1
|
Shao J, Wang T, Tang C, Yu J, Chen Y, Guo X, Wang H, Zhou L, Zhang G, Li Y, Yu H, Zheng R. The chemokine receptor type 5 inhibitor maraviroc alleviates sepsis-associated liver injury by regulating MAPK/NF-κB signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3655-3666. [PMID: 39352530 PMCID: PMC11978715 DOI: 10.1007/s00210-024-03477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/19/2024] [Indexed: 04/10/2025]
Abstract
Sepsis-related organ damage, as the most intractable problem in intensive care units (ICUs), receives a great deal of attention from healthcare professionals. Sepsis-associated liver injury (SALI) often leads to poor clinical outcomes due to its complex physiological mechanism. In previous studies, chemokine receptor 5 (CCR5) inhibitors were shown to exert unique anti-inflammatory effects. As the therapeutic effect of maraviroc (MVC) on SALI is still unclear, we aimed to explore whether MVC is effective in treating SALI. We established a model of SALI by cecal ligation and puncture (CLP) and intraperitoneally injected 20 mg/kg MVC 2 h after CLP. The results showed that MVC could significantly ameliorate liver injury after CLP. Furthermore, we demonstrated that MVC reduced inflammatory infiltration and apoptosis after SALI. In addition, we found that the function of MVC in reducing inflammation was obtained through the inhibition of the two inflammatory signaling pathways mentioned above. Finally, the JNK agonist AN was chosen for reverse research. As shown by the results, the therapeutic effects of MVC disappeared after AN treatment, indicating that MVC exerted anti-inflammatory and antiapoptotic effects through JNK. Our study revealed that MVC could reduce liver injury after SALI by inhibiting liver inflammation and hepatocyte apoptosis induced by CLP and that MVC exerted diminish inflammatory effects by inhibiting the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Jun Shao
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Tianwei Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Chengbin Tang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Jiangquan Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Ying Chen
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225009, China
| | - Xin Guo
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
| | - Haoran Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Lulu Zhou
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Guozhong Zhang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Yuping Li
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Hailong Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China.
- Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China.
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225009, China.
| | - Ruiqiang Zheng
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China.
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| |
Collapse
|
2
|
Gu S, Maurya S, Lona A, Borrega Roman L, Salanga C, Gonzalez DJ, Kufareva I, Handel TM. Traffic control: Mechanisms of ligand-specific internalization and intracellular distribution of CCR5. Mol Pharmacol 2025; 107:100020. [PMID: 40199068 DOI: 10.1016/j.molpha.2025.100020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 04/10/2025] Open
Abstract
CC chemokine receptor (CCR) 5 promotes inflammatory responses by driving cell migration and scavenging chemokine. A CCR5 inhibitor Maraviroc has been approved for blocking HIV entry; however, inhibitors for the treatment of other diseases have had limited success, likely because of the complexity of CCR5 pharmacology and biology. CCR5 is activated by natural and engineered chemokines that elicit distinct signaling and trafficking responses, including receptor sequestration inside the cell. Intracellular sequestration may be therapeutically exploitable as a strategy for receptor inhibition, but the mechanisms by which different ligands promote receptor intracellular retention versus presence on the cell membrane are poorly understood. In this study, we systematically compared the time-dependent trafficking behavior of CCR5 following stimulation with its endogenous agonist, CCL5, and 2 CCL5 variants that promote CCR5 intracellular retention. Using a broad panel of pharmacologic assays, fluorescence microscopy, and live cell ascorbic acid peroxidase proximity labeling proteomics, we identified distinct ligand-dependent CCR5 trafficking patterns with temporal and spatial resolution. All 3 chemokines internalize CCR5 via β-arrestin-dependent, clathrin-mediated endocytosis but to different extents, with different kinetics and varying dependencies on G protein-coupled receptor kinase subtypes. The agonists differ in their ability to target the receptor to lysosomes for degradation, as well as to the Golgi compartment and the trans-Golgi network, and these trafficking patterns translate into distinct levels of ligand scavenging. The results provide insight into the cellular mechanisms behind CCR5 intracellular sequestration and suggest how trafficking can be exploited for the development of functional antagonists of CCR5. SIGNIFICANCE STATEMENT: CC chemokine receptor (CCR) 5 plays a crucial role in the immune system and is important in numerous physiological and pathological processes such as inflammation, cancer, and transmission of HIV. It responds to different ligands with distinct signaling and trafficking behaviors; notably, some ligands induce retention of the receptor inside the cell. This study reveals the cellular basis for receptor sequestration that can be exploited as a therapeutic strategy for inhibiting CCR5 function.
