1
|
Tan Y, Chen P, Xin X, Wang J. Protocol for whole-cell patch-clamp assay for HBV-mediated sodium influx in THP-1 macrophages. STAR Protoc 2024; 5:103462. [PMID: 39630584 DOI: 10.1016/j.xpro.2024.103462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/24/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Sodium influx induced by hepatitis B virus (HBV) enhances activation of the NLRP3 inflammasome, in synergy with potassium efflux signals in macrophages. Here, we present a protocol for conducting patch-clamp assays to measure HBV-mediated sodium influx in THP-1 macrophages. We describe steps for differentiating, resting, and activating THP-1 macrophages and then outline the procedures for whole-cell patch-clamp recordings. For complete details on the use and execution of this protocol, please refer to Wang et al.1.
Collapse
Affiliation(s)
- Yingbao Tan
- Department of Immunology, Army Medical University, Chongqing 400038, P.R. China
| | - Penghui Chen
- Department of Neurobiology, Army Medical University, Chongqing 400038, P.R. China
| | - Xiaojuan Xin
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, P.R. China
| | - Jingxue Wang
- Department of Immunology, Army Medical University, Chongqing 400038, P.R. China.
| |
Collapse
|
2
|
Zhou Y, Huang X, Jin Y, Qiu M, Ambe PC, Basharat Z, Hong W. The role of mitochondrial damage-associated molecular patterns in acute pancreatitis. Biomed Pharmacother 2024; 175:116690. [PMID: 38718519 DOI: 10.1016/j.biopha.2024.116690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 06/03/2024] Open
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal tract diseases with significant morbidity and mortality. Current treatments remain unspecific and supportive due to the severity and clinical course of AP, which can fluctuate rapidly and unpredictably. Mitochondria, cellular power plant to produce energy, are involved in a variety of physiological or pathological activities in human body. There is a growing evidence indicating that mitochondria damage-associated molecular patterns (mtDAMPs) play an important role in pathogenesis and progression of AP. With the pro-inflammatory properties, released mtDAMPs may damage pancreatic cells by binding with receptors, activating downstream molecules and releasing inflammatory factors. This review focuses on the possible interaction between AP and mtDAMPs, which include cytochrome c (Cyt c), mitochondrial transcription factor A (TFAM), mitochondrial DNA (mtDNA), cardiolipin (CL), adenosine triphosphate (ATP) and succinate, with focus on experimental research and potential therapeutic targets in clinical practice. Preventing or diminishing the release of mtDAMPs or targeting the mtDAMPs receptors might have a role in AP progression.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaoyi Huang
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yinglu Jin
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Minhao Qiu
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Peter C Ambe
- Department of General Surgery, Visceral Surgery and Coloproctology, Vinzenz-Pallotti-Hospital Bensberg, Vinzenz-Pallotti-Str. 20-24, Bensberg 51429, Germany
| | | | - Wandong Hong
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
3
|
Golino M, Harding D, Del Buono MG, Fanti S, Mohiddin S, Toldo S, Smyth J, Sanna T, Marelli-Berg F, Abbate A. Innate and adaptive immunity in acute myocarditis. Int J Cardiol 2024; 404:131901. [PMID: 38403204 PMCID: PMC11450758 DOI: 10.1016/j.ijcard.2024.131901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
Acute myocarditis is an acute inflammatory cardiomyopathy associated with cardiac damage triggered by a virus or a pathological immune activation. It may present with a wide range of clinical presentations, ranging from mild symptoms to severe forms like fulminant myocarditis, characterized by hemodynamic compromise and cardiogenic shock. The immune system plays a central role in the pathogenesis of myocarditis. In fact, while its function is primarily protective, aberrant responses can be detrimental. In this context, both innate and adaptive immunity play pivotal roles; notably, the innate system offers a non-specific and immediate defense, while the adaptive provides specialized protection with immunological memory. However, dysregulation in these systems can misidentify cardiac tissue, triggering autoimmune reactions and possibly leading to significant cardiac tissue damage. This review highlights the importance of innate and adaptive immune responses in the progression and treatment of acute myocarditis.
Collapse
Affiliation(s)
- Michele Golino
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America; Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Daniel Harding
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Saidi Mohiddin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom; Barts Heart Centre, London, United Kingdom
| | - Stefano Toldo
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
| | - James Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, United States of America; Virginia Tech Carilion School of Medicine, Roanoke, VA, United States of America; Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, United States of America
| | - Tommaso Sanna
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom.
| | - Antonio Abbate
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America.
| |
Collapse
|
4
|
Cohen MV, Downey JM. Initial Despair and Current Hope of Identifying a Clinically Useful Treatment of Myocardial Reperfusion Injury: Insights Derived from Studies of Platelet P2Y 12 Antagonists and Interference with Inflammation and NLRP3 Assembly. Int J Mol Sci 2024; 25:5477. [PMID: 38791515 PMCID: PMC11122283 DOI: 10.3390/ijms25105477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Myocardial necrosis following the successful reperfusion of a coronary artery occluded by thrombus in a patient presenting with ST-elevation myocardial infarction (STEMI) continues to be a serious problem, despite the multiple attempts to attenuate the necrosis with agents that have shown promise in pre-clinical investigations. Possible reasons include confounding clinical risk factors, the delayed application of protective agents, poorly designed pre-clinical investigations, the possible effects of routinely administered agents that might unknowingly already have protected the myocardium or that might have blocked protection, and the biological differences of the myocardium in humans and experimental animals. A better understanding of the pathobiology of myocardial infarction is needed to stem this reperfusion injury. P2Y12 receptor antagonists minimize platelet aggregation and are currently part of the standard treatment to prevent thrombus formation and propagation in STEMI protocols. Serendipitously, these P2Y12 antagonists also dramatically attenuate reperfusion injury in experimental animals and are presumed to provide a similar protection in STEMI patients. However, additional protective agents are needed to further diminish reperfusion injury. It is possible to achieve additive protection if the added intervention protects by a mechanism different from that of P2Y12 antagonists. Inflammation is now recognized to be a critical factor in the complex intracellular response to ischemia and reperfusion that leads to tissue necrosis. Interference with cardiomyocyte inflammasome assembly and activation has shown great promise in attenuating reperfusion injury in pre-clinical animal models. And the blockade of the executioner protease caspase-1, indeed, supplements the protection already seen after the administration of P2Y12 antagonists. Importantly, protective interventions must be applied in the first minutes of reperfusion, if protection is to be achieved. The promise of such a combination of protective strategies provides hope that the successful attenuation of reperfusion injury is attainable.
