infection, the generation of therapies for chronic and auto-inflammatory disorders, and the ongoing battle against cancer, diabetes and atherosclerosis will all benefit from a greater understanding of innate immunity. The rate of knowledge acquisition in this area has been outstanding. It has been underpinned and driven by the use of model organisms. Information obtained from Drospohila melanogaster, knock-out and knock-in mice, and through the use of forward genetics has resulted in discoveries that have opened our eyes to the functionality and complexity of the innate immune system. With the current increase in genomic information, the range of innate immune receptors and pathways of other species available to study is rapidly increasing, and provides a rich resource to continue the development of innate immune research. Here, we address some of the highlights of cross-species study in the innate immune field and consider the benefits of widening the species-field further.
Collapse
Affiliation(s)
- Clare E Bryant
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
36
|
He X, Jia H, Jing Z, Liu D. Recognition of pathogen-associated nucleic acids by endosomal nucleic acid-sensing toll-like receptors. Acta Biochim Biophys Sin (Shanghai) 2013; 45:241-58. [PMID: 23369718 PMCID: PMC7109797 DOI: 10.1093/abbs/gms122] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Foreign nucleic acids, the essential signature molecules of invading pathogens that act as danger signals for host cells, are detected by endosomal nucleic acid-sensing toll-like receptors (TLRs) 3, 7, 8, 9, and 13. These TLRs have evolved to recognize ‘non-self’ nucleic acids within endosomal compartments and rapidly initiate innate immune responses to ensure host protection through induction of type I interferons, inflammatory cytokines, chemokines, and co-stimulatory molecules and maturation of immune cells. In this review, we highlight our understanding of the recognition of pathogen-associated nucleic acids and activation of corresponding signaling pathways through endosomal nucleic acid-sensing TLRs 3, 7, 8, 9, and 13 for an enormous diversity of pathogens, with particular emphasis on their compartmentalization, intracellular trafficking, proteolytic cleavage, autophagy, and regulatory programs.
Collapse
Affiliation(s)
- Xiaobing He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Huaijie Jia
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Zhizhong Jing
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
- Correspondence address. Tel: +86-931-8341979; Fax: +86-931-8340977; E-mail: (Z.J.)/ (D.L.)
| | - Dingxiang Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
- Correspondence address. Tel: +86-931-8341979; Fax: +86-931-8340977; E-mail: (Z.J.)/ (D.L.)
| |
Collapse
|
37
|
Yu H, Jin H, Sun L, Zhang L, Sun G, Wang Z, Yu Y. Toll-like receptor 7 agonists: chemical feature based pharmacophore identification and molecular docking studies. PLoS One 2013; 8:e56514. [PMID: 23526932 PMCID: PMC3603940 DOI: 10.1371/journal.pone.0056514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/10/2013] [Indexed: 12/19/2022] Open
Abstract
Chemical feature based pharmacophore models were generated for Toll-like receptors 7 (TLR7) agonists using HypoGen algorithm, which is implemented in the Discovery Studio software. Several methods tools used in validation of pharmacophore model were presented. The first hypothesis Hypo1 was considered to be the best pharmacophore model, which consists of four features: one hydrogen bond acceptor, one hydrogen bond donor, and two hydrophobic features. In addition, homology modeling and molecular docking studies were employed to probe the intermolecular interactions between TLR7 and its agonists. The results further confirmed the reliability of the pharmacophore model. The obtained pharmacophore model (Hypo1) was then employed as a query to screen the Traditional Chinese Medicine Database (TCMD) for other potential lead compounds. One hit was identified as a potent TLR7 agonist, which has antiviral activity against hepatitis virus in vitro. Therefore, our current work provides confidence for the utility of the selected chemical feature based pharmacophore model to design novel TLR7 agonists with desired biological activity.