Collapse
Affiliation(s)
- Siyi Gu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California; Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Svetlana Maurya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California; Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Alexis Lona
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California
| | - Leire Borrega Roman
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California; Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Catherina Salanga
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California; Department of Pharmacology, University of California San Diego, La Jolla, California
| | - David J Gonzalez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California; Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California.
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California; Department of Pharmacology, University of California San Diego, La Jolla, California.
| |
Collapse
|
3
|
Xie J, Idris A, Feng R. The complex interplay between encephalomyocarditis virus and the host defence system. Virulence 2024; 15:2383559. [PMID: 39066684 PMCID: PMC11285270 DOI: 10.1080/21505594.2024.2383559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
A variety of animals can be infected by encephalomyocarditis virus (EMCV). EMCV is the established causative agent of myocarditis and encephalitis in some animals. EMCV causes high fatality in suckling and weaning piglets, making pigs the most susceptible domestic animal species. Importantly, EMCV has zoonotic potential to infect the human population. The ability of the pathogen to avoid and undermine the initial defence mechanism of the host contributes to its virulence and pathogenicity. A large body of literature highlights the intricate strategies employed by EMCV to escape the innate immune machinery to suit its "pathogenic needs." Here, we also provide examples on how EMCV interacts with certain host proteins to dampen the infection process. Hence, this concise review aims to summarize these findings in a compendium of decades of research on this exciting yet underappreciated topic.
Collapse
Affiliation(s)
- Jingying Xie
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Adi Idris
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| |
Collapse
|
4
|
Gu S, Maurya S, Lona A, Borrega-Roman L, Salanga C, Gonzalez DJ, Kufareva I, Handel TM. Ligand-Dependent Mechanisms of CC Chemokine Receptor 5 (CCR5) Trafficking Revealed by APEX2 Proximity Labeling Proteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.01.565224. [PMID: 37961097 PMCID: PMC10635066 DOI: 10.1101/2023.11.01.565224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
CC chemokine receptor 5 (CCR5) promotes inflammatory responses by driving cell migration and scavenging chemokine to shape directional chemokine gradients. A CCR5 inhibitor has been approved for blocking HIV entry into cells. However, targeting CCR5 for the treatment of other diseases has had limited success, likely because of the complexity of CCR5 pharmacology and biology. CCR5 is activated by natural and engineered chemokines that elicit distinct signaling and trafficking responses, including receptor sequestration inside the cell. Intracellular sequestration may be therapeutically exploitable as a strategy for receptor inhibition, but the mechanisms by which different ligands promote receptor retention in the cell versus presence on the cell membrane are poorly understood. We employed live cell ascorbic acid peroxidase (APEX2) proximity labeling and quantitative mass spectrometry proteomics for unbiased discovery of temporally resolved protein neighborhoods of CCR5 following stimulation with its endogenous agonist, CCL5, and two CCL5 variants that promote intracellular retention of the receptor. Along with targeted pharmacological assays, the data reveal distinct ligand-dependent CCR5 trafficking patterns with temporal and spatial resolution. All three chemokines internalize CCR5 via β-arrestin-dependent, clathrin-mediated endocytosis but to different extents, with different kinetics and varying dependencies on GPCR kinase subtypes. The agonists differ in their ability to target the receptor to lysosomes for degradation, as well as to the Golgi compartment and the trans-Golgi network, and these trafficking patterns translate into distinct levels of ligand scavenging. The results provide insight into the cellular mechanisms behind CCR5 intracellular sequestration and suggest how trafficking can be exploited for the development of functional antagonists of CCR5. Significance Statement CCR5 plays a crucial role in the immune system and is important in numerous physiological and pathological processes such as inflammation, cancer and transmission of HIV. It responds to different ligands with distinct signaling and trafficking behaviors; notably some ligands induce retention of the receptor inside the cell. Using time-resolved proximity labeling proteomics and targeted pharmacological experiments, this study reveals the cellular basis for receptor sequestration that can be exploited as a therapeutic strategy for inhibiting CCR5 function.