Collapse
Affiliation(s)
- Michael V. Cohen
- The Departments of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA;
- The Departments of Medicine, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA
| | - James M. Downey
- The Departments of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA;
| |
Collapse
|
5
|
Zhang H, Hu H, Zhai C, Jing L, Tian H. Cardioprotective Strategies After Ischemia-Reperfusion Injury. Am J Cardiovasc Drugs 2024; 24:5-18. [PMID: 37815758 PMCID: PMC10806044 DOI: 10.1007/s40256-023-00614-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Acute myocardial infarction (AMI) is associated with high morbidity and mortality worldwide. Although early reperfusion is the most effective strategy to salvage ischemic myocardium, reperfusion injury can develop with the restoration of blood flow. Therefore, it is important to identify protection mechanisms and strategies for the heart after myocardial infarction. Recent studies have shown that multiple intracellular molecules and signaling pathways are involved in cardioprotection. Meanwhile, device-based cardioprotective modalities such as cardiac left ventricular unloading, hypothermia, coronary sinus intervention, supersaturated oxygen (SSO2), and remote ischemic conditioning (RIC) have become important areas of research. Herein, we review the molecular mechanisms of cardioprotection and cardioprotective modalities after ischemia-reperfusion injury (IRI) to identify potential approaches to reduce mortality and improve prognosis in patients with AMI.
Collapse
Affiliation(s)
- Honghong Zhang
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Huilin Hu
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China.
| | - Changlin Zhai
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Lele Jing
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Hongen Tian
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| |
Collapse
|
6
|
Mamsa R, Prabhavalkar KS, Bhatt LK. Crosstalk between NLRP3 inflammasome and calpain in Alzheimer's disease. Eur J Neurosci 2023; 58:3719-3731. [PMID: 37652164 DOI: 10.1111/ejn.16139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/12/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Amyloid plaques are considered to be the pathological hallmark of Alzheimer's disease (AD). Neuroinflammation further aggravates the pathogenesis of Alzheimer's disease. Calpains and NOD-like receptor protein-3 (NLRP3) inflammasomes are involved in the neuroinflammatory pathway and affect the progression of Alzheimer's disease. Hyperactivation of calpains is responsible for the activation of NLRP3 inflammasome, thereby affecting each other's molecular mechanism and causing astrogliosis, microgliosis, and neuronal dysfunction. Further, calpain hyperactivation is also associated with calcium homeostasis that acts as one of the triggers in the activation of NLRP3 inflammasome. Calpain activity is required for the maturation of interleukin-1β, a key mediator of neuroinflammatory responses. The membrane potential/calcium/calpain/caspase-1 axis acts as an unconventional regulator of inflammasomes. The complex crosstalk between NLRP3 inflammasome and calpain leads to a series of events. Targeting the molecular mechanism associated with calpain-NLRP3 inflammasome activation and regulation can be a therapeutic and prophylactic perspective towards Alzheimer's disease. This review discusses calpains and NLRP3 inflammasome crosstalk in the pathogenesis of AD.
Collapse
Affiliation(s)
- Rumaiza Mamsa
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
7
|
Mu W, Xu G, Wang Z, Li Q, Sun S, Qin Q, Li Z, Shi W, Dai W, Zhan X, Wang J, Bai Z, Xiao X. Tricyclic antidepressants induce liver inflammation by targeting NLRP3 inflammasome activation. Cell Commun Signal 2023; 21:123. [PMID: 37231437 DOI: 10.1186/s12964-023-01128-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 04/15/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Idiosyncratic drug-induced liver injury (IDILI) is common in hepatology practices and, in some cases, lethal. Increasing evidence show that tricyclic antidepressants (TCAs) can induce IDILI in clinical applications but the underlying mechanisms are still poorly understood. METHODS We assessed the specificity of several TCAs for NLRP3 inflammasome via MCC950 (a selective NLRP3 inhibitor) pretreatment and Nlrp3 knockout (Nlrp3-/-) BMDMs. Meanwhile, the role of NLRP3 inflammasome in the TCA nortriptyline-induced hepatotoxicity was demonstrated in Nlrp3-/- mice. RESULTS We reported here that nortriptyline, a common TCA, induced idiosyncratic hepatotoxicity in a NLRP3 inflammasome-dependent manner in mildly inflammatory states. In parallel in vitro studies, nortriptyline triggered the inflammasome activation, which was completely blocked by Nlrp3 deficiency or MCC950 pretreatment. Furthermore, nortriptyline treatment led to mitochondrial damage and subsequent mitochondrial reactive oxygen species (mtROS) production resulting in aberrant activation of the NLRP3 inflammasome; a selective mitochondrial ROS inhibitor pretreatment dramatically abrogated nortriptyline-triggered the NLRP3 inflammasome activation. Notably, exposure to other TCAs also induced aberrant activation of the NLRP3 inflammasome by triggering upstream signaling events. CONCLUSION Collectively, our findings revealed that the NLRP3 inflammasome may act as a crucial target for TCA agents and suggested that the core structures of TCAs may contribute to the aberrant activation of NLRP3 inflammasome induced by them, an important factor involved in the pathogenesis of TCA-induced liver injury. Video Abstract.