Collapse
Affiliation(s)
- Hui Yu
- Department of Laboratory Centre, the Affiliated Tenth People's Hospital, Tongji University, Shanghai, China
- The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Lidan Sun
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Gang Sun
- The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Zhanli Wang
- The First Affiliated Hospital, Baotou Medical College, Baotou, China
- * E-mail: (ZW); (YY)
| | - Yongchun Yu
- Department of Laboratory Centre, the Affiliated Tenth People's Hospital, Tongji University, Shanghai, China
- * E-mail: (ZW); (YY)
| |
Collapse
|
38
|
Li B, Baylink DJ, Deb C, Zannetti C, Rajaallah F, Xing W, Walter MH, Lau KHW, Qin X. 1,25-Dihydroxyvitamin D3 suppresses TLR8 expression and TLR8-mediated inflammatory responses in monocytes in vitro and experimental autoimmune encephalomyelitis in vivo. PLoS One 2013; 8:e58808. [PMID: 23516559 PMCID: PMC3597563 DOI: 10.1371/journal.pone.0058808] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 02/07/2013] [Indexed: 01/16/2023] Open
Abstract
1,25-Dihydroxyvitamin D3 (1,25(OH)2D3) suppresses autoimmunity and inflammation; however, the mechanism of its action has not been fully understood. We sought in this study to determine whether the anti-immune/anti-inflammatory action of 1,25(OH)2D3 is in part mediated through an interplay between 1,25(OH)2D3 and toll-like receptor (TLR)7/8 signaling. 1,25(OH)2D3 treatment prior to and/or following experimental autoimmune encephalomyelitis (EAE) induction effectively reduced inflammatory cytokine expression in the spinal cord and ameliorated EAE. These effects were accompanied with a reduction in expression of several TLRs with the most profound effect observed for TLR8. The expression of TLR8 adaptor protein MyD88 was also significantly reduced by 1,25(OH)2D3. To determine the molecular mechanism by which 1,25(OH)2D3 suppresses EAE induction of TLR8 and inflammatory cytokine expression, we evaluated whether 1,25(OH)2D3 can directly inhibit TLR8 signaling and the resulting inflammatory responses in human THP-1 monocytes. 1,25(OH)2D3 treatment not only significantly reduced TLR8 expression but also the expression or activity of MyD88, IRF-4, IRF-7 and NF-kB in monocytes challenged with TLR8 ligands. TLR8 promoter-luciferase reporter assays indicated that 1,25(OH)2D3 decreases TLR8 mRNA level in part via inhibiting TLR8 gene transcription activity. As a result of inhibition on TLR8 signaling cascade at various stages, 1,25(OH)2D3 significantly diminished the TLR8 target gene expression (TNF-α and IL-1β). In summary, our novel findings suggest that TLR8 is a new target of 1,25(OH)2D3 and may mediate the anti-inflammatory action of 1,25(OH)2D3. Our findings also point to a destructive role of TLR8 in EAE and shed lights on pathogenesis of multiple sclerosis.
Collapse
Affiliation(s)
- Bo Li
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dowling DJ, Tan Z, Prokopowicz ZM, Palmer CD, Matthews MAH, Dietsch GN, Hershberg RM, Levy O. The ultra-potent and selective TLR8 agonist VTX-294 activates human newborn and adult leukocytes. PLoS One 2013; 8:e58164. [PMID: 23483986 PMCID: PMC3587566 DOI: 10.1371/journal.pone.0058164] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 02/01/2013] [Indexed: 11/20/2022] Open
Abstract
Background Newborns display distinct immune responses that contribute to susceptibility to infection and reduced vaccine responses. Toll-like receptor (TLR) agonists may serve as vaccine adjuvants, when given individually or in combination, but responses of neonatal leukocytes to many TLR agonists are diminished. TLR8 agonists are more effective than other TLR agonists in activating human neonatal leukocytes in vitro, but little is known about whether different TLR8 agonists may distinctly activate neonatal leukocytes. We characterized the in vitro immuno-stimulatory activities of a novel benzazepine TLR8 agonist, VTX-294, in comparison to imidazoquinolines that activate TLR8 (R-848; (TLR7/8) CL075; (TLR8/7)), with respect to activation of human newborn and adult leukocytes. Effects of VTX-294 and R-848 in combination with monophosphoryl lipid A (MPLA; TLR4) were also assessed. Methods TLR agonist specificity was assessed using TLR-transfected HEK293 cells expressing a NF-κB reporter gene. TLR agonist-induced cytokine production was measured in human newborn cord and adult peripheral blood using ELISA and multiplex assays. Newborn and adult monocytes were differentiated into monocyte-derived dendritic cells (MoDCs) and TLR agonist-induced activation assessed by cytokine production (ELISA) and co-stimulatory molecule expression (flow cytometry). Results VTX-294 was ∼100x more active on TLR8- than TLR7-transfected HEK cells (EC50, ∼50 nM vs. ∼5700 nM). VTX-294-induced TNF and IL-1β production were comparable in newborn cord and adult peripheral blood, while VTX-294 was ∼ 1 log more potent in inducing TNF and IL-1β production than MPLA, R848 or CL075. Combination of VTX-294 and MPLA induced greater blood TNF and IL-1β responses than combination of R-848 and MPLA. VTX-294 also potently induced expression of cytokines and co-stimulatory molecules HLA-DR and CD86 in human newborn MoDCs. Conclusions VTX-294 is a novel ultra-potent TLR8 agonist that activates newborn and adult leukocytes and is a candidate vaccine adjuvant in both early life and adulthood.