Collapse
|
5
|
Song YD, Song YH, Liu SX, Huang HR, Zhong YQ. Inhibition of inflammatory cell infiltration by antagonistic peptides that specifically bind to CCR5 via regulation of the NF-κB signaling pathway in rats with colitis. Arch Med Sci 2021; 20:2022-2033. [PMID: 39967933 PMCID: PMC11831354 DOI: 10.5114/aoms/127587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/17/2020] [Indexed: 02/20/2025] Open
Abstract
Introduction CC chemokine receptor 5 (CCR5) and the NF-κB signaling pathway play important roles in the pathophysiology of inflammatory bowel disease (IBD). Previously, we synthesized two peptides (GH and HY peptides) that specifically bind to the first and the second extracellular loops (ECL1 and ECL2, respectively) of CCR5 and preliminarily found an inhibitory effect of these peptides on colitis. However, the specific mechanism by which these two peptides regulate trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats remains unclear. Aim of the study was to further investigate the effect and mechanism of CCR5 binding peptides in rat colitis. Material and methods Experimental colitis was induced by 5% TNBS. CCR5 antagonist peptides were administered intravenously once a day for a week. Histological evaluation, real-time PCR, western blotting and correlation analysis were performed to examine the effects of the CCR5 binding peptides on the infiltration of inflammatory cells and the NF-κB signaling pathway. Results Administration of GH and HY peptides ameliorated mucosal damage and reduced the infiltration of neutrophils, lymphocytes and macrophages in experimental colitis (p < 0.05). The expression of NF-κB-related genes, including p105, p100, IKK and TNF-α, was reduced after GH and HY peptide administration (p < 0.01), and the protein level of TNF-α and the phosphorylation of IKK, IκBα and p65 were also suppressed. Furthermore, the CCR5 antagonist peptides inhibited nuclear translocation of p65. Spearman correlation analysis showed that the infiltration of inflammatory cells was significantly correlated with the NF-κB pathway. Conclusions Antagonistic peptides that specifically bind to ECL1 and ECL2 of CCR5 inhibit the infiltration of neutrophils, lymphocytes and macrophages in the colonic mucosa of Sprague-Dawley rats with TNBS-induced colitis via regulation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yang-Da Song
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes of Sun Yat-Sen University, Guangzhou, China
| | - Yi-Hang Song
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes of Sun Yat-Sen University, Guangzhou, China
| | - Si-Xue Liu
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes of Sun Yat-Sen University, Guangzhou, China
| | - Hua-Rong Huang
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes of Sun Yat-Sen University, Guangzhou, China
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ying-Qiang Zhong
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
6
|
Stafford JD, Yeo CT, Corbett JA. Inhibition of oxidative metabolism by nitric oxide restricts EMCV replication selectively in pancreatic beta-cells. J Biol Chem 2020; 295:18189-18198. [PMID: 33100269 PMCID: PMC7939444 DOI: 10.1074/jbc.ra120.015893] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/08/2020] [Indexed: 01/27/2023] Open
Abstract
Environmental factors, such as viral infection, are proposed to play a role in the initiation of autoimmune diabetes. In response to encephalomyocarditis virus (EMCV) infection, resident islet macrophages release the pro-inflammatory cytokine IL-1β, to levels that are sufficient to stimulate inducible nitric oxide synthase (iNOS) expression and production of micromolar levels of the free radical nitric oxide in neighboring β-cells. We have recently shown that nitric oxide inhibits EMCV replication and EMCV-mediated β-cell lysis and that this protection is associated with an inhibition of mitochondrial oxidative metabolism. Here we show that the protective actions of nitric oxide against EMCV infection are selective for β-cells and associated with the metabolic coupling of glycolysis and mitochondrial oxidation that is necessary for insulin secretion. Inhibitors of mitochondrial respiration attenuate EMCV replication in β-cells, and this inhibition is associated with a decrease in ATP levels. In mouse embryonic fibroblasts (MEFs), inhibition of mitochondrial metabolism does not modify EMCV replication or decrease ATP levels. Like most cell types, MEFs have the capacity to uncouple the glycolytic utilization of glucose from mitochondrial respiration, allowing for the maintenance of ATP levels under conditions of impaired mitochondrial respiration. It is only when MEFs are forced to use mitochondrial oxidative metabolism for ATP generation that mitochondrial inhibitors attenuate viral replication. In a β-cell selective manner, these findings indicate that nitric oxide targets the same metabolic pathways necessary for glucose stimulated insulin secretion for protection from viral lysis.
