1
|
Poppenborg T, Saljic A, Bruns F, Abu-Taha I, Dobrev D, Fender AC. A short history of the atrial NLRP3 inflammasome and its distinct role in atrial fibrillation. J Mol Cell Cardiol 2025; 202:13-23. [PMID: 40057301 DOI: 10.1016/j.yjmcc.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 04/23/2025]
Abstract
Inflammasomes are multiprotein complexes of the innate immune system that mediate inflammatory responses to infection and to local and systemic stress and tissue injury. The principal function is to facilitate caspase-1 auto-activation and subsequently maturation and release of the effectors interleukin (IL)-1β and IL-18. The atrial-specific NLRP3 inflammasome is a unifying causal feature of atrial fibrillation (AF) development, progression and recurrence after ablation. Many AF-associated risk factors and co-morbidities converge mechanistically on the activation of this central inflammatory signaling platform. This review presents the historical conceptual development of a distinct atrial inflammasome and its potential causal involvement in AF. We follow the early observations linking systemic and local inflammation with AF, to the emergence of an atrial-intrinsic NLRP3 inflammasome operating within not just immune cells but also in resident atrial fibroblasts and cardiomyocytes. We outline the key developments in understanding how the atrial NLRP3 inflammasome and its effector IL-1β contribute causally to cellular and tissue-level arrhythmogenesis in different pathological settings, and outline candidate therapeutic concepts verified in preclinical models of atrial cardiomyopathy and AF.
Collapse
Affiliation(s)
| | - Arnela Saljic
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Florian Bruns
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Issam Abu-Taha
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Anke C Fender
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2
|
Liao D, Su X, Wang J, Yu J, Luo H, Tian W, Ye Z, He J. Pushing the envelope: Immune mechanism and application landscape of macrophage-activating lipopeptide-2. Front Immunol 2023; 14:1113715. [PMID: 36761746 PMCID: PMC9902699 DOI: 10.3389/fimmu.2023.1113715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/10/2023] [Indexed: 01/25/2023] Open
Abstract
Mycoplasma fermentans can cause respiratory diseases, arthritis, genitourinary tract infections, and chronic fatigue syndrome and have been linked to the development of the human immunodeficiency virus. Because mycoplasma lacks a cell wall, its outer membrane lipoproteins are one of the main factors that induce inflammation in the organism and contribute to disease development. Macrophage-activating lipopeptide-2 (MALP-2) modulates the inflammatory response of monocytes/macrophages in a bidirectional fashion, indirectly enhances the cytotoxicity of NK cells, promotes oxidative bursts in neutrophils, upregulates surface markers on lymphocytes, enhances antigen presentation on dendritic cells and induces immune inflammatory responses in sebocytes and mesenchymal cells. MALP-2 is a promising vaccine adjuvant for this application. It also promotes vascular healing and regeneration, accelerates wound and bone healing, suppresses tumors and metastasis, and reduces lung infections and inflammation. MALP-2 has a simple structure, is easy to synthesize, and has promising prospects for clinical application. Therefore, this paper reviews the mechanisms of MALP-2 activation in immune cells, focusing on the application of MALP-2 in animals/humans to provide a basis for the study of pathogenesis in Mycoplasma fermentans and the translation of MALP-2 into clinical applications.
Collapse
Affiliation(s)
- Daoyong Liao
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaoling Su
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Jingyun Wang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Jianwei Yu
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Haodang Luo
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China,Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Wei Tian
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Zufeng Ye
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun He
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China,*Correspondence: Jun He,
| |
Collapse
|
3
|
Komal S, Komal N, Mujtaba A, Wang SH, Zhang LR, Han SN. Potential therapeutic strategies for myocardial infarction: the role of Toll-like receptors. Immunol Res 2022; 70:607-623. [PMID: 35608723 DOI: 10.1007/s12026-022-09290-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/06/2022] [Indexed: 11/27/2022]
Abstract
Myocardial infarction (MI) is a life-threatening condition among patients with cardiovascular diseases. MI increases the risk of stroke and heart failure and is a leading cause of morbidity and mortality worldwide. Several genetic and epigenetic factors contribute to the development of MI, suggesting that further understanding of the pathomechanism of MI might help in the early management and treatment of this disease. Toll-like receptors (TLRs) are well-known members of the pattern recognition receptor (PRR) family and contribute to both adaptive and innate immunity. Collectively, studies suggest that TLRs have a cardioprotective effect. However, prolonged TLR activation in the response to signals generated by damage-associated molecular patterns (DAMPs) results in the release of inflammatory cytokines and contributes to the development and exacerbation of myocardial inflammation, MI, ischemia-reperfusion injury, myocarditis, and heart failure. The objective of this review is to discuss and summarize the association of TLRs with MI, highlighting their therapeutic potential for the development of advanced TLR-targeted therapies for MI.
Collapse
Affiliation(s)
- Sumra Komal
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Nimrah Komal
- Department of Pharmacology, Mohi-Ud-Din Islamic Medical College, Azad Jammu & Kashmir, Mirpur, 10250, Pakistan
| | - Ali Mujtaba
- Department of Pharmacology, Mohi-Ud-Din Islamic Medical College, Azad Jammu & Kashmir, Mirpur, 10250, Pakistan
| | - Shu-Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Sheng-Na Han
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
4
|
Chen F, Wang D, Li X, Wang H. Molecular Mechanisms Underlying Intestinal Ischemia/Reperfusion Injury: Bioinformatics Analysis and In Vivo Validation. Med Sci Monit 2020; 26:e927476. [PMID: 33290384 PMCID: PMC7733309 DOI: 10.12659/msm.927476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury is a serious clinical complication. This study aimed to explore the hub genes and pathways of intestinal I/R injury. Material/Methods GSE96733 from the GEO website was extracted to analyze the differentially expressed genes (DEGs) of intestinal I/R injured and sham-operated mice at 3 h and 6 h after surgery. The DAVID and STRING databases were used to construct functional enrichment analyses of DEGs and the protein–protein interaction (PPI) network. In Cytoscape software, cytoHubba was used to identify hub genes, and MCODE was used for module analysis. Testing by qRT-PCR detected the expression of hub genes in intestinal I/R injury. Western blot analysis detected the key proteins involved with the important pathways of intestinal I/R injury. Results IL-6, IL-10, CXCL1, CXCL2, and IL-1β were identified as critical upregulated genes, while IRF7, IFIT3, IFIT1, Herc6, and Oasl2 were identified as hub genes among the downregulated genes. The qRT-PCR testing showed the expression of critical upregulated genes was significantly increased in intestinal I/R injury (P<0.05), while the expression of hub downregulated genes was notably reduced (P<0.05). The proteins of CXCL1 and CXCR2 were upregulated following intestinal I/R injury (P<0.05) and the CXCL1/CXCR2 axis was involved with intestinal I/R injury. Conclusions The results of the present study identified IL-6, IL-10, CXCL1, CXCL2, IL-1β, IRF7, IFIT3, IFIT1, Herc6, and Oasl2 as hub genes in intestinal I/R injury and identified the involvement of the CXCL1/CXCR2 axis in intestinal I/R injury.
Collapse
Affiliation(s)
- Fengshou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Dan Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xiaoqian Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - He Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
5
|
Chen E, Chen C, Niu Z, Gan L, Wang Q, Li M, Cai X, Gao R, Katakam S, Chen H, Zhang S, Zhou R, Cheng X, Qiu Y, Yu H, Zhu T, Liu J. Poly(I:C) preconditioning protects the heart against myocardial ischemia/reperfusion injury through TLR3/PI3K/Akt-dependent pathway. Signal Transduct Target Ther 2020; 5:216. [PMID: 33154351 PMCID: PMC7644758 DOI: 10.1038/s41392-020-00257-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/28/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
Emerging evidence suggests that Toll-like receptors (TLRs) ligands pretreatment may play a vital role in the progress of myocardial ischemia/reperfusion (I/R) injury. As the ligand of TLR3, polyinosinic-polycytidylic acid (poly(I:C)), a synthetic double-stranded RNA, whether its preconditioning can exhibit a cardioprotective phenotype remains unknown. Here, we report the protective effect of poly(I:C) pretreatment in acute myocardial I/R injury by activating TLR3/PI3K/Akt signaling pathway. Poly(I:C) pretreatment leads to a significant reduction of infarct size, improvement of cardiac function, and downregulation of inflammatory cytokines and apoptotic molecules compared with controls. Subsequently, our data demonstrate that phosphorylation of TLR3 tyrosine residue and its interaction with PI3K is enhanced, and protein levels of phospho-PI3K and phospho-Akt are both increased after poly(I:C) pretreatment, while knock out of TLR3 suppresses the cardioprotection of poly(I:C) preconditioning through a decreased activation of PI3K/Akt signaling. Moreover, inhibition of p85 PI3K by the administration of LY294002 in vivo and knockdown of Akt by siRNA in vitro significantly abolish poly(I:C) preconditioning-induced cardioprotective effect. In conclusion, our results reveal that poly(I:C) preconditioning exhibits essential protection in myocardial I/R injury via its modulation of TLR3, and the downstream PI3K/Akt signaling, which may provide a potential pharmacologic target for perioperative cardioprotection.
Collapse
Affiliation(s)
- Erya Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhendong Niu
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Gan
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Wang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ming Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - XingWei Cai
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Gao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sruthi Katakam
- Institute of Cell Engineering, Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hai Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shu Zhang
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ronghua Zhou
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xu Cheng
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanhua Qiu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai Yu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Zhu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Zhang XY, Huang Z, Li QJ, Zhong GQ, Meng JJ, Wang DX, Tu RH. Ischemic postconditioning attenuates the inflammatory response in ischemia/reperfusion myocardium by upregulating miR‑499 and inhibiting TLR2 activation. Mol Med Rep 2020; 22:209-218. [PMID: 32377693 PMCID: PMC7248531 DOI: 10.3892/mmr.2020.11104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/25/2020] [Indexed: 12/18/2022] Open
Abstract
Toll-like receptor 2 (TLR2)-mediated myocardial inflammation serves an important role in promoting myocardial ischemic/reperfusion (I/R) injury. Previous studies have shown that miR-499 is critical for cardioprotection after ischemic postconditioning (IPostC). Therefore, the present study evaluated the protective effect of IPostC on the myocardium by inhibiting TLR2, and also assessed the involvement of microRNA (miR)-499. Rat hearts were subjected to 30 min of ischemia and 2 h of reperfusion. The IPostC was 3 cycles of 30 sec of reperfusion and 30 sec of re-occlusion prior to reperfusion. In total, 90 rats were randomly divided into six groups (n=15 per group): Sham; I/R; IPostC; miR-499 negative control adeno-associated virus (AAV) vectors + IPostC; miR-499 inhibitor AAV vectors + IPostC; and miR-499 mimic AAV vectors + IPostC. It was identified that IPostC significantly decreased the I/R-induced cardiomyocyte apoptotic index (29.4±2.03% in IPostC vs. 42.64±2.27% in I/R; P<0.05) and myocardial infarct size (48.53±2.49% in IPostC vs. 66.52±3.1% in I/R; P<0.05). Moreover, these beneficial effects were accompanied by increased miR-499 expression levels (as demonstrated by reverse transcription-quantitative PCR) in the myocardial tissue and decreased TLR2, protein kinase C (PKC), interleukin (IL)-1β and IL-6 expression levels (as demonstrated by western blotting and ELISA) in the myocardium and serum. The results indicated that IPostC + miR-499 mimics significantly inhibited inflammation and the PKC signaling pathway and enhanced the anti-inflammatory and anti-apoptotic effects of IPostC. However, IPostC + miR-499 inhibitors had the opposite effect. Therefore, it was speculated that IPostC may have a miR-499-dependent cardioprotective effect. The present results suggested that miR-499 may be involved in IPostC-mediated ischemic cardioprotection, which may occur via local and systemic TLR2 inhibition, subsequent inhibition of the PKC signaling pathway and a decrease in inflammatory cytokine release, including IL-1β and IL-6. Moreover, these effects will ultimately lead to a decrease in the myocardial apoptotic index and myocardial infarct size via the induction of the anti-apoptotic protein Bcl-2, and inhibition of the pro-apoptotic protein Bax in myocardium.