Collapse
Affiliation(s)
- David J. Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Zhen Tan
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pediatrics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zofia M. Prokopowicz
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christine D. Palmer
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Gregory N. Dietsch
- VentiRx Pharmaceuticals, Inc., Seattle, Washington, United States of America
| | - Robert M. Hershberg
- VentiRx Pharmaceuticals, Inc., Seattle, Washington, United States of America
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
40
|
Kokatla HP, Yoo E, Salunke DB, Sil D, Ng CF, Balakrishna R, Malladi SS, Fox LM, David SA. Toll-like receptor-8 agonistic activities in C2, C4, and C8 modified thiazolo[4,5-c]quinolines. Org Biomol Chem 2013; 11:1179-98. [PMID: 23314908 PMCID: PMC3577938 DOI: 10.1039/c2ob26705e] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptor (TLR)-8 agonists typified by the 2-alkylthiazolo[4,5-c]quinolin-4-amine (CL075) chemotype are uniquely potent in activating adaptive immune responses by inducing robust production of T helper 1-polarizing cytokines, suggesting that TLR8-active compounds could be promising candidate vaccine adjuvants, especially for neonatal vaccines. Alkylthiazoloquinolines with methyl, ethyl, propyl and butyl groups at C2 displayed comparable TLR8-agonistic potencies; activity diminished precipitously in the C2-pentyl compound, and higher homologues were inactive. The C2-butyl compound was unique in possessing substantial TLR7-agonistic activity. Analogues with branched alkyl groups at C2 displayed poor tolerance of terminal steric bulk. Virtually all modifications at C8 led to abrogation of agonistic activity. Alkylation on the C4-amine was not tolerated, whereas N-acyl analogues with short acyl groups (other than acetyl) retained TLR8 agonistic activity, but were substantially less water-soluble. Immunization in rabbits with a model subunit antigen adjuvanted with the lead C2-butyl thiazoloquinoline showed enhancements of antigen-specific antibody titers.
Collapse
Affiliation(s)
- Hari Prasad Kokatla
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Euna Yoo
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Deepak B. Salunke
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Diptesh Sil
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Cameron F. Ng
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Rajalakshmi Balakrishna
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Subbalakshmi S. Malladi
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Lauren M. Fox
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| | - Sunil A. David
- Department of Medicinal Chemistry, University of Kansas, Multidisciplinary Research Building, Room 320D, 2030 Becker Drive, Lawrence KS 66047
| |
Collapse
|
41
|
Sarvestani ST, Williams BRG, Gantier MP. Human Toll-like receptor 8 can be cool too: implications for foreign RNA sensing. J Interferon Cytokine Res 2012; 32:350-61. [PMID: 22817608 DOI: 10.1089/jir.2012.0014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Recent advances in our understanding of foreign nucleic acid sensing indicate an important role for the human Toll-like receptor (TLR) 8 in the initiation of immune responses to certain pathogens. However, TLR8, far too often grouped together with TLR7 for its common ability to detect RNA, has a function on its own in the initiation of specific proinflammatory responses to viruses and bacteria. Here, we present an overview of what is currently known of human TLR8 biology, from genetic regulation to its function in innate immunity, and discuss how TLR8 could present novel therapeutic opportunities in viral and cancer diseases.