Collapse
Affiliation(s)
- Joshua D Stafford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
7
|
Stafford JD, Shaheen ZR, Yeo CT, Corbett JA. Inhibition of mitochondrial oxidative metabolism attenuates EMCV replication and protects β-cells from virally mediated lysis. J Biol Chem 2020; 295:16655-16664. [PMID: 32972972 PMCID: PMC7864063 DOI: 10.1074/jbc.ra120.014851] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/06/2020] [Indexed: 12/15/2022] Open
Abstract
Viral infection is one environmental factor that may contribute to the initiation of pancreatic β-cell destruction during the development of autoimmune diabetes. Picornaviruses, such as encephalomyocarditis virus (EMCV), induce a pro-inflammatory response in islets leading to local production of cytokines, such as IL-1, by resident islet leukocytes. Furthermore, IL-1 is known to stimulate β-cell expression of iNOS and production of the free radical nitric oxide. The purpose of this study was to determine whether nitric oxide contributes to the β-cell response to viral infection. We show that nitric oxide protects β-cells against virally mediated lysis by limiting EMCV replication. This protection requires low micromolar, or iNOS-derived, levels of nitric oxide. At these concentrations nitric oxide inhibits the Krebs enzyme aconitase and complex IV of the electron transport chain. Like nitric oxide, pharmacological inhibition of mitochondrial oxidative metabolism attenuates EMCV-mediated β-cell lysis by inhibiting viral replication. These findings provide novel evidence that cytokine signaling in β-cells functions to limit viral replication and subsequent β-cell lysis by attenuating mitochondrial oxidative metabolism in a nitric oxide-dependent manner.
Collapse
Affiliation(s)
- Joshua D Stafford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Zachary R Shaheen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
8
|
Steinbach K, Vincenti I, Egervari K, Kreutzfeldt M, van der Meer F, Page N, Klimek B, Rossitto-Borlat I, Di Liberto G, Muschaweckh A, Wagner I, Hammad K, Stadelmann C, Korn T, Hartley O, Pinschewer DD, Merkler D. Brain-resident memory T cells generated early in life predispose to autoimmune disease in mice. Sci Transl Med 2020; 11:11/498/eaav5519. [PMID: 31243152 DOI: 10.1126/scitranslmed.aav5519] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/13/2019] [Accepted: 04/25/2019] [Indexed: 12/17/2022]
Abstract
Epidemiological studies associate viral infections during childhood with the risk of developing autoimmune disease during adulthood. However, the mechanistic link between these events remains elusive. We report that transient viral infection of the brain in early life, but not at a later age, precipitates brain autoimmune disease elicited by adoptive transfer of myelin-specific CD4+ T cells at sites of previous infection in adult mice. Early-life infection of mouse brains imprinted a chronic inflammatory signature that consisted of brain-resident memory T cells expressing the chemokine (C-C motif) ligand 5 (CCL5). Blockade of CCL5 signaling via C-C chemokine receptor type 5 prevented the formation of brain lesions in a mouse model of autoimmune disease. In mouse and human brain, CCL5+ TRM were located predominantly to sites of microglial activation. This study uncovers how transient brain viral infections in a critical window in life might leave persisting chemotactic cues and create a long-lived permissive environment for autoimmunity.