Collapse
Affiliation(s)
- Wenqing Mu
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, Jiangsu, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Guang Xu
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Zhilei Wang
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Qiang Li
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Siqiao Sun
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Qin Qin
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhiyong Li
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wenzhang Dai
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaoyan Zhan
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jiabo Wang
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhaofang Bai
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiaohe Xiao
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
8
|
Wang Q, Zuurbier CJ, Huhn R, Torregroza C, Hollmann MW, Preckel B, van den Brom CE, Weber NC. Pharmacological Cardioprotection against Ischemia Reperfusion Injury-The Search for a Clinical Effective Therapy. Cells 2023; 12:1432. [PMID: 37408266 DOI: 10.3390/cells12101432] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
Pharmacological conditioning aims to protect the heart from myocardial ischemia-reperfusion injury (IRI). Despite extensive research in this area, today, a significant gap remains between experimental findings and clinical practice. This review provides an update on recent developments in pharmacological conditioning in the experimental setting and summarizes the clinical evidence of these cardioprotective strategies in the perioperative setting. We start describing the crucial cellular processes during ischemia and reperfusion that drive acute IRI through changes in critical compounds (∆GATP, Na+, Ca2+, pH, glycogen, succinate, glucose-6-phosphate, mitoHKII, acylcarnitines, BH4, and NAD+). These compounds all precipitate common end-effector mechanisms of IRI, such as reactive oxygen species (ROS) generation, Ca2+ overload, and mitochondrial permeability transition pore opening (mPTP). We further discuss novel promising interventions targeting these processes, with emphasis on cardiomyocytes and the endothelium. The limited translatability from basic research to clinical practice is likely due to the lack of comorbidities, comedications, and peri-operative treatments in preclinical animal models, employing only monotherapy/monointervention, and the use of no-flow (always in preclinical models) versus low-flow ischemia (often in humans). Future research should focus on improved matching between preclinical models and clinical reality, and on aligning multitarget therapy with optimized dosing and timing towards the human condition.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Ragnar Huhn
- Department of Anesthesiology, Kerckhoff-Clinic-Center for Heart, Lung, Vascular and Rheumatic Disease, Justus-Liebig-University Giessen, Benekestr. 2-8, 61231 Bad Nauheim, Germany
| | - Carolin Torregroza
- Department of Anesthesiology, Kerckhoff-Clinic-Center for Heart, Lung, Vascular and Rheumatic Disease, Justus-Liebig-University Giessen, Benekestr. 2-8, 61231 Bad Nauheim, Germany
| | - Markus W Hollmann
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Charissa E van den Brom
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
9
|
Yang C, Pan Q, Ji K, Tian Z, Zhou H, Li S, Luo C, Li J. Review on the protective mechanism of astragaloside IV against cardiovascular diseases. Front Pharmacol 2023; 14:1187910. [PMID: 37251311 PMCID: PMC10213926 DOI: 10.3389/fphar.2023.1187910] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Cardiovascular disease is a global health problem. Astragaloside IV (AS-IV) is a saponin compound extracted from the roots of the Chinese herb Astragalus. Over the past few decades, AS-IV has been shown to possess various pharmacological properties. It can protect the myocardium through antioxidative stress, anti-inflammatory effects, regulation of calcium homeostasis, improvement of myocardial energy metabolism, anti-apoptosis, anti-cardiomyocyte hypertrophy, anti-myocardial fibrosis, regulation of myocardial autophagy, and improvement of myocardial microcirculation. AS-IV exerts protective effects on blood vessels. For example, it can protect vascular endothelial cells through antioxidative stress and anti-inflammatory pathways, relax blood vessels, stabilize atherosclerotic plaques, and inhibit the proliferation and migration of vascular smooth muscle cells. Thus, the bioavailability of AS-IV is low. Toxicology indicates that AS-IV is safe, but should be used cautiously in pregnant women. In this paper, we review the mechanisms of AS-IV prevention and treatment of cardiovascular diseases in recent years to provide a reference for future research and drug development.