Collapse
Affiliation(s)
- Soroush T Sarvestani
- Centre for Cancer Research, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | | |
Collapse
|
42
|
Alzabin S, Kong P, Medghalchi M, Palfreeman A, Williams R, Sacre S. Investigation of the role of endosomal Toll-like receptors in murine collagen-induced arthritis reveals a potential role for TLR7 in disease maintenance. Arthritis Res Ther 2012; 14:R142. [PMID: 22691272 PMCID: PMC3446525 DOI: 10.1186/ar3875] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 05/23/2012] [Accepted: 06/12/2012] [Indexed: 12/20/2022] Open
Abstract
Introduction Endosomal toll-like receptors (TLRs) have recently emerged as potential contributors to the inflammation observed in human and rodent models of rheumatoid arthritis (RA). This study aims to evaluate the role of endosomal TLRs and in particular TLR7 in the murine collagen induced arthritis (CIA) model. Methods CIA was induced by injection of collagen in complete Freund's adjuvant. To investigate the effect of endosomal TLRs in the CIA model, mianserin was administered daily from the day of disease onset. The specific role of TLR7 was examined by inducing CIA in TLR7-deficient mice. Disease progression was assessed by measuring clinical score, paw swelling, serum anti-collagen antibodies histological parameters, cytokine production and the percentage of T regulatory (Treg) cells. Results Therapeutic administration of mianserin to arthritic animals demonstrated a highly protective effect on paw swelling and joint destruction. TLR7-/- mice developed a mild arthritis, where the clinical score and paw swelling were significantly compromised in comparison to the control group. The amelioration of arthritis by mianserin and TLR7 deficiency both corresponded with a reduction in IL-17 responses, histological and clinical scores, and paw swelling. Conclusions These data highlight the potential role for endosomal TLRs in the maintenance of inflammation in RA and support the concept of a role for TLR7 in experimental arthritis models. This study also illustrates the potential benefit that may be afforded by therapeutically inhibiting the endosomal TLRs in RA.
Collapse
Affiliation(s)
- Saba Alzabin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, London, W6 8LH, UK
| | | | | | | | | | | |
Collapse
|
43
|
Areal H, Abrantes J, Esteves PJ. Signatures of positive selection in Toll-like receptor (TLR) genes in mammals. BMC Evol Biol 2011; 11:368. [PMID: 22185391 PMCID: PMC3276489 DOI: 10.1186/1471-2148-11-368] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 12/20/2011] [Indexed: 12/13/2022] Open
Abstract
Background Toll-like receptors (TLRs) are a major class of pattern recognition receptors (PRRs) expressed in the cell surface or membrane compartments of immune and non-immune cells. TLRs are encoded by a multigene family and represent the first line of defense against pathogens by detecting foreigner microbial molecular motifs, the pathogen-associated molecular patterns (PAMPs). TLRs are also important by triggering the adaptive immunity in vertebrates. They are characterized by the presence of leucine-rich repeats (LRRs) in the ectodomain, which are associated with the PAMPs recognition. The direct recognition of different pathogens by TLRs might result in different evolutionary adaptations important to understand the dynamics of the host-pathogen interplay. Ten mammal TLR genes, viral (TLR3, 7, 8, 9) and non-viral (TLR1-6, 10), were selected to identify signatures of positive selection that might have been imposed by interacting pathogens and to clarify if viral and non-viral TLRs might display different patterns of molecular evolution. Results By using Maximum Likelihood approaches, evidence of positive selection was found in all the TLRs studied. The number of positively selected codons (PSC) ranged between 2-26 codons (0.25%-2.65%) with the non-viral TLR4 as the receptor with higher percentage of positively selected codons (2.65%), followed by the viral TLR8 (2.50%). The results indicated that viral and non-viral TLRs are similarly under positive selection. Almost all TLRs have at least one PSC located in the LRR ectodomain which underlies the importance of the pathogen recognition by this region. Conclusions Our results are not in line with previous studies on primates and birds that identified more codons under positive selection in non-viral TLRs. This might be explained by the fact that both primates and birds are homogeneous groups probably being affected by only a restricted number of related viruses with equivalent motifs to be recognized. The analyses performed in this work encompassed a large number of species covering some of the most representative mammalian groups - Artiodactyla, Rodents, Carnivores, Lagomorphs and Primates - that are affected by different families of viruses. This might explain the role of adaptive evolution in shaping viral TLR genes.
Collapse
Affiliation(s)
- Helena Areal
- CIBIO-UP, Centro de Investigacao em Biodiversidade e Recursos Geneticos - Universidade do Porto, Campus Agrario de Vairao, Rua Padre Armando Quintas, nr,7, 4485-661 Vairao, Portugal
| | | | | |
Collapse
|