Collapse
Affiliation(s)
- Karin Steinbach
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Ilena Vincenti
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Kristof Egervari
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Franziska van der Meer
- Department of Neuropathology, University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Nicolas Page
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Bogna Klimek
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Irène Rossitto-Borlat
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Giovanni Di Liberto
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Andreas Muschaweckh
- Klinikum rechts der Isar, Department of Experimental Neuroimmunology, Technical University Munich, 81675 Munich, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Karim Hammad
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Christine Stadelmann
- Department of Neuropathology, University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Thomas Korn
- Klinikum rechts der Isar, Department of Experimental Neuroimmunology, Technical University Munich, 81675 Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), 80539 Munich, Germany
| | - Oliver Hartley
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.,Mintaka Foundation for Medical Research, 1205 Geneva, Switzerland
| | - Daniel D Pinschewer
- Department of Biomedicine-Haus Petersplatz, University of Basel, 4031 Basel, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland. .,Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| |
Collapse
|
9
|
Ellwanger JH, Kulmann-Leal B, Kaminski VDL, Rodrigues AG, Bragatte MADS, Chies JAB. Beyond HIV infection: Neglected and varied impacts of CCR5 and CCR5Δ32 on viral diseases. Virus Res 2020; 286:198040. [PMID: 32479976 PMCID: PMC7260533 DOI: 10.1016/j.virusres.2020.198040] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
CCR5 regulates multiple cell types (e.g., T regulatory and Natural Killer cells) and immune responses. The effects of CCR5, CCR5Δ32 (variant associated with reduced CCR5 expression) and CCR5 antagonists vary between infections. CCR5 affects the pathogenesis of flaviviruses, especially in the brain. The genetic variant CCR5Δ32 increases the risk of symptomatic West Nile virus infection. The triad “CCR5, extracellular vesicles and infections” is an emerging topic.
The interactions between chemokine receptors and their ligands may affect susceptibility to infectious diseases as well as their clinical manifestations. These interactions mediate both the traffic of inflammatory cells and virus-associated immune responses. In the context of viral infections, the human C-C chemokine receptor type 5 (CCR5) receives great attention from the scientific community due to its role as an HIV-1 co-receptor. The genetic variant CCR5Δ32 (32 base-pair deletion in CCR5 gene) impairs CCR5 expression on the cell surface and is associated with protection against HIV infection in homozygous individuals. Also, the genetic variant CCR5Δ32 modifies the CCR5-mediated inflammatory responses in various conditions, such as inflammatory and infectious diseases. CCR5 antagonists mimic, at least in part, the natural effects of the CCR5Δ32 in humans, which explains the growing interest in the potential benefits of using CCR5 modulators for the treatment of different diseases. Nevertheless, beyond HIV infection, understanding the effects of the CCR5Δ32 variant in multiple viral infections is essential to shed light on the potential effects of the CCR5 modulators from a broader perspective. In this context, this review discusses the involvement of CCR5 and the effects of the CCR5Δ32 in human infections caused by the following pathogens: West Nile virus, Influenza virus, Human papillomavirus, Hepatitis B virus, Hepatitis C virus, Poliovirus, Dengue virus, Human cytomegalovirus, Crimean-Congo hemorrhagic fever virus, Enterovirus, Japanese encephalitis virus, and Hantavirus. Subsequently, this review addresses the impacts of CCR5 gene editing and CCR5 modulation on health and viral diseases. Also, this article connects recent findings regarding extracellular vesicles (e.g., exosomes), viruses, and CCR5. Neglected and emerging topics in “CCR5 research” are briefly described, with focus on Rocio virus, Zika virus, Epstein-Barr virus, and Rhinovirus. Finally, the potential influence of CCR5 on the immune responses to coronaviruses is discussed.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunologia Aplicada, Instituto de Ciência e Tecnologia - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos, São Paulo, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Marcelo Alves de Souza Bragatte