Collapse
Affiliation(s)
- Xin-Yue Zhang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zheng Huang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qing-Jie Li
- Department of Cardiology, Second Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guo-Qiang Zhong
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jian-Jun Meng
- Department of Geriatric Health Care Center, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Dong-Xiao Wang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rong-Hui Tu
- Guangxi Key Laboratory of Precision Medicine in Cardio‑Cerebrovascular Diseases Control and Prevention, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
7
|
Raimann FJ, Dröse S, Bonke E, Schneider L, Tybl E, Wittig I, Heidler J, Heide H, Josipovic I, Leisegang M, Brandes RP, Roeper J, Zacharowski K, Mersmann J. TLR2-Dependent Reversible Oxidation of Connexin 43 at Cys260 Modifies Electrical Coupling After Experimental Myocardial Ischemia/Reperfusion. J Cardiovasc Transl Res 2019; 12:478-487. [PMID: 30963423 DOI: 10.1007/s12265-019-09887-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/27/2019] [Indexed: 12/27/2022]
Abstract
We have shown previously that during myocardial ischemia/reperfusion (MI/R), toll-like receptor 2 (TLR2) signaling regulates connexin 43 (Cx43) subcellular localization and function and dampens arrhythmia formation. We aimed to identify sites capable of TLR2-dependent redox modification within Cx43. Post-ischemic TLR2-/- or wild-type (WT) mouse hearts were analyzed by OxICAT. Cx43 was mutated to exclude redox modification and transfected into HL-1 cardiomyocytes (CM) that were challenged with a TLR2 agonist. We identified Cys260 of Cx43 to be susceptible to reversible oxidation MI/R; TLR2-/- leads to reduced H2O2 production in post-ischemic isolated mitochondria and subsequently reduced oxidation of Cx43 at Cys260. Cx43 was dephosphorylated in WT, while phosphorylation was preserved in TLR2-/-. Mutation of Cx43 (C260A) and lentiviral transfection in HL-1 CM accelerated pacemaker activity and reduced activity after TLR2 ligand stimulation. We here provide evidence for TLR2-dependent reversible oxidation of Cx43 at Cys260, which led to decreased Cx43 phosphorylation and affected CM pacemaker frequency and intercellular communication.
Collapse
MESH Headings
- Action Potentials
- Animals
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/physiopathology
- Cell Communication
- Cell Line
- Connexin 43/deficiency
- Connexin 43/genetics
- Connexin 43/metabolism
- Cysteine
- Disease Models, Animal
- Heart Rate
- Hydrogen Peroxide/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Reperfusion Injury/genetics
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/physiopathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Oxidation-Reduction
- Phosphorylation
- Signal Transduction
- Toll-Like Receptor 2/deficiency
- Toll-Like Receptor 2/genetics
- Toll-Like Receptor 2/metabolism
Collapse
Affiliation(s)
- Florian Jürgen Raimann
- Department of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Stefan Dröse
- Department of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Erik Bonke
- Department of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Lea Schneider
- Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt, Germany
| | - Elisabeth Tybl
- Department of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt, Germany
| | - Juliana Heidler
- Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt, Germany
| | - Heinrich Heide
- Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt, Germany
- Thermo Fisher Scientific GmbH, Dreieich, Germany
| | - Ivana Josipovic
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany
| | - Matthias Leisegang
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany
| | - Ralf Peter Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany
| | - Jochen Roeper
- Institute of Neurophysiology, Goethe University, Frankfurt, Germany
| | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Jan Mersmann
- Department of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
8
|
Murayama S, Kurganov E, Miyata S. Activation of microglia and macrophages in the circumventricular organs of the mouse brain during TLR2-induced fever and sickness responses. J Neuroimmunol 2019; 334:576973. [PMID: 31170673 DOI: 10.1016/j.jneuroim.2019.576973] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/26/2019] [Accepted: 05/27/2019] [Indexed: 01/28/2023]
Abstract
Toll-like receptor 2 (TLR2) recognizes cell wall components from Gram-positive bacteria. Until now, however, little has been known about the significance of brain TLR2 in controlling inflammation and thermoregulatory responses during systemic Gram-positive bacterial infection. In the present study, the TLR2 immunoreactivity was seen to be prominent in the microglia/macrophages of the circumventricular organs (CVOs) of the mouse brain. The intraperitoneal injection of Pam3CSK4, a TLR2 agonist, induced nuclear factor-κ B activation in the microglia/macrophages of the CVOs. The injection of Pam3CSK4 also produced the expression of Fos at astrocytes and neurons in the CVOs and the regions neighboring the CVOs. The Pam3CSK4 injection induced fever and sickness responses. Pretreatment with lipopolysaccharide, a TLR4 agonist, augmented the Pam3CSK4-induced fever together with the increased TLR2 immunoreactivity. These results indicate that the TLR2 in microglia/macrophages of the CVOs are possibly associated with initiating and transmitting inflammatory responses in the brain.
Collapse
Affiliation(s)
- Saki Murayama
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Erkin Kurganov
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
9
|
Quinlan S, Merino-Serrais P, Di Grande A, Dussmann H, Prehn JHM, Ní Chonghaile T, Henshall DC, Jimenez-Mateos EM. The Anti-inflammatory Compound Candesartan Cilexetil Improves Neurological Outcomes in a Mouse Model of Neonatal Hypoxia. Front Immunol 2019; 10:1752. [PMID: 31396238 PMCID: PMC6667988 DOI: 10.3389/fimmu.2019.01752] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/10/2019] [Indexed: 11/15/2022] Open
Abstract
Recent studies suggest that mild hypoxia-induced neonatal seizures can trigger an acute neuroinflammatory response leading to long-lasting changes in brain excitability along with associated cognitive and behavioral deficits. The cellular elements and signaling pathways underlying neuroinflammation in this setting remain incompletely understood but could yield novel therapeutic targets. Here we show that brief global hypoxia-induced neonatal seizures in mice result in transient cytokine production, a selective expansion of microglia and long-lasting changes to the neuronal structure of pyramidal neurons in the hippocampus. Treatment of neonatal mice after hypoxia-seizures with the novel anti-inflammatory compound candesartan cilexetil suppressed acute seizure-damage and mitigated later-life aggravated seizure responses and hippocampus-dependent learning deficits. Together, these findings improve our understanding of the effects of neonatal seizures and identify potentially novel treatments to protect against short and long-lasting harmful effects.
Collapse
Affiliation(s)
- Sean Quinlan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula Merino-Serrais
- Division for Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Departamento de Neurobiologia Funcional y de Sistemas, Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Alessandra Di Grande
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tríona Ní Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David C Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.,INFANT Research Centre, UCC, Cork, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Li Y, Deng SL, Lian ZX, Yu K. Roles of Toll-Like Receptors in Nitroxidative Stress in Mammals. Cells 2019; 8:cells8060576. [PMID: 31212769 PMCID: PMC6627996 DOI: 10.3390/cells8060576] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/20/2023] Open
Abstract
Free radicals are important antimicrobial effectors that cause damage to DNA, membrane lipids, and proteins. Professional phagocytes produce reactive oxygen species (ROS) and reactive nitrogen species (RNS) that contribute towards the destruction of pathogens. Toll-like receptors (TLRs) play a fundamental role in the innate immune response and respond to conserved microbial products and endogenous molecules resulting from cellular damage to elicit an effective defense against invading pathogens, tissue injury, or cancer. In recent years, several studies have focused on how the TLR-mediated activation of innate immune cells leads to the production of pro-inflammatory factors upon pathogen invasion. Here, we review recent findings that indicate that TLRs trigger a signaling cascade that induces the production of reactive oxygen and nitrogen species.
Collapse
Affiliation(s)
- Yao Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Shou-Long Deng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zheng-Xing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
11
|
Wietzorrek G, Drexel M, Trieb M, Santos-Sierra S. Anti-inflammatory activity of small-molecule antagonists of Toll-like receptor 2 (TLR2) in mice. Immunobiology 2019; 224:1-9. [PMID: 30509503 DOI: 10.1016/j.imbio.2018.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022]
Abstract
Toll-like receptor 2 (TLR2) is currently investigated as a potential therapeutic target in diseases with underlying inflammation like sepsis and arthritis. We reported the discovery, by virtual screening and biological testing, of eight TLR2 antagonists (AT1-AT8) which showed TLR2-inhibitory activity in human cells (Murgueitio et al., 2014). In this study, we have deepened in the mechanism of action and selectivity (TLR2/1 or TLR2/6) of those compounds in mouse primary cells and in vivo. The antagonists reduced, in a dose-dependent way the TNFα production (e.g. AT5 IC50 7.4 μM) and also reduced the nitric oxide (NO) formation in mouse bone marrow-derived macrophages (BMDM). Treatment of BMDM with the antagonists showed that downstream of TLR2, MAPKs phosphorylation and IkBα degradation was reduced. Notably, in a mouse model of tri-acylated lipopeptide (Pam3CSK4)-induced inflammation, AT5 attenuated the TNFα and IL-6 inflammatory response. Further, the effect of AT5 in the stimulation of BMDM by the endogenous alarmin HMGB1 was investigated. Our results indicate that AT4-AT7 and, particularly AT5 appear as good starting points for the development of inhibitors targeting TLR2 in inflammatory disorders.
Collapse
Affiliation(s)
- G Wietzorrek
- Section of Molecular and Cellular Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - M Drexel
- Department of Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - M Trieb
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - S Santos-Sierra
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria.
| |
Collapse
|
12
|
Ubc9 overexpression and SUMO1 deficiency blunt inflammation after intestinal ischemia/reperfusion. J Transl Med 2018; 98:799-813. [PMID: 29472640 PMCID: PMC6397426 DOI: 10.1038/s41374-018-0035-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/27/2017] [Accepted: 01/10/2018] [Indexed: 11/08/2022] Open
Abstract
The intestinal epithelium constitutes a crucial defense to the potentially life-threatening effects of gut microbiota. However, due to a complex underlying vasculature, hypoperfusion and resultant tissue ischemia pose a particular risk to function and integrity of the epithelium. The small ubiquitin-like modifier (SUMO) conjugation pathway critically regulates adaptive responses to metabolic stress and is of particular significance in the gut, as inducible knockout of the SUMO-conjugating enzyme Ubc9 results in rapid intestinal epithelial disintegration. Here we analyzed the pattern of individual SUMO isoforms in intestinal epithelium and investigated their roles in intestinal ischemia/reperfusion (I/R) damage. Immunostaining revealed that epithelial SUMO2/3 expression was almost exclusively limited to crypt epithelial nuclei in unchallenged mice. However, intestinal I/R or overexpression of Ubc9 caused a remarkable enhancement of epithelial SUMO2/3 staining along the crypt-villus axis. Unexpectedly, a similar pattern was found in SUMO1 knockout mice. Ubc9 transgenic mice, but also SUMO1 knockout mice were protected from I/R injury as evidenced by better preserved barrier function and blunted inflammatory responses. PCR array analysis of microdissected villus-tip epithelia revealed a specific epithelial contribution to reduced inflammatory responses in Ubc9 transgenic mice, as key chemotactic signaling molecules such as IL17A were significantly downregulated. Together, our data indicate a critical role particularly of the SUMO2/3 isoforms in modulating responses to I/R and provide the first evidence that SUMO1 deletion activates a compensatory process that protects from ischemic damage.