Collapse
Affiliation(s)
- Chunkun Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Zhuang Tian
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Hongyuan Zhou
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Chuanchao Luo
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Jun Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Yang XM, Cohen MV, Sayner S, Audia JP, Downey JM. Lethal Caspase-1/4-Dependent Injury Occurs in the First Minutes of Coronary Reperfusion and Requires Calpain Activity. Int J Mol Sci 2023; 24:3801. [PMID: 36835212 PMCID: PMC9960231 DOI: 10.3390/ijms24043801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
To study the relationship between caspase-1/4 and reperfusion injury, we measured infarct size (IS) in isolated mouse hearts undergoing 50 min global ischemia/2 h reperfusion. Starting VRT-043198 (VRT) at reperfusion halved IS. The pan-caspase inhibitor emricasan duplicated VRT's protection. IS in caspase-1/4-knockout hearts was similarly reduced, supporting the hypothesis that caspase-1/4 was VRT's only protective target. NLRC4 inflammasomes activate caspase-1. NLRC4 knockout hearts were not protected, eliminating NLRC4 as caspase-1/4's activator. The amount of protection that could be achieved by only suppressing caspase-1/4 activity was limited. In wild-type (WT) hearts, ischemic preconditioning (IPC) was as protective as caspase-1/4 inhibitors. Combining IPC and emricasan in these hearts or preconditioning caspase-1/4-knockout hearts produced an additive IS reduction, indicating that more protection could be achieved by combining treatments. We determined when caspase-1/4 exerted its lethal injury. Starting VRT after 10 min of reperfusion in WT hearts was no longer protective, revealing that caspase-1/4 inflicted its injury within the first 10 min of reperfusion. Ca++ influx at reperfusion might activate caspase-1/4. We tested whether Ca++-dependent soluble adenylyl cyclase (AC10) could be responsible. However, IS in AC10-/- hearts was not different from that in WT control hearts. Ca++-activated calpain has been implicated in reperfusion injury. Calpain could be releasing actin-bound procaspase-1 in cardiomyocytes, which would explain why caspase-1/4-related injury is confined to early reperfusion. The calpain inhibitor calpeptin duplicated emricasan's protection. Unlike IPC, adding calpain to emricasan offered no additional protection, suggesting that caspase-1/4 and calpain may share the same protective target.
Collapse
Affiliation(s)
- Xi-Ming Yang
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Michael V. Cohen
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
- Department of Medicine, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Sarah Sayner
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Jonathon P. Audia
- Department of Microbiology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - James M. Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
11
|
Long JX, Tian MZ, Chen XY, Yu HH, Ding H, Liu F, Du K. The role of NLRP3 inflammasome-mediated pyroptosis in ischemic stroke and the intervention of traditional Chinese medicine. Front Pharmacol 2023; 14:1151196. [PMID: 37153784 PMCID: PMC10160381 DOI: 10.3389/fphar.2023.1151196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/07/2023] [Indexed: 05/10/2023] Open
Abstract
Ischemic stroke (IS) is the second leading cause of death and disability in the world. Pyroptosis, a form of programmed cell death initiated by caspases, participates in the occurrence and development of IS. Because it can increase cell membrane permeability, mediate the release of inflammatory factors, and aggravate inflammation, inhibiting this process can significantly reduce the pathological injury of IS. The nucleotide binding oligomerization domain-like receptor family pyrin domain protein 3 (NLRP3) is a multiprotein complex whose activation is the core link of pyroptosis. In recent years, studies have reported that traditional Chinese medicine (TCM) could regulate pyroptosis mediated by NLRP3 inflammasome through multi-channel and multi-target networks and thus exert the effect against IS. This article reviews 107 papers published in recent years in PubMed, Chinese National Knowledge Infrastructure (CNKI), and WanFang Data in recent years. It has found that the activation factors of NLRP3 inflammasome include ROS, mitochondrial dysfunction, K+, Ca2+, lysosome rupture, and trans-Golgi breakdown. TLR4/NF-κB/NLRP3, ROS/TXNIP/NLRP3, AMPK/Nrf2/NLRP3, DRP1/NLRP3, TAK1/JNK/NLRP3 signaling pathways regulate the initiation and assembly of the NLRP3 inflammasome, subsequently induce pyroptosis, affecting the occurrence and development of IS. TCM can affect the above signaling pathways and regulate the pyroptosis mediated by NLRP3 inflammasome, so as to play a protective role against IS, which provides a new entry point for discussing the pathological mechanism of IS and a theoretical basis for developing TCM treasure house.
Collapse
Affiliation(s)
- Jia-Xin Long
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Meng-Zhi Tian
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Xiao-Yi Chen
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Huang-He Yu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Huang Ding
- College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fang Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Ke Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Ke Du,
| |
Collapse
|
12
|
Zheng C, Yang C, Gao D, Zhang L, Li Y, Li L, Zhang L. Cornel Iridoid Glycoside Alleviates Microglia-Mediated Inflammatory Response via the NLRP3/Calpain Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11967-11980. [PMID: 36104266 DOI: 10.1021/acs.jafc.2c03851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vascular dementia (VaD) is associated with cerebral hypoperfusion, which results in long-term cognitive impairment and memory loss. Cornel iridoid glycoside (CIG) is the major active constituent isolated from the ripe fruit of Cornus officinalis. Previous studies have shown that CIG enhances neurological function in VaD rats. In the present research, we attempted to clarify the molecular processes underlying the role of CIG in neuroinflammation in VaD. We created a chronic cerebral ischemia rat model by ligation of the bilateral common carotid arteries (2VO) and then treated rats with different concentrations of CIG. Comprehensive analyses revealed that CIG ameliorated myelin integrity and neuronal loss. Furthermore, we also found that CIG inhibited polarized microglia activation and attenuated inflammasome-mediated production of proinflammatory cytokines in BV2 microglia cells induced by LPS/IFN-γ and in the brains of 2VO rats. To further elucidate the role of CIG in microglia-mediated inflammatory response, we investigated the expression and activity of calpain. CIG inhibited the expression and activity of calpain 1/2, which was characterized by decreased calpastatin and spectrin αII expression. In particular, intra- and extracellular calpain 1 levels were reduced by CIG. However, CIG showed weak interaction with calpain 1. In addition, we found that CG administration significantly repressed the assembly of the NOD-like receptor protein 3 (NLRP3) inflammasome, including NLRP3, ASC, and caspase-1. In conclusion, our knowledge of the mechanisms by which CIG regulates NLRP3/calpain signaling to influence inflammatory responses offers further insights into potential therapeutic strategies to treat VaD.