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Núcleo de Bioinformática do Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
10
|
Shaheen ZR, Stafford JD, Voss MG, Oleson BJ, Stancill JS, Corbett JA. The location of sensing determines the pancreatic β-cell response to the viral mimetic dsRNA. J Biol Chem 2020; 295:2385-2397. [PMID: 31915247 DOI: 10.1074/jbc.ra119.010267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/11/2019] [Indexed: 12/18/2022] Open
Abstract
Viral infection is an environmental trigger that has been suggested to initiate pancreatic β-cell damage, leading to the development of autoimmune diabetes. Viruses potently activate the immune system and can damage β cells by either directly infecting them or stimulating the production of secondary effector molecules (such as proinflammatory cytokines) during bystander activation. However, how and where β cells recognize viruses is unclear, and the antiviral responses that are initiated following virus recognition are incompletely understood. In this study, we show that the β-cell response to dsRNA, a viral replication intermediate known to activate antiviral responses, is determined by the cellular location of sensing (intracellular versus extracellular) and differs from the cellular response to cytokine treatment. Using biochemical and immunological methods, we show that β cells selectively respond to intracellular dsRNA by expressing type I interferons (IFNs) and inducing apoptosis, but that they do not respond to extracellular dsRNA. These responses differ from the activities of cytokines on β cells, which are mediated by inducible nitric oxide synthase expression and β-cell production of nitric oxide. These findings provide evidence that the antiviral activities of type I IFN production and apoptosis are elicited in β cells via the recognition of intracellular viral replication intermediates and that β cells lack the capacity to respond to extracellular viral intermediates known to activate innate immune responses.
Collapse
Affiliation(s)
- Zachary R Shaheen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Joshua D Stafford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Michael G Voss
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Bryndon J Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.
| |
Collapse
|
11
|
Yu X, Wang H, Shao H, Zhang C, Ju X, Yang J. PolyI:C Upregulated CCR5 and Promoted THP-1-Derived Macrophage Chemotaxis via TLR3/JMJD1A Signalling. CELL JOURNAL 2019; 22:325-333. [PMID: 31863658 PMCID: PMC6947015 DOI: 10.22074/cellj.2020.6713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 02/15/2019] [Indexed: 12/03/2022]
Abstract
Objective This study aimed to evaluate the specific roles of polyinosinic:polycytidylic acid (polyI:C) in macrophage
chemotaxis and reveal the potential regulatory mechanisms related to chemokine receptor 5 (CCR5).
Materials and Methods In this experimental study, THP-1-derived macrophages (THP1-Mφs) induced from THP-
1 monocytes were treated with 25 μg/mL polyI:C. Toll-like receptor 3 (TLR3), Jumonji domain-containing protein
(JMJD)1A, and JMJD1C small interfering RNA (siRNAs) were transfected into THP1-Mφs. Quantitative real-time
reverse transcriptase polymerase chain reaction (qRT-PCR) was used to detect the expression levels of TLR3, CCR5,
23 Jumonji C domain-containing histone demethylase family members, JMJD1A, and JMJD1C in THP1-Mφs with
different siRNAs transfections. Western blot was performed to detect JMJD1A, JMJD1C, H3K9me2, and H3K9me3
expressions. A transwell migration assay was conducted to detect THP1-Mφ chemotaxis toward chemokine ligand 3
(CCL3). A chromatin immunoprecipitation (ChIP) assay was performed to detect H3K9me2-CCR5 complexes in THP1-
Mφs.
Results PolyI:C significantly upregulated CCR5 in THP1-Mφs and promoted chemotaxis toward CCL3 (P<0.05);
these effects were significantly inhibited by TLR3 siRNA (P<0.01). JMJD1A and JMJD1C expression was significantly
upregulated in polyI:C-stimulated THP1-Mφs, while only JMJD1A siRNA decreased CCR5 expression (P<0.05).
JMJD1A siRNA significantly increased H3K9me2 expression in THP1-Mφs but not in polyI:C-stimulated THP1-Mφs.
The ChIP result revealed that polyI:C significantly downregulated H3K9me2 in the promoter region of CCR5 in THP1-
Mφs.
Conclusion PolyI:C can enhance THP1-Mφ chemotaxis toward CCL3 regulated by TLR3/JMJD1A signalling and
activate CCR5 expression by reducing H3K9me2 in the promoter region of CCR5.