Collapse
|
13
|
Hilbert T, Markowski P, Frede S, Boehm O, Knuefermann P, Baumgarten G, Hoeft A, Klaschik S. Synthetic CpG oligonucleotides induce a genetic profile ameliorating murine myocardial I/R injury. J Cell Mol Med 2018; 22:3397-3407. [PMID: 29671939 PMCID: PMC6010716 DOI: 10.1111/jcmm.13616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
We previously demonstrated that pre‐conditioning with CpG oligonucleotide (ODN) 1668 induces quick up‐regulation of gene expression 3 hours post‐murine myocardial ischaemia/reperfusion (I/R) injury, terminating inflammatory processes that sustain I/R injury. Now, performing comprehensive microarray and biocomputational analyses, we sought to further enlighten the “black box” beyond these first 3 hours. C57BL/6 mice were pretreated with either CpG 1668 or with control ODN 1612, respectively. Sixteen hours later, myocardial ischaemia was induced for 1 hour in a closed‐chest model, followed by reperfusion for 24 hours. RNA was extracted from hearts, and labelled cDNA was hybridized to gene microarrays. Data analysis was performed with BRB ArrayTools and Ingenuity Pathway Analysis. Functional groups mediating restoration of cellular integrity were among the top up‐regulated categories. Genes known to influence cardiomyocyte survival were strongly induced 24 hours post‐I/R. In contrast, proinflammatory pathways were down‐regulated. Interleukin‐10, an upstream regulator, suppressed specifically selected proinflammatory target genes at 24 hours compared to 3 hours post‐I/R. The IL1 complex is supposed to be one regulator of a network increasing cardiovascular angiogenesis. The up‐regulation of numerous protective pathways and the suppression of proinflammatory activity are supposed to be the genetic correlate of the cardioprotective effects of CpG 1668 pre‐conditioning.
Collapse
Affiliation(s)
- Tobias Hilbert
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Paul Markowski
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Stilla Frede
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Olaf Boehm
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Pascal Knuefermann
- Department of Anesthesiology and Intensive Care Medicine, Gemeinschaftskrankenhaus Bonn St. Elisabeth - St. Petrus - St. Johannes gGmbH, Bonn, Germany
| | - Georg Baumgarten
- Department of Anesthesiology and Intensive Care Medicine, Johanniter Hospital Bonn, Bonn, Germany
| | - Andreas Hoeft
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sven Klaschik
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
14
|
Zhang P, Shao L, Ma J. Toll-Like Receptors 2 and 4 Predict New-Onset Atrial Fibrillation in Acute Myocardial Infarction Patients. Int Heart J 2018; 59:64-70. [PMID: 29375116 DOI: 10.1536/ihj.17-084] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Myocardial infarction (MI) can cause new-onset atrial fibrillation (AF) due to cardiac remodeling. As a recent study has shown, inflammatory factors are closely tied to cell death and survival in myocardial ischemia injury. Toll-like receptors (TLRs) have been shown to participate in the process of myocardial infarction as innate immune factors.The subjects were divided into 3 groups: healthy controls (n = 82), MI patients (n = 84), and AFMI (new-onset atrial fibrillation after myocardial infarction) patients (n = 85). Peripheral blood mononuclear cell (PBMC) TLR mRNA expression was detected by rt-PCR. Western blot was used to analyze PBMC TLRs and their downstream signal protein expression. PBMCs were presented as TLR2 expression or TLR4 expression using flow cytometry.From mRNA to protein detection, PBMC TLR2 and TLR4 were significantly higher in the AFMI group than in the control group and MI group. A similar tendency was also observed in the expression of downstream signaling proteins. When further analyzed with TLR2 and TLR4 antibodies by flow cytometry, PBMC levels also appeared to be higher in AFMI patients than those in MI patients and the healthy control group.In our study, PBMC TLRs and their downstream signaling proteins were significantly higher in the acute myocardial infarction patients with new-onset atrial fibrillation compared with healthy people and acute myocardial infarction patients without new-onset atrial fibrillation. They have the potential to be novel biomarkers for new-onset atrial fibrillation after acute myocardial infarction.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Geriatrics & Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
| | - Liang Shao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
| | - Jun Ma
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
| |
Collapse
|
15
|
Deng S, Yu K, Jiang W, Li Y, Wang S, Deng Z, Yao Y, Zhang B, Liu G, Liu Y, Lian Z. Over-expression of Toll-like receptor 2 up-regulates heme oxygenase-1 expression and decreases oxidative injury in dairy goats. J Anim Sci Biotechnol 2017; 8:3. [PMID: 28078083 PMCID: PMC5223356 DOI: 10.1186/s40104-016-0136-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/12/2016] [Indexed: 12/14/2022] Open
Abstract
Background Mastitis, an infection caused by Gram-positive bacteria, produces udder inflammation and oxidative injury in milk-producing mammals. Toll-like receptor 2 (TLR2) is important for host recognition of invading Gram-positive microbes. Over-expression of TLR2 in transgenic dairy goats is a useful model for studying various aspects of infection with Gram-positive bacteria, in vivo. Methods We over-expressed TLR2 in transgenic dairy goats. Pam3CSK4, a component of Gram-positive bacteria, triggered the TLR2 signal pathway by stimulating the monocytes-macrophages from the TLR2-positive transgenic goats, and induced over-expression of activator protein-1 (AP-1), phosphatidylinositol 3-kinase (PI3K) and transcription factor nuclear factor kappa B (NF-κB) and inflammation factors downstream of the signal pathway. Results Compared with wild-type controls, measurements of various oxidative stress-related molecules showed that TLR2, when over-expressed in transgenic goat monocytes-macrophages, resulted in weak lipid damage, high level expression of anti-oxidative stress proteins, and significantly increased mRNA levels of transcription factor NF-E2-related factor-2 (Nrf2) and the downstream gene, heme oxygenase-1 (HO-1). When Pam3CSK4 was used to stimulate ear tissue in vivo the HO-1 protein of the transgenic goats had a relatively high expression level. Conclusions The results indicate that the oxidative injury in goats over-expressing TLR2 was reduced following Pam3CSK4 stimulation. The underlying mechanism for this reduction was increased expression of the anti-oxidation gene HO-1 by activation of the Nrf2 signal pathway.
Collapse
Affiliation(s)
- Shoulong Deng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Kun Yu
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China.,National key Lab of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Wuqi Jiang
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Yan Li
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Shuotian Wang
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Zhuo Deng
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078 USA
| | - Yuchang Yao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030 People's Republic of China
| | - Baolu Zhang
- State Oceanic Administration, Beijing, 100860 People's Republic of China
| | - Guoshi Liu
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Yixun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Zhengxing Lian
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China.,National key Lab of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193 People's Republic of China
| |
Collapse
|
16
|
Goulopoulou S, McCarthy CG, Webb RC. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol Rev 2016; 68:142-67. [PMID: 26721702 PMCID: PMC4709508 DOI: 10.1124/pr.114.010090] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptors (TLRs) are components of the innate immune system that respond to exogenous infectious ligands (pathogen-associated molecular patterns, PAMPs) and endogenous molecules that are released during host tissue injury/death (damage-associated molecular patterns, DAMPs). Interaction of TLRs with their ligands leads to activation of downstream signaling pathways that induce an immune response by producing inflammatory cytokines, type I interferons (IFN), and other inflammatory mediators. TLR activation affects vascular function and remodeling, and these molecular events prime antigen-specific adaptive immune responses. Despite the presence of TLRs in vascular cells, the exact mechanisms whereby TLR signaling affects the function of vascular tissues are largely unknown. Cardiovascular diseases are considered chronic inflammatory conditions, and accumulating data show that TLRs and the innate immune system play a determinant role in the initiation and development of cardiovascular diseases. This evidence unfolds a possibility that targeting TLRs and the innate immune system may be a novel therapeutic goal for these conditions. TLR inhibitors and agonists are already in clinical trials for inflammatory conditions such as asthma, cancer, and autoimmune diseases, but their study in the context of cardiovascular diseases is in its infancy. In this article, we review the current knowledge of TLR signaling in the cardiovascular system with an emphasis on atherosclerosis, hypertension, and cerebrovascular injury. Furthermore, we address the therapeutic potential of TLR as pharmacological targets in cardiovascular disease and consider intriguing research questions for future study.
Collapse
Affiliation(s)
- Styliani Goulopoulou
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - R Clinton Webb
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
17
|
Stallmann-Jorgensen I, Ogbi S, Szasz T, Webb RC. A Toll-Like Receptor 1/2 Agonist Augments Contractility in Rat Corpus Cavernosum. J Sex Med 2015; 12:1722-31. [PMID: 26234560 DOI: 10.1111/jsm.12960] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Activation of the innate immune Toll-like receptor 2 (TLR2) initiates inflammation and has been implicated in vascular dysfunction. Increased contraction and decreased relaxation responses in the penile vasculature lead to erectile dysfunction, a condition associated with inflammation. However, whether TLR2 activation plays a role in penile vascular function has not been established. AIM We hypothesized that activation of the TLR 1/2 heterodimer (TLR1/2) augments contractile and impairs relaxation responses of corpus cavernosum and that these perturbations of vascular function are mediated by low nitric oxide (NO) availability and enhanced activity of the RhoA/Rho-kinase pathway. METHODS Contraction and relaxation responses were measured in rat cavernosal strips using a myograph after incubation with a TLR1/2-activating ligand Pam3 CSK4 (Pam3), the TLR1/2 inhibitor CuCPT 22 (CuCPT), and inhibitors of NO synthase (LNAME) and Rho-kinase (Y27632). TLR2 protein expression was assessed by immunohistochemistry. MAIN OUTCOME MEASURES Cumulative concentration response curves, sensitivity (pEC50), and maximal response (Emax ) of cavernosal strips to vasodilatory and vasocontractile agonists were compared between treatments. RESULTS Pam3-treated cavernosal strips exhibited greater pEC50 and higher Emax to phenylephrine (PE) than control tissues. Inhibition of NO synthase increased Emax to PE in Pam3-treated cavernosal strips. Pam3 treatment reduced relaxation to Y27632 compared with control tissues. Inhibition of TLR1/2 activation with CuCPT returned the augmented contraction to PE and the decreased relaxation to Y27632 of Pam3-treated cavernosal strips to control values. CONCLUSIONS The TLR1/2 heterodimer mediates augmented contraction and reduced relaxation in rat cavernosal strips. Thus, TLR1/2 activation antagonizes vascular responses crucial for normal erectile function and implicates immune activation in vasculogenic erectile dysfunction. Immune signaling via TLR2 may offer novel targets for treating inflammation-mediated vascular dysfunction in the penis.