Collapse
Affiliation(s)
- Cengceng Zheng
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Cuicui Yang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Dan Gao
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Yali Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| |
Collapse
|
13
|
Activation and Pharmacological Regulation of Inflammasomes. Biomolecules 2022; 12:biom12071005. [PMID: 35883561 PMCID: PMC9313256 DOI: 10.3390/biom12071005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammasomes are intracellular signaling complexes of the innate immune system, which is part of the response to exogenous pathogens or physiological aberration. The multiprotein complexes mainly consist of sensor proteins, adaptors, and pro-caspase-1. The assembly of the inflammasome upon extracellular and intracellular cues drives the activation of caspase-1, which processes pro-inflammatory cytokines IL-1β and IL-18 to maturation and gasdermin-D for pore formation, leading to pyroptosis and cytokine release. Inflammasome signaling functions in numerous infectious or sterile inflammatory diseases, including inherited autoinflammatory diseases, metabolic disorders, cardiovascular diseases, cancers, neurodegenerative disorders, and COVID-19. In this review, we summarized current ideas on the organization and activation of inflammasomes, with details on the molecular mechanisms, regulations, and interventions. The recent developments of pharmacological strategies targeting inflammasomes as disease therapeutics were also covered.
Collapse
|
14
|
Zhao Y, Sun L. Bacillus cereus cytotoxin K triggers gasdermin D-dependent pyroptosis. Cell Death Dis 2022; 8:305. [PMID: 35788609 PMCID: PMC9253000 DOI: 10.1038/s41420-022-01091-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022]
Abstract
Bacillus cereus is well known as a causative agent of foodborne gastrointestinal diseases and systemic non-gastrointestinal diseases. We have recently identified a pathogenic B. cereus (named H2) from a deep-sea cold-seep. H2 possesses the pyroptosis-inducing capacity and contains a number of enterotoxins including cytotoxin K (CytK). In the present work, we examined the cytotoxicity of the CytK of H2 to human macrophages. CytK bound macrophages by interaction with the plasma membrane and caused cellular structure damage. CytK−cell interaction triggered rapid pyroptosis mediated by caspase 1-activated gasdermin D (GSDMD). CytK-induced pyroptosis required NLRP3 inflammasome activation, K+ efflux, and intracellular Ca2+ accumulation. CytK exhibited apparent binding to several cytomembrane lipids, in particular phosphatidic acid, which proved to be essential to CytK-elicited cell death. Together, these results add new insights into the cytotoxic mechanism of CytK.
Collapse
Affiliation(s)
- Yan Zhao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China. .,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China. .,College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
15
|
Penna C, Comità S, Tullio F, Alloatti G, Pagliaro P. Challenges facing the clinical translation of cardioprotection: 35 years after the discovery of ischemic preconditioning. Vascul Pharmacol 2022; 144:106995. [PMID: 35470102 DOI: 10.1016/j.vph.2022.106995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/17/2022] [Accepted: 04/16/2022] [Indexed: 12/19/2022]
|
16
|
Miao Z, Miao Z, Teng X, Xu S. Chlorpyrifos triggers epithelioma papulosum cyprini cell pyroptosis via miR-124-3p/CAPN1 axis. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127318. [PMID: 34879549 DOI: 10.1016/j.jhazmat.2021.127318] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/08/2021] [Accepted: 09/19/2021] [Indexed: 06/13/2023]
Abstract
Chlorpyrifos (CPF), a widely used organophosphorus pesticide has caused water pollution, threatening aquatic organisms. MicroRNAs (miRNAs) highly conserved noncoding RNAs, that regulate various cell death processes, including pyroptosis. To investigate the effect of CPF exposure on epithelioma papulosum cyprini (EPC) cell pyroptosis and the role of the miR-124-3p/CAPN1 axis, we established miR-124 overexpression and inhibition EPC cell models of CPF exposure. The target of the miR-124-3p/CAPN1 axis was primarily confirmed by the double luciferase reporter assay. Pyroptosis was demonstrated to occur in CPF-exposed EPC cells and was accompanied by mitochondrial membrane potential depletion, ROS level elevation and pyroptotic indicator expression upregulation. PD150606 was supplied as a CAPN1 inhibitor, alleviating CPF-induced mitochondrial dysfunction, and alleviating the increased expression of NLRP3, CASP1, IL1β and GSDMD. In conclusion, CPF induces pyroptosis by regulating the miR-124-3p/CAPN1 axis. This study enriches the cytotoxicity study of CPF, and provides new theoretical fundamentals for exploration of miRNA and its target protein response to water contaminants.
Collapse
Affiliation(s)
- Zhiying Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Zhiruo Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, People's Republic of China.