Collapse
Affiliation(s)
- Xiaoxiao Yu
- Department of Paediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huayang Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hongjia Shao
- Department of Paediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Cuijuan Zhang
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiuli Ju
- Department of Paediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jie Yang
- Department of Paediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China. Electronic Address
| |
Collapse
|
12
|
Shaheen ZR, Christmann BS, Stafford JD, Moran JM, Buller RML, Corbett JA. CCR5 is a required signaling receptor for macrophage expression of inflammatory genes in response to viral double-stranded RNA. Am J Physiol Regul Integr Comp Physiol 2019; 316:R525-R534. [PMID: 30811246 DOI: 10.1152/ajpregu.00019.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Double-stranded (ds) RNA, both synthetic and produced during virus replication, rapidly stimulates MAPK and NF-κB signaling that results in expression of the inflammatory genes inducible nitric oxide synthase, cyclooxygenase 2, and IL-1β by macrophages. Using biochemical and genetic approaches, we have identified the chemokine ligand-binding C-C chemokine receptor type 5 (CCR5) as a cell surface signaling receptor required for macrophage expression of inflammatory genes in response to dsRNA. Activation of macrophages by synthetic dsRNA does not require known dsRNA receptors, as poly(inosinic:cytidylic) acid [poly(I:C)] activates signaling pathways leading to expression of inflammatory genes to similar levels in wild-type and Toll-like receptor 3- or melanoma differentiation antigen 5-deficient macrophages. In contrast, macrophage activation in response to poly(I:C) is attenuated in macrophages isolated from mice lacking CCR5. These findings support a role for CCR5 as a cell surface signaling receptor that participates in activation of inflammatory genes in macrophages in response to the viral dsRNA mimetic poly(inosinic:cytidylic) acid by pathways that are distinct from classical dsRNA receptor-mediated responses.
Collapse
Affiliation(s)
- Zachary R Shaheen
- Department of Biochemistry, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Benjamin S Christmann
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine , St. Louis, Missouri
| | - Joshua D Stafford
- Department of Biochemistry, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Jason M Moran
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine , St. Louis, Missouri
| | - R Mark L Buller
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine , St. Louis, Missouri
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
13
|
Shaheen ZR, Naatz A, Corbett JA. CCR5-Dependent Activation of mTORC1 Regulates Translation of Inducible NO Synthase and COX-2 during Encephalomyocarditis Virus Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:4406-14. [PMID: 26408666 DOI: 10.4049/jimmunol.1500704] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/27/2015] [Indexed: 11/19/2022]
Abstract
Encephalomyocarditis virus (EMCV) infection of macrophages results in the expression of a number of inflammatory and antiviral genes, including inducible NO synthase (iNOS) and cyclooxygenase (COX)-2. EMCV-induced macrophage activation has been shown to require the presence of CCR5 and the activation of PI3K-dependent signaling cascades. The purpose of this study was to determine the role of PI3K in regulating the macrophage responses to EMCV. We show that PI3K regulates EMCV-stimulated iNOS and COX-2 expression by two independent mechanisms. In response to EMCV infection, Akt is activated and regulates the translation of iNOS and COX-2 through the mammalian target of rapamycin complex (mTORC)1. The activation of mTORC1 during EMCV infection is CCR5-dependent and appears to function in a manner that promotes the translation of iNOS and COX-2. CCR5-dependent mTORC1 activation functions as an antiviral response, as mTORC1 inhibition increases the expression of EMCV polymerase. PI3K also regulates the transcriptional induction of iNOS and COX-2 in response to EMCV infection by a mechanism that is independent of Akt and mTORC1 regulation. These findings indicate that macrophage expression of the inflammatory genes iNOS and COX-2 occurs via PI3K- and Akt-dependent translational control of mTORC1 and PI3K-dependent, Akt-independent transcriptional control.