Collapse
Affiliation(s)
| | - Safia Ogbi
- Department of Physiology, Georgia Regents University, Augusta, GA, USA
| | - Theodora Szasz
- Department of Physiology, Georgia Regents University, Augusta, GA, USA
| | - R Clinton Webb
- Department of Physiology, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
18
|
Cross talk of the first-line defense TLRs with PI3K/Akt pathway, in preconditioning therapeutic approach. MOLECULAR AND CELLULAR THERAPIES 2015; 3:4. [PMID: 26056605 PMCID: PMC4456045 DOI: 10.1186/s40591-015-0041-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 05/19/2015] [Indexed: 01/04/2023]
Abstract
Toll-like receptor family (TLRs), pattern recognition receptors, is expressed not only on immune cells but also on non-immune cells, including cardiomyocytes, fibroblasts, and vascular endothelial cells. One main function of TLRs in the non-immune system is to regulate apoptosis. TLRs are the central mediators in hepatic, pulmonary, brain, and renal ischemic/reperfusion (I/R) injury. Up-regulation of TLRs and their ligation by either exogenous or endogenous danger signals plays critical roles in ischemia/reperfusion-induced tissue damage. Conventional TLR-NF-κB pathways are markedly activated in failing and ischemic myocardium. Recent studies have identified a cross talk between TLR activation and the PI3K/Akt pathway. The activation of TLRs is proposed to be the most potent preconditioning method after ischemia, to improve the cell survival via the mechanism involved the PI3K/Akt signaling pathway and to attenuate the subsequent TLR-NF-κB pathway stimulation. Thus, TLRs could be a great target in the new treatment approaches for myocardial I/R injury.
Collapse
|
19
|
Pourrajab F, Yazdi MB, Zarch MB, Zarch MB, Hekmatimoghaddam S. Cross talk of the first-line defense TLRs with PI3K/Akt pathway, in preconditioning therapeutic approach. MOLECULAR AND CELLULAR THERAPIES 2015; 3:4. [PMID: 26056605 PMCID: PMC4456045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 05/19/2015] [Indexed: 11/21/2023]
Abstract
Toll-like receptor family (TLRs), pattern recognition receptors, is expressed not only on immune cells but also on non-immune cells, including cardiomyocytes, fibroblasts, and vascular endothelial cells. One main function of TLRs in the non-immune system is to regulate apoptosis. TLRs are the central mediators in hepatic, pulmonary, brain, and renal ischemic/reperfusion (I/R) injury. Up-regulation of TLRs and their ligation by either exogenous or endogenous danger signals plays critical roles in ischemia/reperfusion-induced tissue damage. Conventional TLR-NF-κB pathways are markedly activated in failing and ischemic myocardium. Recent studies have identified a cross talk between TLR activation and the PI3K/Akt pathway. The activation of TLRs is proposed to be the most potent preconditioning method after ischemia, to improve the cell survival via the mechanism involved the PI3K/Akt signaling pathway and to attenuate the subsequent TLR-NF-κB pathway stimulation. Thus, TLRs could be a great target in the new treatment approaches for myocardial I/R injury.
Collapse
Affiliation(s)
- Fatemeh Pourrajab
- />School of Medicine, Shahid Sadoughi University of Medical Sciences, Professor Hessabi 11 BLV, Shohadaye Gomnam BLV, Yazd, Iran P.O. 8915173149
- />Department of Clinical Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Baghi Yazdi
- />School of Medicine, Shahid Sadoughi University of Medical Sciences, Professor Hessabi 11 BLV, Shohadaye Gomnam BLV, Yazd, Iran P.O. 8915173149
| | - Mojtaba Babaei Zarch
- />School of Medicine, Shahid Sadoughi University of Medical Sciences, Professor Hessabi 11 BLV, Shohadaye Gomnam BLV, Yazd, Iran P.O. 8915173149
| | - Mohammadali Babaei Zarch
- />School of Medicine, Shahid Sadoughi University of Medical Sciences, Professor Hessabi 11 BLV, Shohadaye Gomnam BLV, Yazd, Iran P.O. 8915173149
| | | |
Collapse
|
20
|
Vilahur G, Badimon L. Ischemia/reperfusion activates myocardial innate immune response: the key role of the toll-like receptor. Front Physiol 2014; 5:496. [PMID: 25566092 PMCID: PMC4270170 DOI: 10.3389/fphys.2014.00496] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/02/2014] [Indexed: 01/04/2023] Open
Abstract
Recent data have indicated that the myocardium may act as an immune organ initiating cardiac innate immune response and inflammation. It has been suggested that activation of the immune system occurs upon the interaction of damage-associated molecular patterns (DAMPs) generated and released during ischemic damage with pattern recognition receptors (Toll like receptors; TLR) present in cardiac cells. Among TLRs, TLR4, and TLR2 are the ones mostly expressed in cardiac tissue. Whereas TLR4 has shown to play a detrimental role in myocardial ischemia/reperfusion (I/R) injury, the effect elicited by TLR2 activation remains controversial. Once activated, TLR signaling may occur via the Myd88- and Trif- dependent pathways leading to NFκB and IFN-3 activation, respectively, and subsequent stimulation of pro-inflammatory and immunomodulatory cytokine gene expression. Cytokine release contributes to neutrophils activation, recruitment, adhesion and infiltration to the site of cardiac injury further perpetuating the inflammatory process. This mini-review will focus on the current knowledge regarding the role of the heart in inducing and coordinating the innate inflammatory response via the TLR signaling pathway in myocardial I/R injury.
Collapse
Affiliation(s)
- Gemma Vilahur
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau Barcelona, Spain ; Cardiovascular Research Chair, Universitat Autònoma de Barcelona Barcelona, Spain
| |
Collapse
|
21
|
Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Role of toll-like receptors in myocardial infarction. J Recept Signal Transduct Res 2014; 35:420-2. [PMID: 25515816 DOI: 10.3109/10799893.2014.993649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Toll-like receptors (TLRs) play a pivotal role in both innate and adaptive immunity, and TLRs recognize invading pathogens through molecular pattern recognition, and ultimately lead to the activation of transcription factors and inflammatory responses. Myocardial infarction leads to changes in the remodeling of the left ventricle of the heart, and the degree and type of remodeling provides important diagnostic information for the therapeutic management of ischemic heart disease. Innate immune takes a most important role in myocardial infarction. There are some studies reporting that TLRs play an important role in the myocardial infarction. The literatures were searched extensively and this review was performed to review the role of TLRs in myocardial infarction.
Collapse
Affiliation(s)
- Yu Sun
- a Department of Cardio-Thoracic Surgery , The First Affiliated Hospital of GuangXi Medical University , NanNing , China
| | - Wen-Zhou Liu
- a Department of Cardio-Thoracic Surgery , The First Affiliated Hospital of GuangXi Medical University , NanNing , China
| | - Tao Liu
- a Department of Cardio-Thoracic Surgery , The First Affiliated Hospital of GuangXi Medical University , NanNing , China
| | - Xu Feng
- a Department of Cardio-Thoracic Surgery , The First Affiliated Hospital of GuangXi Medical University , NanNing , China
| | - Nuo Yang
- a Department of Cardio-Thoracic Surgery , The First Affiliated Hospital of GuangXi Medical University , NanNing , China
| | - Hua-Fu Zhou
- a Department of Cardio-Thoracic Surgery , The First Affiliated Hospital of GuangXi Medical University , NanNing , China
| |
Collapse
|
22
|
Srijaya TC, Ramasamy TS, Kasim NHA. Advancing stem cell therapy from bench to bedside: lessons from drug therapies. J Transl Med 2014; 12:243. [PMID: 25182194 PMCID: PMC4163166 DOI: 10.1186/s12967-014-0243-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022] Open
Abstract
The inadequacy of existing therapeutic tools together with the paucity of organ donors have always led medical researchers to innovate the current treatment methods or to discover new ways to cure disease. Emergence of cell-based therapies has provided a new framework through which it has given the human world a new hope. Though relatively a new concept, the pace of advancement clearly reveals the significant role that stem cells will ultimately play in the near future. However, there are numerous uncertainties that are prevailing against the present setting of clinical trials related to stem cells: like the best route of cell administration, appropriate dosage, duration and several other applications. A better knowledge of these factors can substantially improve the effectiveness of disease cure or organ repair using this latest therapeutic tool. From a certain perspective, it could be argued that by considering certain proven clinical concepts and experience from synthetic drug system, we could improve the overall efficacy of cell-based therapies. In the past, studies on synthetic drug therapies and their clinical trials have shown that all the aforementioned factors have critical ascendancy over its therapeutic outcomes. Therefore, based on the knowledge gained from synthetic drug delivery systems, we hypothesize that by employing many of the clinical approaches from synthetic drug therapies to this new regenerative therapeutic tool, the efficacy of stem cell-based therapies can also be improved.
Collapse
Affiliation(s)
| | - Thamil Selvee Ramasamy
- />Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- />Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Ohm IK, Gao E, Belland Olsen M, Alfsnes K, Bliksøen M, Øgaard J, Ranheim T, Nymo SH, Holmen YD, Aukrust P, Yndestad A, Vinge LE. Toll-like receptor 9-activation during onset of myocardial ischemia does not influence infarct extension. PLoS One 2014; 9:e104407. [PMID: 25126943 PMCID: PMC4134200 DOI: 10.1371/journal.pone.0104407] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 07/14/2014] [Indexed: 02/01/2023] Open
Abstract
Aim Myocardial infarction (MI) remains a major cause of death and disability worldwide, despite available reperfusion therapies. Inflammatory signaling is considered nodal in defining final infarct size. Activation of the innate immune receptor toll-like receptors (TLR) 9 prior to ischemia and reperfusion (I/R) reduces infarct size, but the consequence of TLR9 activation timed to the onset of ischemia is not known. Methods and Results The TLR9-agonist; CpG B was injected i.p. in C57BL/6 mice immediately after induction of ischemia (30 minutes). Final infarct size, as well as area-at-risk, was measured after 24 hours of reperfusion. CpG B injection resulted in a significant increase in circulating granulocytes and monocytes both in sham and I/R mice. Paradoxically, clear evidence of reduced cardiac infiltration of both monocytes and granulocytes could be demonstrated in I/R mice treated with CpG B (immunocytochemistry, myeloperoxidase activity and mRNA expression patterns). In addition, systemic TLR9 activation elicited significant alterations of cardiac inflammatory genes. Despite these biochemical and cellular changes, there was no difference in infarct size between vehicle and CpG B treated I/R mice. Conclusion Systemic TLR9-stimulation upon onset of ischemia and subsequent reperfusion does not alter final infarct size despite causing clear alterations of both systemic and cardiac inflammatory parameters. Our results question the clinical usefulness of TLR9 activation during cardiac I/R.
Collapse
Affiliation(s)
- Ingrid Kristine Ohm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- * E-mail:
| | - Erhe Gao
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Katrine Alfsnes
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marte Bliksøen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Jonas Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ståle Haugset Nymo
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Yangchen Dhondup Holmen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Leif Erik Vinge
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Liu Y, Li G, Lu H, Li W, Li X, Liu H, Li X, Li T, Yu B. Expression profiling and ontology analysis of long noncoding RNAs in post-ischemic heart and their implied roles in ischemia/reperfusion injury. Gene 2014; 543:15-21. [PMID: 24726549 DOI: 10.1016/j.gene.2014.04.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 03/22/2014] [Accepted: 04/07/2014] [Indexed: 02/03/2023]
Abstract
Long noncoding RNAs (lncRNAs) play important regulatory roles in cellular physiology. The contributions of lncRNAs to ischemic heart disease remain largely unknown. The aim of this study was to investigate the profile of myocardial lncRNAs and their potential roles at early stage of reperfusion. lncRNAs and mRNAs were profiled by microarray and the expression of some highly-dysregulated lncRNAs was further validated using polymerase chain reaction. Our results revealed that 64 lncRNAs were up-regulated and 87 down-regulated, while 50 mRNAs were up-regulated and 60 down-regulated in infarct region at all reperfusion sampled. Gene ontology analysis indicated that dysregulated transcripts were associated with immune response, spermine catabolic process, taxis, chemotaxis, polyamine catabolic process, spermine metabolic process, chemokine activity and chemokine receptor binding. Target gene-related pathway analysis showed significant changes in cytokine-cytokine receptor interaction, the chemokine signaling pathway and nucleotide oligomerization domain (NOD)-like receptor signaling pathway which have a close relationship with myocardial ischemia/reperfusion injury (MI/RI). Besides, a gene co-expression network was constructed to identify correlated targets of 10 highly-dysregulated lncRNAs. These lncRNAs may play their roles by this network in post-ischemic heart. Such results provide a foundation for understanding the roles and mechanisms of myocardial lncRNAs at early stage of reperfusion.