| |
Collapse
|
17
|
Wu Y, Yang H, Cheng M, Shi J, Zhang W, Liu S, Zhang M. Calpain Inhibitor Calpeptin Alleviates Ischemia/Reperfusion-Induced Acute Kidney Injury via Suppressing AIM2 Inflammasome and Upregulating Klotho Protein. Front Med (Lausanne) 2022; 9:811980. [PMID: 35155498 PMCID: PMC8831790 DOI: 10.3389/fmed.2022.811980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/06/2022] [Indexed: 11/29/2022] Open
Abstract
Renal ischemia/reperfusion injury is a major contributor of acute kidney injury (AKI), leading to renal cell necrosis, apoptosis, and inflammation. Calpains, a family of Ca2+-dependent cysteine proteases, play a pivotal role in the pathogenesis of renal diseases. Several studies have reported calpain inhibitors showing remarkable reno-protective effects against proteinuria and α-klotho deficiency-induced renal aging symptoms, particularly against glomerulus injury. However, little is known about the role of the calpain inhibitor calpeptin in acute kidney injury. The present study aims to investigate the potential mechanism of downregulation of Calpain 1 and 2 activity by calpeptin in the ischemia/reperfusion (IR)-induced AKI model. Firstly, we observed that the contents of Calpain 1 and 2 were significantly increased in the renal biopsy of clinical AKI patients, especially in the diseased tubules space. To investigate the impacts of calpain activity inhibition, we further pretreated with calpeptin in both the IR mouse model and in the HK-2 cells hypoxia model. We found that the calpain inhibitor calpeptin improved renal functional deterioration, attenuated pathological structure damage, and decreased tubular cell apoptosis in the IR injury-induced AKI mice model. Mechanistically, calpeptin significantly suppressed the AIM2 (absent in melanoma 2) and NLRP3 (NOD-like receptor protein 3) inflammasome signaling pathways and increased Klotho protein levels. Furthermore, immunofluorescence assays demonstrated that the application of calpeptin effectively inhibited Calpain 1 activation and gasdermin D (GSDMD) cleavage in the renal tubules of IR mice. Taken together, our both in vivo and in vitro experiments suggest that calpeptin conveyed reno-protection in AKI might be mediated by the inhibition of AIM2 inflammasome activation and upregulation of Klotho protein. As such, we provide new evidence that Calpain 1 and 2 activation may be closely associated with the pathogenesis of clinical AKI. The calpain-mediated AIM2 inflammasome signaling pathway and distinct interaction between calpain and Klotho may provide a potential novel preventative and therapeutic target for acute kidney injury.
Collapse
|
18
|
Zhang Y, Shi Y. Intracellular Potassium Ion Measurements by Inductively Coupled Plasma Optical Emission Spectrometer (ICP-OES). Methods Mol Biol 2022; 2459:85-92. [PMID: 35212957 DOI: 10.1007/978-1-0716-2144-8_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Potassium ion (K+) efflux is often considered as an upstream signaling event of NLRP3 activation. The main evidence to demonstrate the importance of K+ efflux is that high concentration of extracellular K+ inhibits NLRP3 inflammasome assembly. However, the conditions used to prevent K+ flowing also breaks down a basic parameter of eukaryotic biology, leading to sustained membrane potential depolarization and affecting normal signal transduction in cells. Therefore, direct measurement of intracellular ion concentration can more truly reflect the role of K+ flow during the activation of NLRP3. In this chapter, we will provide the rationale and a method to evaluate intracellular K+ concentration by ICP-OES (Inductively Coupled Plasma Optical Emission Spectroscopy), which helps us understand how disturbances in intracellular K+ level orchestrates NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yifei Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Yan Shi
- Tsinghua Institute for Immunology and Department of Basic Medical Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
- Snyder Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
19
|
The roles of cellular protease interactions in viral infections and programmed cell death: a lesson learned from the SARS-CoV-2 outbreak and COVID-19 pandemic. Pharmacol Rep 2022; 74:1149-1165. [PMID: 35997950 PMCID: PMC9395814 DOI: 10.1007/s43440-022-00394-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022]
Abstract
The unprecedented pandemic of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), which leads to COVID-19, is threatening global health. Over the last 2 years, we have witnessed rapid progress in research focusing on developing new antiviral vaccines and drugs, as well as in academic and clinical efforts to understand the biology and pathology of COVID-19. The roles of proteases among master regulators of SARS-CoV-2 invasion and replication and their pivotal roles in host defence against this pathogen, including programmed cell death, have not been well established. Our understanding of protease function in health and disease has increased considerably over the last two decades, with caspases, matrix metalloproteases, and transmembrane serine proteases representing the most prominent examples. Therefore, during the COVID-19 pandemic, these enzymes have been investigated as potential molecular targets for therapeutic interventions. Proteases that are responsible for SARS-CoV-2 cell entry and replication, such as TMPRSS2, ACE2 or cathepsins, are screened with inhibitor libraries to discover lead structures for further drug design that would prevent virus multiplication. On the other hand, proteases that orchestrate programmed cell death can also be harnessed to enhance the desired demise of infected cells through apoptosis or to attenuate highly inflammatory lytic cell death that leads to undesired cytokine storms, a major hallmark of severe COVID-19. Given the prominent role of proteases in SARS-CoV-2-induced cell death, we discuss the individual roles of these enzymes and their catalytic interactions in the pathology of COVID-19 in this article. We provide a rationale for targeting proteases participating in cell death as potential COVID-19 treatments and identify knowledge gaps that might be investigated to better understand the mechanism underlying SARS-CoV-2-induced cell death.