Collapse
Affiliation(s)
- Zachary R Shaheen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI 53226
| | - Aaron Naatz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI 53226
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI 53226
| |
Collapse
|
14
|
Shaheen ZR, Corbett JA. Macrophage Expression of Inflammatory Genes in Response to EMCV Infection. Biomolecules 2015; 5:1938-54. [PMID: 26295266 PMCID: PMC4598781 DOI: 10.3390/biom5031938] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/06/2015] [Accepted: 08/08/2015] [Indexed: 12/24/2022] Open
Abstract
The expression and production of type 1 interferon is the classic cellular response to virus infection. In addition to this antiviral response, virus infection also stimulates the production of proinflammatory mediators. In this review, the pathways controlling the induction of inflammatory genes and the roles that these inflammatory mediators contribute to host defense against viral pathogens will be discussed. Specific focus will be on the role of the chemokine receptor CCR5, as a signaling receptor controlling the activation of pathways leading to virus-induced inflammatory gene expression.
Collapse
Affiliation(s)
- Zachary R Shaheen
- Department of Biochemistry, Medical College of Wisconsin, 8701 W. Watertown Plank Rd, Milwaukee, WI 53226, USA.
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, 8701 W. Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
15
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 PMCID: PMC3880466 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 691] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Burke SJ, Goff MR, Lu D, Proud D, Karlstad MD, Collier JJ. Synergistic Expression of the CXCL10 Gene in Response to IL-1β and IFN-γ Involves NF-κB, Phosphorylation of STAT1 at Tyr701, and Acetylation of Histones H3 and H4. THE JOURNAL OF IMMUNOLOGY 2013; 191:323-36. [DOI: 10.4049/jimmunol.1300344] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
17
|
Portales P, Psomas KC, Tuaillon E, Mura T, Vendrell JP, Eliaou JF, Reynes J, Corbeau P. The intensity of immune activation is linked to the level of CCR5 expression in human immunodeficiency virus type 1-infected persons. Immunology 2012; 137:89-97. [PMID: 22862553 DOI: 10.1111/j.1365-2567.2012.03609.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Immune activation is a main driver of AIDS- and non-AIDS-linked morbidities in the course of HIV-1 infection. As CCR5, the main HIV-1 co-receptor, is not only a chemokine receptor but also a co-activation molecule expressed at the surface of T cells, it could be directly involved in this immune activation. To test this hypothesis, we measured by flow cytometry the mean number of CCR5 molecules at the surface of non-activated CD4(+) T cells (CCR5 density), which determines the intensity of CCR5 signalling, and the percentage of CD8(+) T cells over-expressing CD38 (CD38 expression), a major marker of immune activation, in the blood of 67 HIV-1-infected, non-treated individuals. CCR5 density was correlated with CD38 expression independently of viral load (P=0.016). CCR5 density remained unchanged after highly active anti-retroviral therapy (HAART) introduction or cessation, whereas CD38 expression decreased and increased, respectively. Moreover, pre-therapeutic CCR5 density was highly predictive (r=0.736, P<10(-4) ) of residual CD38 over-expression after 9 months of HAART. Hence, CCR5 might play an immunological role in HIV-1 infection as a driver of immune activation. This could explain why CCR5 antagonists may have an inhibitory effect on immune activation.
Collapse
Affiliation(s)
- Pierre Portales
- Département d'Immunologie, CHU de Montpellier, Université Montpellier 1, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
The encephalomyocarditis virus (EMCV) is a small non-enveloped single-strand RNA virus, the causative agent of not only myocarditis and encephalitis, but also neurological diseases, reproductive disorders and diabetes in many mammalian species. EMCV pathogenesis appears to be viral strain- and host-specific, and a better understanding of EMCV virulence factors is increasingly required. Indeed, EMCV is often used as a model for diabetes and viral myocarditis, and is also widely used in immunology as a double-stranded RNA stimulus in the study of Toll-like as well as cytosolic receptors. However, EMCV virulence and properties have often been neglected. Moreover, EMCV is able to infect humans albeit with a low morbidity. Progress on xenografts, such as pig heart transplantation in humans, has raised safety concerns that need to be explored. In this review we will highlight the biology of EMCV and all known and potential virulence factors.
Collapse
Affiliation(s)
- Margot Carocci
- Microbiology Immunology Department, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|