Collapse
Affiliation(s)
- Youbin Liu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Heilongjiang Province, China; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Guangnan Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Huimin Lu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Wei Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Xianglu Li
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Heilongjiang Province, China; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Huimin Liu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Heilongjiang Province, China; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Xingda Li
- Department of Pharmacology, Harbin Medical University, Harbin, PR China
| | - Tianyu Li
- Department of Pharmacology, Harbin Medical University, Harbin, PR China
| | - Bo Yu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Heilongjiang Province, China; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| |
Collapse
|
25
|
Nguyen NT, Zhang X, Wu C, Lange RA, Chilton RJ, Lindsey ML, Jin YF. Integrative computational and experimental approaches to establish a post-myocardial infarction knowledge map. PLoS Comput Biol 2014; 10:e1003472. [PMID: 24651374 PMCID: PMC3961365 DOI: 10.1371/journal.pcbi.1003472] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 01/02/2014] [Indexed: 01/04/2023] Open
Abstract
Vast research efforts have been devoted to providing clinical diagnostic markers of myocardial infarction (MI), leading to over one million abstracts associated with “MI” and “Cardiovascular Diseases” in PubMed. Accumulation of the research results imposed a challenge to integrate and interpret these results. To address this problem and better understand how the left ventricle (LV) remodels post-MI at both the molecular and cellular levels, we propose here an integrative framework that couples computational methods and experimental data. We selected an initial set of MI-related proteins from published human studies and constructed an MI-specific protein-protein-interaction network (MIPIN). Structural and functional analysis of the MIPIN showed that the post-MI LV exhibited increased representation of proteins involved in transcriptional activity, inflammatory response, and extracellular matrix (ECM) remodeling. Known plasma or serum expression changes of the MIPIN proteins in patients with MI were acquired by data mining of the PubMed and UniProt knowledgebase, and served as a training set to predict unlabeled MIPIN protein changes post-MI. The predictions were validated with published results in PubMed, suggesting prognosticative capability of the MIPIN. Further, we established the first knowledge map related to the post-MI response, providing a major step towards enhancing our understanding of molecular interactions specific to MI and linking the molecular interaction, cellular responses, and biological processes to quantify LV remodeling. Heart attack, known medically as myocardial infarction, often occurs as a result of partial shortage of blood supply to a portion of the heart, leading to the death of heart muscle cells. Following myocardial infarction, complications might arise, including arrhythmia, myocardial rupture, left ventricular dysfunction, and heart failure. Although myocardial infarction can be quickly diagnosed using a various number of tests, including blood tests and electrocardiography, there have been no available prognostic tests to predict the long-term outcome in response to myocardial infarction. Here, we present a framework to analyze how the left ventricle responds to myocardial infarction by combining protein interactome and experimental results retrieved from published human studies. The framework organized current understanding of molecular interactions specific to myocardial infarction, cellular responses, and biological processes to quantify left ventricular remodeling process. Specifically, our knowledge map showed that transcriptional activity, inflammatory response, and extracellular matrix remodeling are the main functional themes post myocardial infarction. In addition, text analytics of relevant abstracts revealed differentiated protein expressions in plasma or serum expressions from patients with myocardial infarction. Using this data, we predicted expression levels of other proteins following myocardial infarction.
Collapse
Affiliation(s)
- Nguyen T. Nguyen
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
- San Antonio Cardiovascular Proteomics Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Xiaolin Zhang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Cathy Wu
- Center for Bioinformatics and Computational Biology and Protein Information Resource, University of Delaware, Newark, Delaware, United States of America
| | - Richard A. Lange
- San Antonio Cardiovascular Proteomics Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Robert J. Chilton
- San Antonio Cardiovascular Proteomics Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Merry L. Lindsey
- San Antonio Cardiovascular Proteomics Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi, United States of America
| | - Yu-Fang Jin
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
- San Antonio Cardiovascular Proteomics Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
26
|
Okun E, Griffioen KJ, Sarah R, Wan R, Cong WN, De Cabo R, Montalvo AM, Levette A, Maudsley S, Martin B, Arumugam TV, Mattson MP. Toll-like receptors 2 and 4 modulate autonomic control of heart rate and energy metabolism. Brain Behav Immun 2014; 36:90-100. [PMID: 24145051 PMCID: PMC3947180 DOI: 10.1016/j.bbi.2013.10.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 09/24/2013] [Accepted: 10/12/2013] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptors (TLR) are innate immune receptors typically activated by microbial-associated molecular patterns (MAMPs) during infection or damage-associated molecular patterns (DAMPs) as a result of tissue injury. Recent findings suggest that TLR2 and TLR4 signaling play important roles in developmental and adult neuroplasticity, and in learning and memory. In addition, activation of TLR2 and TLR4 worsens ischemic injury to the heart and brain in animal models of myocardial infarction and stroke. TLR activation is also implicated in thermoregulation and fever in response to infection. However, it is not known whether TLRs participate in the regulation of the sympathetic and/or parasympathetic components of the autonomic nervous system (ANS). Here we provide evidence that TLR2 and TLR4 influence autonomic regulation of heart rate (HR) body temperature and energy metabolism in mice. We show that mice lacking TLR2 or TLR4 exhibit reduced basal HR, which results from an increase of parasympathetic tone. In addition, thermoregulatory responses to stress are altered in TLR2-/- and TLR4-/- mice, and brown fat-dependent thermoregulation is altered in TLR4-/- mice. Moreover, TLR2-/- and TLR4-/- mice consume less food and exhibit a greater mass compared to wild type mice. Collectively, our findings suggest important roles for TLR2 and TLR4 in the ANS regulation of cardiovascular function, thermoregulation, and energy metabolism.
Collapse
Affiliation(s)
- Eitan Okun
- The Mina and Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 52900, Israel.
| | - Kathleen J. Griffioen
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Rothman Sarah
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Ruiqian Wan
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Wei-Na Cong
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Rafael De Cabo
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Alejandro Martin Montalvo
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Andrew Levette
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Stuart Maudsley
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Bronwen Martin
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | | | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
27
|
Attenuation of myocardial injury by HMGB1 blockade during ischemia/reperfusion is toll-like receptor 2-dependent. Mediators Inflamm 2013; 2013:174168. [PMID: 24371373 PMCID: PMC3859028 DOI: 10.1155/2013/174168] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 11/17/2022] Open
Abstract
Genetic or pharmacological ablation of toll-like receptor 2 (TLR2) protects against myocardial ischemia/reperfusion injury (MI/R). However, the endogenous ligand responsible for TLR2 activation has not yet been detected. The objective of this study was to identify HMGB1 as an activator of TLR2 signalling during MI/R. C57BL/6 wild-type (WT) or TLR2(-/-)-mice were injected with vehicle, HMGB1, or HMGB1 BoxA one hour before myocardial ischemia (30 min) and reperfusion (24 hrs). Infarct size, cardiac troponin T, leukocyte infiltration, HMGB1 release, TLR4-, TLR9-, and RAGE-expression were quantified. HMGB1 plasma levels were measured in patients undergoing coronary artery bypass graft (CABG) surgery. HMGB1 antagonist BoxA reduced cardiomyocyte necrosis during MI/R in WT mice, accompanied by reduced leukocyte infiltration. Injection of HMGB1 did, however, not increase infarct size in WT animals. In TLR2(-/-)-hearts, neither BoxA nor HMGB1 affected infarct size. No differences in RAGE and TLR9 expression could be detected, while TLR2(-/-)-mice display increased TLR4 and HMGB1 expression. Plasma levels of HMGB1 were increased MI/R in TLR2(-/-)-mice after CABG surgery in patients carrying a TLR2 polymorphism (Arg753Gln). We here provide evidence that absence of TLR2 signalling abrogates infarct-sparing effects of HMGB1 blockade.
Collapse
|
28
|
TLR-3 receptor activation protects the very immature brain from ischemic injury. J Neuroinflammation 2013; 10:104. [PMID: 23965176 PMCID: PMC3765441 DOI: 10.1186/1742-2094-10-104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/06/2013] [Indexed: 12/22/2022] Open
Abstract
Background We have shown that preconditioning by lipopolysaccharide (LPS) will result in 90% reduction in ischemic brain damage in P7 rats. This robust LPS neuroprotection was not observed in P3 or P5 pups (corresponding to human premature infant). LPS is a known Toll-like receptor 4 (TLR-4) ligand. We hypothesized that TLRs other than TLR-4 may mediate preconditioning against cerebral ischemic injury in the developing brain. Methods TLR-2, TLR-3, TLR-4, and TLR-9 expression was detected in brain sections from P3, P5, and P7 rats by immuno-staining. In subsequent experiments, P5 rats were randomly assigned to TLR-3 specific agonist, poly I:C, or saline treated group. At 48 h after the injections, hypoxic-ischemic (HI) injury was induced by unilateral carotid artery ligation followed by hypoxia for 65 min. Brains were removed 1 week after HI injury and infarct volumes were compared in H&E stained sections between the two groups. Results TLR-2 and TLR-3 were highly expressed in brains of P3 and P5 but not in P7 rats. The number of TLR-4 positive cells was lower in P3 and P5 compared to P7 brains (P <0.05). TLR-3 was predominately expressed in P5 pups (P <0.05). There was no significant difference in TLR-9 expression in the three age groups. There was a significant reduction in infarct volume (P = 0.01) in poly I:C compared to saline pre-treated P5 pups. Pre-treatment with poly I:C downregulated NF-κB and upregulated IRF3 expression in P5 rat ischemic brains. Pre-treatment with poly I:C did not offer neuroprotection in P7 rat brains. Conclusion TLRs expression and function is developmentally determined. Poly I:C-induced preconditioning against ischemic injury may be mediated by modulation of TLR-3 signaling pathways. This is the first study to show that TLR-3 is expressed in the immature brain and mediates preconditioning against ischemic injury.