Collapse
|
20
|
Mitochondrial calpain-1 activates NLRP3 inflammasome by cleaving ATP5A1 and inducing mitochondrial ROS in CVB3-induced myocarditis. Basic Res Cardiol 2022; 117:40. [PMID: 35997820 PMCID: PMC9399059 DOI: 10.1007/s00395-022-00948-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 01/31/2023]
Abstract
Treatment options for myocarditis are currently limited. Inhibition of calpains has been shown to prevent Coxsackievirus B3 (CVB3)-induced cardiac injuries, but the underlying mechanism of action of calpains has not been elucidated. We investigated whether NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome participated in CVB3-induced myocarditis, and investigated the effects of calpain-1 on CVB3-induced cardiac injury. NLRP3 inflammasome was activated in CVB3-infected hearts, evidenced by elevated protein levels of NLRP3, N-terminal domain of Gasdermin D, and cleaved caspase-1, and the increased co-localization of NLRP3 and apoptosis-associated speck-like protein. The intraperitoneal administration of MCC950, a selective inhibitor of the NLRP3 inflammasome, led to decreased levels of serum creatine kinase-MB, cardiac troponin I, lactate dehydrogenase, interleukin-18, interleukin-1β, prevention of the infiltration of inflammatory cells, and improvement of cardiac function under CVB3 infection. Transgenic mice overexpressing the endogenous calpain inhibitor calpastatin (Tg-CAST mice) exhibited not only decreased apoptosis, inflammation, fibrosis, and enhanced cardiac function but also inhibition of NLRP3 inflammasome and pyroptosis. The selective inhibition of calpain-1 using PD151746 protected cardiomyocytes in vitro from CVB3 infection by downregulating NLRP3 inflammasome and, thus, preserved cell viability. Mechanistically, we showed that mitochondrial dysfunction preceded inflammatory response after CVB3 treatment and elimination of mitochondrial reactive oxygen species (ROS) using mitochondria-targeted antioxidants (mito-TEMPO) recapitalized the phenotype observed in Tg-CAST mice. Furthermore, the promotion or inhibition of calpain-1 activation in vitro regulated the mitochondrial respiration chain. Mito-TEMPO reversed calpain-1-mediated NLRP3 inflammation activation and cell death. We also found that mitochondrial calpain-1, which was increased after CVB3 stimulation, activated the NLRP3 inflammasome and resulted in cell death. Furthermore, ATP synthase-α (ATP5A1) was revealed to be the cleaving target of calpain-1 after CVB3 treatment. Downregulating ATP5A1 using ATP5A1-small interfering RNA impaired mitochondrial function, decreased cell viability, and induced NLRP3 inflammasome activation. In conclusion, CVB3 infection induced calpain-1 accumulation in mitochondria, and led to subsequent ATP5A1 cleavage, mitochondrial ROS overproduction, and impaired mitochondrial function, eventually causing NLRP3 inflammasome activation and inducing pyroptosis. Therefore, our findings established the role of calpain in viral myocarditis and unveiled its underlying mechanism of its action. Calpain appears as a promising target for the treatment of viral myocarditis.
Collapse
|
21
|
Inflammasome activation in neurodegenerative diseases. Essays Biochem 2021; 65:885-904. [PMID: 34846519 DOI: 10.1042/ebc20210021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/08/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022]
Abstract
Approximately ten million people are diagnosed with dementia annually since they experience difficulties with memory and thinking skills. Since neurodegenerative diseases are diagnosed late, most of them are difficult to treat. This is due to the increased severity of the disease during the progression when neuroinflammation plays a critical role. The activation of immune cells, especially microglia, plays a crucial role in the development of neurodegenerative diseases. Molecular sensors within these microglia, such as the NLRP3 inflammasome, are activated by signals that represent the hallmarks of neurodegenerative diseases. Here, we first summarize the two activation steps of NLRP3 inflammasome activation. Furthermore, we discuss the key factors that contribute to NLRP3 inflammasome activation in the different neuroinflammatory diseases, like Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). The prominent NLRP3 inflammasome triggers include amyloid β and tau oligomers in AD, α-synuclein in PD, and superoxide dismutase (SOD1) and TAR DNA-binding protein 43 (TDP43) in ALS. NLRP3 inhibitor treatment has shown promising results in several preclinical mouse models of AD, PD, and ALS. Finally, we postulate that current understandings underpin the potential for NLRP3 inhibitors as a therapeutic target in neurodegenerative diseases.
Collapse
|
22
|
Arabiyat AS, Chen H, Erndt-Marino J, Burkhard K, Scola L, Fleck A, Wan LQ, Hahn MS. Hyperosmolar Ionic Solutions Modulate Inflammatory Phenotype and sGAG Loss in a Cartilage Explant Model. Cartilage 2021; 13:713S-721S. [PMID: 32975437 PMCID: PMC8804856 DOI: 10.1177/1947603520961167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The objective of this study was to compare the effects of hyperosmolar sodium (Na+), lithium (Li+) and potassium (K+) on catabolic and inflammatory osteoarthritis (OA) markers and sulfated glycosaminoglycan (sGAG) loss in TNF-α-stimulated cartilage explants. METHODS Explants from bovine stifle joints were stimulated with TNF-α for 1 day to induce cartilage degradation followed by supplementation with 50 mM potassium chloride (KCl), 50 mM lithium chloride (LiCl), 50 mM sodium chloride (NaCl), or 100 nM dexamethasone for an additional 6 days. We assessed the effect of TNF-α stimulation and hyperosmolar ionic treatment on sGAG loss and expression of OA-associated proteins: ADAMTS-5, COX-2, MMP-1, MMP-13, and VEGF. RESULTS TNF-α treatment increased sGAG loss (P < 0.001) and expression of COX-2 (P = 0.018), MMP-13 (P < 0.001), and VEGF (P = 0.017) relative to unstimulated controls. Relative to activated controls, LiCl and dexamethasone treatment attenuated sGAG loss (P = 0.008 and P = 0.042, respectively) and expression of MMP-13 (P = 0.005 and P = 0.036, respectively). In contrast, KCl treatment exacerbated sGAG loss (P = 0.032) and MMP-1 protein expression (P = 0.010). NaCl treatment, however, did not alter sGAG loss or expression of OA-related proteins. Comparing LiCl and KCl treatment shows a potent reduction (P < 0.05) in catabolic and inflammatory mediators following LiCl treatment. CONCLUSION These results suggest that these ionic species elicit varying responses in TNF-α-stimulated explants. Cumulatively, these findings support additional studies of hyperosmolar ionic solutions for potential development of novel intraarticular injections targeting OA.