Collapse
|
29
|
Markowski P, Boehm O, Goelz L, Haesner AL, Ehrentraut H, Bauerfeld K, Tran N, Zacharowski K, Weisheit C, Langhoff P, Schwederski M, Hilbert T, Klaschik S, Hoeft A, Baumgarten G, Meyer R, Knuefermann P. Pre-conditioning with synthetic CpG-oligonucleotides attenuates myocardial ischemia/reperfusion injury via IL-10 up-regulation. Basic Res Cardiol 2013; 108:376. [PMID: 23929312 PMCID: PMC3778842 DOI: 10.1007/s00395-013-0376-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 07/18/2013] [Accepted: 07/26/2013] [Indexed: 01/26/2023]
Abstract
The aim of the study was to investigate whether pre-conditioning with CpG-oligodeoxynucleotides (CpG-ODN) may change cardiac ischemia/reperfusion (I/R)-dependent inflammation and modulates infarct size and cardiac performance. WT and TLR9-deficient mice were pre-treated with 1668-, 1612- and H154-thioate or D-Gal as control. Priming with 1668-thioate significantly induced inflammatory mediators in the serum and a concomitant increase of immune cells in the blood and spleen of WT mice. Furthermore, it induced myocardial pattern recognition receptors and pro-inflammatory cytokines peaking 2 h after priming and a continuous increase of IL-10. 16 h after pre-conditioning, myocardial ischemia was induced for 1 h. Infarct size determined after 24 h of I/R was reduced by 75 % due to pre-conditioning with 1668-thioate but not in the other groups. During reperfusion, cytokine expression in 1668-thioate primed mice increased further with IL-10 exceeding the other mediators by far. These changes were observed neither in animals pre-treated with 1612- or H154-thioate nor in TLR9-deficient mice. The 1668-thioate-dependent increase of IL-10 was further supported by results of a micro-array analysis 3 h after begin of reperfusion. Block of IL-10 signaling increased I/R size and prevented influence of priming. In the group pre-treated with 1668-thioate, cardiac function was preserved 24 h, 14 days and 28 days after I/R, whereas animals without pre-conditioning exhibited impaired heart function 24 h and 14 days after I/R. The excessive 1668-thioate-dependent IL-10 up-regulation during pre-conditioning and after I/R seems to be the key factor for reducing infarct size and improving cardiac function. This is in agreement with the finding that IL-10 block prevents cardioprotection by pre-conditioning.
Collapse
Affiliation(s)
- P Markowski
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Grote K, Sonnenschein K, Kapopara PR, Hillmer A, Grothusen C, Salguero G, Kotlarz D, Schuett H, Bavendiek U, Schieffer B. Toll-like receptor 2/6 agonist macrophage-activating lipopeptide-2 promotes reendothelialization and inhibits neointima formation after vascular injury. Arterioscler Thromb Vasc Biol 2013; 33:2097-104. [PMID: 23868938 DOI: 10.1161/atvbaha.113.301799] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Reendothelialization after vascular injury (ie, balloon angioplasty or stent implantation) is clinically extremely relevant to promote vascular healing. We here investigated the therapeutic potential of the toll-like receptor 2/6 agonist macrophage-activating lipopeptide (MALP)-2 on reendothelialization and neointima formation in a murine model of vascular injury. APPROACH AND RESULTS The left common carotid artery was electrically injured, and reendothelialization was quantified by Evans blue staining after 3 days. A single injection of MALP-2 (1 or 10 µg, IV) after vascular injury accelerated reendothelialization (P<0.001). Proliferation of endothelial cells at the wound margins determined by 5-ethynyl-2'-deoxyuridine incorporation was significantly higher in MALP-2-treated animals (P<0.05). Furthermore, wire injury-induced neointima formation of the left common carotid artery was completely prevented by a single injection of MALP-2 (10 µg, IV). In vitro, MALP-2 induced proliferation (BrdU incorporation) and closure of an artificial wound of endothelial cells (P<0.05) but not of smooth muscle cells. Protein array and ELISA analysis of isolated primary endothelial cells and ex vivo stimulated carotid segments revealed that MALP-2 stimulated the release of multiple growth factors and cytokines predominantly from endothelial cells. MALP-2 induced a strong activation of the mitogen-activated protein kinase cascade in endothelial cells, which was attenuated in smooth muscle cells. Furthermore, MALP-2 significantly enhanced circulating monocytes and hematopoietic progenitor cells. CONCLUSIONS The toll-like receptor 2/6 agonist MALP-2 promotes reendothelialization and inhibits neointima formation after experimental vascular injury via enhanced proliferation and migration of endothelial cells. Thus, MALP-2 represents a novel therapeutic option to accelerate reendothelialization after vascular injury.
Collapse
Affiliation(s)
- Karsten Grote
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Xu Y, Zhou Y, Lin H, Hu H, Wang Y, Xu G. Toll-like receptor 2 in promoting angiogenesis after acute ischemic injury. Int J Mol Med 2013; 31:555-60. [PMID: 23314218 DOI: 10.3892/ijmm.2013.1240] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/06/2012] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is an important mechanism that protects tissue against necrosis following acute ischemic injury. The aim of this study was to investigate whether the Toll-like receptor 2 (TLR2) signaling pathway is involved in angiogenesis following ischemic injury by cell migration and lymphocyte invasion assays in vitro, and a mouse model of hindlimb ischemia by ligation in vivo, respectively. To assess the potential role of TLR2 activation in endothelial cell permeability, HUVECs were pretreated with Pam3CSK4 and analyzed using wound repair and transwell assays. The results showed that the TLR2 agonist induced human umbilical vein endothelial cell (HUVEC) migration and increased the permeability of HUVECs to lymphocyte. The lymphocyte invasion of TLR2 knockout (TLR2-/-) mice was inhibited as compared to that of wild-type (WT) mice. In the mouse model of hindlimb ischemia by ligation, blood perfusion of operated limbs was significantly lower in TLR2-/- compared to WT mice, 7 and 14 days after ligation. TLR2-/- mice showed a decreased CD31 expression in ischemic gastrocnemius at 7 and 14 days after ligation, reduced interleukin-6 (IL-6) level and lowered tumor necrosis factor-α (TNF-α) levels. These findings demonstrated that TLR2 activation promotes cell migration, cell permeability and the lymphocyte invasion of endothelial cells. TLR2 activation promotes angiogenesis in vivo, which may be associated with the serum of TNF-α levels and IL-6 release.
Collapse
Affiliation(s)
- Yifei Xu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, PR China
| | | | | | | | | | | |
Collapse
|
32
|
Koch A, Pernow M, Barthuber C, Mersmann J, Zacharowski K, Grotemeyer D. Systemic inflammation after aortic cross clamping is influenced by Toll-like receptor 2 preconditioning and deficiency. J Surg Res 2012; 178:833-41. [DOI: 10.1016/j.jss.2012.04.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/17/2012] [Accepted: 04/24/2012] [Indexed: 01/04/2023]
|
33
|
Akt or phosphoinositide-3-kinase inhibition reverses cardio-protection in Toll-like receptor 2 deficient mice. Resuscitation 2012; 83:1404-10. [DOI: 10.1016/j.resuscitation.2012.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 03/27/2012] [Accepted: 04/03/2012] [Indexed: 01/06/2023]
|
34
|
Therapeutic role of toll-like receptor modification in cardiovascular dysfunction. Vascul Pharmacol 2012; 58:231-9. [PMID: 23070056 DOI: 10.1016/j.vph.2012.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 09/28/2012] [Accepted: 10/05/2012] [Indexed: 01/04/2023]
Abstract
Toll-like receptors (TLR) are key pattern recognition receptors in the innate immune system. The TLR-mediated immune response against pathogens is usually protective however inappropriate TLR activation may lead to excessive tissue damage. It is well recognised that TLRs respond to a variety of endogenous as well as exogenous ligands. By responding to endogenous ligands that are exposed during cellular damage, TLRs have been implicated in a range of pathological conditions associated with cardiovascular dysfunction. Increasing knowledge on the mechanisms involved in TLR signalling has encouraged the exploration of therapeutic pharmacological modulation of TLR activation in conditions such as atherosclerosis, ischaemic heart disease, heart failure and ischaemic reperfusion injury. The aim of this review is to explore the translational potentials of TLR modification in cardiovascular dysfunction, where these agents have been studied.
Collapse
|
35
|
Cao Z, Ren D, Ha T, Liu L, Wang X, Kalbfleisch J, Gao X, Kao R, Williams D, Li C. CpG-ODN, the TLR9 agonist, attenuates myocardial ischemia/reperfusion injury: involving activation of PI3K/Akt signaling. Biochim Biophys Acta Mol Basis Dis 2012; 1832:96-104. [PMID: 22917564 DOI: 10.1016/j.bbadis.2012.08.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/31/2012] [Accepted: 08/08/2012] [Indexed: 01/04/2023]
Abstract
BACKGROUND Toll-like receptors (TLRs) have been implicated in myocardial ischemia/reperfusion (I/R) injury. The TLR9 ligand, CpG-ODN has been reported to improve cell survival. We examined effect of CpG-ODN on myocardial I/R injury. METHODS Male C57BL/6 mice were treated with either CpG-ODN, control-ODN, or inhibitory CpG-ODN (iCpG-ODN) 1h prior to myocardial ischemia (60min) followed by reperfusion. Untreated mice served as I/R control (n=10/each group). Infarct size was determined by TTC straining. Cardiac function was examined by echocardiography before and after myocardial I/R up to 14days. RESULTS CpG-ODN administration significantly decreased infarct size by 31.4% and improved cardiac function after myocardial I/R up to 14days. Neither control-ODN nor iCpG-ODN altered I/R-induced myocardial infarction and cardiac dysfunction. CpG-ODN attenuated I/R-induced myocardial apoptosis and prevented I/R-induced decrease in Bcl2 and increase in Bax levels in the myocardium. CpG-ODN increased Akt and GSK-3β phosphorylation in the myocardium. In vitro data suggested that CpG-ODN treatment induced TLR9 tyrosine phosphorylation and promoted an association between TLR9 and the p85 subunit of PI3K. Importantly, PI3K/Akt inhibition and Akt kinase deficiency abolished CpG-ODN-induced cardioprotection. CONCLUSION CpG-ODN, the TLR9 ligand, induces protection against myocardial I/R injury. The mechanisms involve activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zhijuan Cao
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ha T, Liu L, Kelley J, Kao R, Williams D, Li C. Toll-like receptors: new players in myocardial ischemia/reperfusion injury. Antioxid Redox Signal 2011; 15:1875-93. [PMID: 21091074 PMCID: PMC3159106 DOI: 10.1089/ars.2010.3723] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Innate immune and inflammatory responses have been implicated in myocardial ischemia/reperfusion (I/R) injury. However, the mechanisms by which innate immunity and inflammatory response are involved in myocardial I/R have not been elucidated completely. Recent studies highlight the role of Toll-like receptors (TLRs) in the induction of innate immune and inflammatory responses. Growing evidence has demonstrated that TLRs play a critical role in myocardial I/R injury. Specifically, deficiency of TLR4 protects the myocardium from ischemic injury, whereas modulation of TLR2 induces cardioprotection against ischemic insult. Importantly, cardioprotection induced by modulation of TLRs involves activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, suggesting that there is a crosstalk between TLRs and PI3K/Akt signaling pathways. In addition, TLRs also associate with other coreceptors, such as macrophage scavenger receptors in the recognition of their ligands. TLRs are also involved in the induction of angiogenesis, modulation of stem cell function, and expression of microRNA, which are currently important topic areas in myocardial I/R. Understanding how TLRs contribute to myocardial I/R injury could provide basic scientific knowledge for the development of new therapeutic approaches for the treatment and management of patients with heart attack.
Collapse
Affiliation(s)
- Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | | | | | | | | | | |
Collapse
|
37
|
Lu C, Liu L, Chen Y, Ha T, Kelley J, Schweitzer J, Kalbfleisch JH, Kao RL, Williams DL, Li C. TLR2 ligand induces protection against cerebral ischemia/reperfusion injury via activation of phosphoinositide 3-kinase/Akt signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1458-66. [PMID: 21709150 PMCID: PMC3140609 DOI: 10.4049/jimmunol.1003428] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
This study examined the effect of TLR2 activation by its specific ligand, Pam3CSK4, on cerebral ischemia/reperfusion (I/R) injury. Mice (n = 8/group) were treated with Pam3CSK4 1 h before cerebral ischemia (60 min), followed by reperfusion (24 h). Pam3CSK4 was also given to the mice (n = 8) 30 min after ischemia. Infarct size was determined by triphenyltetrazolium chloride staining. The morphology of neurons in brain sections was examined by Nissl staining. Pam3CSK4 administration significantly reduced infarct size by 55.9% (p < 0.01) compared with untreated I/R mice. Therapeutic treatment with Pam3CSK4 also significantly reduced infarct size by 55.8%. Morphologic examination showed that there was less neuronal damage in the hippocampus of Pam3CSK4-treated mice compared with untreated cerebral I/R mice. Pam3CSK4 treatment increased the levels of Hsp27, Hsp70, and Bcl2, and decreased Bax levels and NF-κB-binding activity in the brain tissues. Administration of Pam3CSK4 significantly increased the levels of phospho-Akt/Akt and phospho-GSK-3β/GSK-3β compared with untreated I/R mice. More significantly, either TLR2 deficiency or PI3K inhibition with LY29004 abolished the protection by Pam3CSK4. These data demonstrate that activation of TLR2 by its ligand prevents focal cerebral ischemic damage through a TLR2/PI3K/Akt-dependent mechanism. Of greater significance, these data indicate that therapy with a TLR2-specific agonist during cerebral ischemia is effective in reducing injury.
Collapse
Affiliation(s)
- Chen Lu
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuling Chen
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Tuanzhu Ha
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - Jim Kelley
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - John Schweitzer
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - John H. Kalbfleisch
- Department of Biometry and Medical Computing, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - Race L. Kao
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - David L. Williams
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - Chuanfu Li
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| |
Collapse
|
38
|
Abstract
The discovery and characterization of the TLR (Toll-like receptor) family has led to a better understanding of the innate immune system. The strategy of innate immune recognition is based on the detection of constitutive and conserved products of micro-organisms. However, host molecules that are released during injury can also activate TLRs. Engagement of TLRs by microbial or host-derived molecules induces the expression of pro-inflammatory cytokines, which may have both beneficial and detrimental effects on the host. In addition to being expressed in immune cells, TLRs are expressed in other tissues such as those of the cardiovascular system. In the present review, the role of TLRs in septic cardiomyopathy, viral myocarditis, atherosclerosis, ischaemia/reperfusion injury and cardiac remodelling after myocardial infarction are outlined, with attention paid to genetically modified murine models. Although much has been learned about stress-induced TLR activation in the tissues of the cardiovascular system, the role of individual TLRs in initiating and integrating homoeostatic responses within the heart remains to be defined. Accumulating evidence indicates that TLRs may play an important role in the pathogenesis of atherosclerosis, viral myocarditis, dilated cardiomyopathy, cardiac allograft rejection and sepsis-induced left ventricular dysfunction. Moreover, heart failure of diverse aetiology is also now recognized to have an important immune component, with TLR signalling influencing the process of cardiac remodelling and prognosis. In the present review, we outline the biology of TLRs as well as the current experimental and clinical evidence for the role of TLRs in cardiovascular diseases.
Collapse
|
39
|
The essential roles of Toll-like receptor signaling pathways in sterile inflammatory diseases. Int Immunopharmacol 2011; 11:1422-32. [PMID: 21600309 DOI: 10.1016/j.intimp.2011.04.026] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Revised: 04/29/2011] [Accepted: 04/30/2011] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) form a family of pattern recognition receptors with at least 11 members in human and 13 in mouse. TLRs recognize a wide variety of putative host-derived agonists that have emerged as key mediators of innate immunity. TLR signaling also plays an important role in the activation of the adaptive immune system by inducing pro-inflammatory cytokines and upregulating costimulatory molecules of antigen presenting cells. Inappropriate activation of TLRs by self-components generated by damaged tissues may result in sterile inflammation. This review discusses the contribution of TLR signaling to the initiation and progression of non-infectious inflammatory processes, such as ischemia and reperfusion (I/R) injury, tissue repair and regeneration and autoimmune diseases. The involvement of TLR signaling in the pathogenesis of sterile inflammation-related diseases may provide novel targets for the development of therapeutics.
Collapse
|
40
|
Du X, Fleiss B, Li H, D'angelo B, Sun Y, Zhu C, Hagberg H, Levy O, Mallard C, Wang X. Systemic stimulation of TLR2 impairs neonatal mouse brain development. PLoS One 2011; 6:e19583. [PMID: 21573120 PMCID: PMC3089625 DOI: 10.1371/journal.pone.0019583] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Accepted: 04/12/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Inflammation is associated with perinatal brain injury but the underlying mechanisms are not completely characterized. Stimulation of Toll-like receptors (TLRs) through specific agonists induces inflammatory responses that trigger both innate and adaptive immune responses. The impact of engagement of TLR2 signaling pathways on the neonatal brain is still unclear. The aim of this study was to investigate the potential effect of a TLR2 agonist on neonatal brain development. METHODOLOGY/PRINCIPAL FINDINGS Mice were injected intraperitoneally (i.p.) once a day from postnatal day (PND) 3 to PND11 with endotoxin-free saline, a TLR2 agonist Pam(3)CSK(4) (5 mg/kg) or Lipopolysaccharide (LPS, 0.3 mg/kg). Pups were sacrificed at PND12 or PND53 and brain, spleen and liver were collected and weighed. Brain sections were stained for brain injury markers. Long-term effects on memory function were assessed using the Trace Fear Conditioning test at PND50. After 9 days of Pam(3)CSK(4) administration, we found a decreased volume of cerebral gray matter, white matter in the forebrain and cerebellar molecular layer that was accompanied by an increase in spleen and liver weight at PND12. Such effects were not observed in Pam3CSK4-treated TLR 2-deficient mice. Pam3CSK4-treated mice also displayed decreased hippocampus neuronal density, and increased cerebral microglia density, while there was no effect on caspase-3 or general cell proliferation at PND12. Significantly elevated levels of IL-1β, IL-6, KC, and MCP-1 were detected after the first Pam3CSK4 injection in brain homogenates of PND3 mice. Pam(3)CSK(4) administration did not affect long-term memory function nor the volume of gray or white matter. CONCLUSIONS/SIGNIFICANCE Repeated systemic exposure to the TLR2 agonist Pam(3)CSK(4) can have a short-term negative impact on the neonatal mouse brain.
Collapse
Affiliation(s)
- Xiaonan Du
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Bobbi Fleiss
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Hongfu Li
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Barbara D'angelo
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yanyan Sun
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Perinatal Center, Department of Obstetrics and Gynecology, University of Gothenburg, Gothenburg, Sweden
- Institute of Reproductive and Developmental Biology, Imperial College, London, United Kingdom
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carina Mallard
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoyang Wang
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
41
|
Mersmann J, Latsch K, Habeck K, Zacharowski K. Measure for measure-determination of infarct size in murine models of myocardial ischemia and reperfusion: a systematic review. Shock 2011; 35:449-55. [PMID: 21192281 DOI: 10.1097/shk.0b013e318208af64] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myocardial ischemia/reperfusion injury (MI/R) is one of the most prominent topics of contemporary research. The frequent failure of potential therapeutic drugs and interventions to transfer to clinical practice demonstrates the limitations in using experimental animal models. Because a variety of transgenic animals are readily available in mice, researchers in recent years have made use of murine models rather than of larger animal models for experimental MI/R. This review focuses on in vivo and ex vivo murine models of MI/R and aims to characterize the source of our mechanistic understanding in mice. A systematic review of the literature demonstrated that there is great diversity among ex vivo (Langendorff) and in vivo models of MI/R.
Collapse
Affiliation(s)
- Jan Mersmann
- Clinic of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | | | | | | |
Collapse
|
42
|
Wang YY, Liu S, Lian F, Yang WG, Xue S. Toll-like receptor 7/8 agonist resiquimod induces late preconditioning in neonatal cardiac myocytes. Acta Pharmacol Sin 2011; 32:565-72. [PMID: 21516132 DOI: 10.1038/aps.2011.6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIM To investigate whether R-848 (resiquimod, toll-like receptor 7/8 agonist) can induce late preconditioning in neonatal cardiac myocytes. METHODS The protective effects of R-848 on neonatal myocytes against anoxia-reoxygenation-induced injury were tested, and intracellular reactive oxygen species (ROS) were determined. The protein synthesis inhibitor cyclohexamide (CH) and the ROS scavenger N-acetylcysteine (NAC) were used in this model to test if new protein synthesis and oxidative stress were necessary for their cardioprotective effects. The activation of nuclear factor kappa B (NFκB) and hypoxia inducible factor 1 (HIF1) was investigated by electrophoretic mobility shift assays (EMSA), and inducible nitric oxide synthase (iNOS) was assessed by immunoblotting. After iNOS was down-regulated by small interfering RNA (siRNA) transfection, the cardioprotective effect was reassessed. RESULTS ROS were triggered soon after R-848 (0.01-1.0 μg/L) administration, however, the cardioprotective effect of which was induced 24 h later. This protection was abolished by CH or NAC pretreatment. NFκB and HIF1 activation and iNOS up-regulation were involved in this protective mechanism. The cardioprotective effect was also attenuated after iNOS was knocked down. CONCLUSION R-848 provided a cardioprotective effect through a late preconditioning mechanism via a ROS/NFκB-HIF1/iNOS-dependent pathway.
Collapse
|
43
|
Bingold TM, Pullmann B, Sartorius S, Geiger EV, Marzi I, Zacharowski K, Wissing H, Scheller B. Soluble triggering receptor on myeloid cells-1 is expressed in the course of non-infectious inflammation after traumatic lung contusion: a prospective cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R115. [PMID: 21496225 PMCID: PMC3219398 DOI: 10.1186/cc10141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 03/25/2011] [Accepted: 04/15/2011] [Indexed: 01/19/2023]
Abstract
INTRODUCTION The triggering receptor expressed on myeloid cells-1 (TREM-1) is known to be expressed during bacterial infections. We investigated whether TREM-1 is also expressed in non-infectious inflammation following traumatic lung contusion. METHODS In a study population of 45 adult patients with multiple trauma and lung contusion, we obtained bronchoalveolar lavage (BAL) (blind suctioning of 20 ml NaCl (0.9%) via jet catheter) and collected blood samples at two time points (16 hours and 40 hours) after trauma. Post hoc patients were assigned to one of four groups radiologically classified according to the severity of lung contusion based on the initial chest tomography. Concentration of soluble TREM-1 (sTREM-1) and bacterial growth were determined in the BAL. sTREM-1, IL-6, IL-10, lipopolysaccharide binding protein, procalcitonin, C-reactive protein and leukocyte count were assessed in blood samples. Pulmonary function was evaluated by the paO2/FiO2 ratio. RESULTS Three patients were excluded due to positive bacterial growth in the initial BAL. In 42 patients the severity of lung contusion correlated with the levels of sTREM-1 16 hours and 40 hours after trauma. sTREM-1 levels were significantly (P < 0.01) elevated in patients with severe contusion (2,184 pg/ml (620 to 4,000 pg/ml)) in comparison with patients with mild (339 pg/ml (135 to 731 pg/ml)) or no (217 pg/ml (97 to 701 pg/ml)) contusion 40 hours following trauma. At both time points the paO2/FiO2 ratio correlated negatively with sTREM-1 levels (Spearman correlation coefficient = -0.446, P < 0.01). CONCLUSIONS sTREM-1 levels are elevated in the BAL of patients following pulmonary contusion. Furthermore, the levels of sTREM-1 in the BAL correlate well with both the severity of radiological pulmonary tissue damage and functional impairment of gas exchange (paO2/FiO2 ratio).
Collapse
Affiliation(s)
- Tobias M Bingold
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt am Main, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hofmann U, Ertl G, Frantz S. Toll-like receptors as potential therapeutic targets in cardiac dysfunction. Expert Opin Ther Targets 2011; 15:753-65. [PMID: 21385118 DOI: 10.1517/14728222.2011.566560] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The innate immune system can detect the highly conserved, relatively invariant structural motifs of pathogens. The most important innate immune receptors, Toll-like receptors (TLRs), represent a first line of defense against infectious pathogens, and play a pivotal role in initiating and shaping innate and adaptive immune responses. TLRs are not only expressed in immune cells, but also in cardiovascular cells. In addition to their role in response to microbial infections, evidence suggests that TLRs can also recognize endogenous ligands and may play a role in mediating cardiomyocyte cell death and survival after non-infectious injury. AREAS COVERED TLRs could be a link between cardiovascular diseases and the immune system. Experimentally, there is good evidence that TLR activation contributes to development and progression of both acute cardiac injury and chronic heart failure. The role of TLRs in myocardial ischemia-reperfusion, remodeling, septic cardiomyoparthy, autoimmune- and viral myocarditis, anthracycline-induced cardiomyopathy and cardiac hypertrophy, in basic as well as clinical science are discussed. EXPERT OPINION Evidence, mainly from animal experiments, indicates that TLRs contribute to all of the myocardial disease states reviewed in this paper. However, the relevance of TLRs as therapeutic targets remains to be defined as clinical data is sparse.
Collapse
Affiliation(s)
- Ulrich Hofmann
- University Hospital Würzburg, Department of Internal Medicine I, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | | | | |
Collapse
|
45
|
Wang E, Feng Y, Zhang M, Zou L, Li Y, Buys ES, Huang P, Brouckaert P, Chao W. Toll-like receptor 4 signaling confers cardiac protection against ischemic injury via inducible nitric oxide synthase- and soluble guanylate cyclase-dependent mechanisms. Anesthesiology 2011; 114:603-13. [PMID: 21270629 PMCID: PMC3044772 DOI: 10.1097/aln.0b013e31820a4d5b] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Prior administration of a small dose of lipopolysaccharide confers a cardiac protection against ischemia-reperfusion injury. However, the signaling mechanisms that control the protection are incompletely understood. We tested the hypothesis that Toll-like receptor 4 (TLR4) mediates the ability of lipopolysaccharide to protect against cardiac ischemia-reperfusion injury through distinct intracellular pathways involving myeloid differentiation factor 88 (MyD88), TIR-domain-containing adaptor protein-inducing interferon-β-mediated transcription factor (Trif), inducible nitric oxide synthase (iNOS), and soluble guanylate cyclase (sGC). METHODS Wild-type mice and genetically modified mice, that is TLR4-deficient (TLR4(-def)), TLR2 knockout (TLR2(-/-)), MyD88(-/-), Trif(-/-), iNOS(-/-), and sGCα1(-/-), were treated with normal saline or 0.1 mg/kg lipopolysaccharide intraperitoneally. Twenty-four hours later, isolated hearts were perfused in a Langendorff apparatus and subsequently subjected to 30 min global ischemia and reperfusion for as long as 60 min. Left ventricular function and myocardial infarction sizes were examined. RESULTS Compared with saline-treated mice, lipopolysaccharide-treated mice had markedly improved left ventricular developed pressure and dP/dt(max) (P < 0.01) and reduced myocardial infarction sizes (37.2 ± 3.4% vs. 19.8 ± 4.9%, P < 0.01) after ischemia-reperfusion. The cardiac protective effect of lipopolysaccharide was abolished in the TLR4(-def) and MyD88(-/-) mice but remained intact in TLR2(-/-) or Trif(-/-) mice. iNOS(-/-) mice or wild-type mice treated with the iNOS inhibitor 1400W failed to respond to the TLR4-induced nitric oxide production and were not protected by the lipopolysaccharide preconditioning. Although sGCα(1)(-/-) mice had robust nitric oxide production in response to lipopolysaccharide, they were not protected by the TLR4-elicited cardiac protection. CONCLUSIONS TLR4 activation confers a potent cardiac protection against ischemia-reperfusion injury via a MyD88-dependent, but Trif-independent, mechanism. iNOS/sGC are essential for the TLR4-induced cardiac protection.
Collapse
Affiliation(s)
- E Wang
- Research Fellow, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Associate Professor, Department of Anesthesiology, Xiangya Hospital, Central South University, China
| | - Yan Feng
- Research Fellow, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ming Zhang
- Research Fellow, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lin Zou
- Research Fellow, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yan Li
- Research Fellow, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emmanuel S. Buys
- Instructor, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peigen Huang
- Instructor, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter Brouckaert
- Professor, Department for Molecular Biomedical Research, Flanders Institute for Biotechnology, and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wei Chao
- Associate Professor, Anesthesia Center for Critical Care Research, Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Mersmann J, Habeck K, Latsch K, Zimmermann R, Jacoby C, Fischer JW, Hartmann C, Schrader J, Kirschning CJ, Zacharowski K. Left ventricular dilation in toll-like receptor 2 deficient mice after myocardial ischemia/reperfusion through defective scar formation. Basic Res Cardiol 2010; 106:89-98. [PMID: 20967453 DOI: 10.1007/s00395-010-0127-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 09/23/2010] [Accepted: 10/06/2010] [Indexed: 12/15/2022]
Abstract
Restoration of myocardial blood flow after ischemia triggers an inflammatory response involving toll-like receptors (TLRs). TLR2(-/-)-mice show short-term advantages upon reperfusion injury as compared with WT controls. Accordingly, it has been shown that transient TLR2-blockade prior to reperfusion is associated with improved left-ventricular performance after myocardial scar formation. We present here adverse myocardial remodeling due to a chronic lack of TLR2 expression. Myocardial ischemia/reperfusion (MI/R) was surgically induced in C3HeN-mice by ligation of the left anterior descending coronary artery for 20 min, followed by 24 h or 28 days of reperfusion. TLR2(-/-)-mice and TLR2-Ab treated (T2.5) WT-mice displayed a reduction of infarct size, plasma troponin T concentrations, and leukocyte infiltration as compared with untreated controls after 24 h of reperfusion. After 28 days, however, magnetic resonance imaging revealed a marked left ventricular dilation in TLR2(-/-)-animals, which was associated with pronounced matrix remodeling characterized by reduced collagen and decorin density in the infarct scar. Our data show adverse effects on myocardial remodeling in TLR2(-/-)-mice. Although interception with TLR2 signaling is a promising concept for the prevention of reperfusion injury after myocardial ischemia, these data give cause for serious concern with respect to the time-point and duration of the potential treatment.
Collapse
Affiliation(s)
- Jan Mersmann
- Clinic of Anaesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Toll-like receptor 2 signaling triggers fatal arrhythmias upon myocardial ischemia-reperfusion. Crit Care Med 2010; 38:1927-32. [PMID: 20855990 DOI: 10.1097/ccm.0b013e3181ef455b] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Restoration of myocardial blood flow after ischemia triggers an inflammatory response involving toll-like receptors. Toll-like receptor 2 deficiency is associated with a reduced infarct size after myocardial ischemia and reperfusion. Because a marked mortality was observed in C3HeN wild-type mice, which was absent in TLR2 mice, we tested whether cardiac arrhythmias are the underlying pathology and aimed to elucidate how toll-like receptor 2 ligation might prevent lethal arrhythmias. DESIGN Experimental animal model. SETTING University hospital research laboratory. SUBJECTS Male C3HeN mice. INTERVENTIONS Myocardial ischemia and reperfusion was surgically induced by ligation of the left anterior descending coronary artery for 20 mins followed by 24 hrs of reperfusion. Electrocardiography was continuously recorded during the observation period through an implantable telemetry transmitter to detect cardiac arrhythmias during reperfusion. MEASUREMENTS AND MAIN RESULTS Toll-like receptor 2 expression was associated with a 51% mortality rate (23 of 45 mice died) after myocardial ischemia and reperfusion. Absence of toll-like receptor 2 improved survival toward 100% (17 of 17 mice survived). Electrocardiography diagnostics in conscious animals and histologic analysis revealed that absence of toll-like receptor 2 signaling prevented the formation of pathologic heart rate turbulence after myocardial ischemia and reperfusion and modulated the density of connexin 43-positive gap junctions in the ischemic area compared with wild-type hearts, indicating arrhythmia as the cause underlying the observed mortality. CONCLUSIONS The results presented here indicate toll-like receptor 2 as a novel target for the prevention of lethal arrhythmic complications after myocardial ischemia and reperfusion.
Collapse
|
48
|
Preconditioning by toll-like receptor 2 agonist Pam3CSK4 reduces CXCL1-dependent leukocyte recruitment in murine myocardial ischemia/reperfusion injury: Erratum. Crit Care Med 2010. [DOI: 10.1097/ccm.0b013e3181e0afca] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Ha T, Hu Y, Liu L, Lu C, McMullen JR, Kelley J, Kao RL, Williams DL, Gao X, Li C. TLR2 ligands induce cardioprotection against ischaemia/reperfusion injury through a PI3K/Akt-dependent mechanism. Cardiovasc Res 2010; 87:694-703. [PMID: 20421349 DOI: 10.1093/cvr/cvq116] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AIMS Toll-like receptor (TLR)-mediated signalling pathways have been implicated in myocardial ischaemia/reperfusion (I/R) injury. Activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway protects the myocardium from ischaemic injury. We hypothesized that the modulation of TLR2 would induce cardioprotection against I/R injury via activation of the PI3K/Akt signalling. METHODS AND RESULTS Mice were treated with TLR2 ligands, peptidoglycan (PGN) or Pam3CSK4, respectively, 1 h before the hearts were subjected to ischaemia (1 h), followed by reperfusion (4 h). Infarct size was determined by triphenyltetrazolium chloride staining. Cardiac function and haemodynamic performance were evaluated. Infarct size was significantly reduced in PGN- or Pam3CSK4-treated mice compared with untreated I/R mice. Administration of TLR2 ligands improved cardiac function following I/R. PGN treatment increased the levels of phospho-Akt and phospho-GSK-3beta (glycogen synthase kinase-3beta), compared with untreated I/R hearts. PGN stimulation increased TLR2 tyrosine phosphorylation and association of the p85 subunit of PI3K with TLR2. To investigate the role of PI3K/Akt signalling in PGN-induced cardioprotection, we administered the PI3K inhibitor, Wortmannin, to the mice 15 min before PGN treatment. We also administered PGN to kinase-deficient Akt (kdAkt) transgenic mice 1 h before myocardial I/R. Both PI3K inhibition and kdAkt mice abolished the cardioprotection induced by PGN. To examine the role of TLR2 in PGN-induced cardioprotection, we administrated PGN to TLR2 knockout mice 1 h before the hearts were subjected to I/R. PGN-induced cardioprotection was lost in TLR2-deficient mice. CONCLUSION These results demonstrate that TLR2 ligands induced cardioprotection, which is mediated through a TLR2/PI3K/Akt-dependent mechanism.
Collapse
Affiliation(s)
- Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70575, Johnson City, TN 37614-0575, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cardiac preconditioning by specific ligands of Toll-like receptors: is it wither or whither? Crit Care Med 2010; 38:1003-4. [PMID: 20168168 DOI: 10.1097/ccm.0b013e3181cd10e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|