Collapse
Affiliation(s)
- Ahmad S. Arabiyat
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Hongyu Chen
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Josh Erndt-Marino
- Department of Biomedical Engineering,
Tufts University, Medford, MA, USA
| | - Katie Burkhard
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Lisa Scola
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Allison Fleck
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Leo Q. Wan
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| |
Collapse
|
23
|
Garrosa-Jiménez J, Sánchez Carro Y, Ovejero-Benito MC, Del Sastre E, García AG, López MG, López-García P, Cano-Abad MF. Intracellular calcium and inflammatory markers, mediated by purinergic stimulation, are differentially regulated in monocytes of patients with major depressive disorder. Neurosci Lett 2021; 765:136275. [PMID: 34606909 DOI: 10.1016/j.neulet.2021.136275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/06/2021] [Accepted: 09/25/2021] [Indexed: 11/24/2022]
Abstract
The P2X7 receptor (P2X7R) is a ligand-gated ion channel that is being recognized as a major player in neuropsychiatric disorders such as Major Depressive Disorder (MDD). P2X7R activation is triggered by high extracellular ATP concentrations, leading to channel opening and inducing an increase in cytosolic calcium concentration ([Ca2+]c), that activates the inflammatory pathway. Those receptors are expressed not only in CNS cells but also in peripheral blood cells, where they are activated in response to inflammatory molecules such as bacterial lipopolysaccharide (LPS). LPS induced-tissue damage promotes an elevation of extracellular ATP, triggering the NRLP3-inflammasome assembly and activation that, sequentially, induces caspase-1 cleavage and IL-1β processing and secretion. In this context, we attempt to understand the role of P2X7R in [Ca2+]c homeostasis regulation, inflammasome expression and its pharmacological modulation in MDD. For this purpose, monocytes were isolated from peripheral blood of MDD patients and [Ca2+]c was monitored with the intracellular probe Fura-2. Our results point out to P2X7R as the responsible of the Ca2+ imbalance, as well as TNF-α-dependent activation of caspase-1 in MDD patients. In addition, P2X7R blockade with its specific antagonist, JNJ-47965567, reduces the Ca2+ entry upon Bz-ATP exposure. Altogether, our results point that MDD patients have both, Ca2+ homeostasis alteration and an inflammatory status, which promote an independent-inflammasome activation of caspase-1. Therefore, we propose the pharmacological modulation of P2X7R as a therapeutic approach against MDD symptoms.
Collapse
Affiliation(s)
- Javier Garrosa-Jiménez
- Instituto Teófilo Hernando de I+D del Medicamento, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, (IIS-IP)., Madrid, Spain
| | - Yolanda Sánchez Carro
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain; Department of Psychiatry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - María C Ovejero-Benito
- Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, (IIS-IP)., Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Eric Del Sastre
- Instituto Teófilo Hernando de I+D del Medicamento, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio G García
- Instituto Teófilo Hernando de I+D del Medicamento, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando de I+D del Medicamento, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, (IIS-IP)., Madrid, Spain
| | - Pilar López-García
- Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, (IIS-IP)., Madrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Spain
| | - María F Cano-Abad
- Instituto Teófilo Hernando de I+D del Medicamento, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, (IIS-IP)., Madrid, Spain.
| |
Collapse
|
24
|
Song Z, Bian Z, Zhang Z, Wang X, Zhu A, Zhu G. Astrocytic Kir4.1 regulates NMDAR/calpain signaling axis in lipopolysaccharide-induced depression-like behaviors in mice. Toxicol Appl Pharmacol 2021; 429:115711. [PMID: 34474083 DOI: 10.1016/j.taap.2021.115711] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 12/16/2022]
Abstract
The activation of Nod-like receptor protein 3 (NLRP3) inflammasome propagates pro-inflammatory signaling cascades linking to depression-like behaviors. However, the signaling pathway contributing to NLRP3 inflammasome activation and depression-like behaviors is still not clear. In this study, we evidenced that lipopolysaccharide (LPS) injection (i.p.) triggered depression-like behaviors, promoted the expression of Kir4.1, p-GluN2B and calpain-1, and activated NLRP3 inflammasome. The blockage of N-methyl-d-aspartate receptors (NMDAR) by memantine reduced LPS-induced depression-like behaviors, NLRP3 inflammasome and astrocyte activation, and calpain-1 expression. Additionally, memantine also inhibited LPS-induced reduction of postsynaptic density protein 95 (PSD-95) and Arc expression. Specific reduction of Kir4.1 in astrocytes attenuated LPS-induced expression of NLRP3 and calpain-1, and phosphorylation of GluN2B. Interestingly, LPS-induced expression of calpain-1 largely co-localized with GFAP, indicating the specific function of calpain-1 in astrocytes. Together, these data indicate that astrocytic Kir4.1 could regulate NMDAR/calpain-1 signaling axis, contributing to depression-like behaviors, likely through regulating NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Zhujin Song
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China; School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhijuan Bian
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Zhengrong Zhang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xuncui Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Aisong Zhu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China.
| |
Collapse
|
25
|
Bai H, Zhang Q. Activation of NLRP3 Inflammasome and Onset of Alzheimer's Disease. Front Immunol 2021; 12:701282. [PMID: 34381452 PMCID: PMC8350495 DOI: 10.3389/fimmu.2021.701282] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
The nucleotide-binding domain leucine-rich repeat and pyrin domain containing receptor protein 3 (NLRP3) is an important pattern recognition receptor in human innate immunity. Activation of the NLRP3 inflammasome play a key role in the pathogenesis of Alzheimer’s disease (AD). Theories explaining activation of the NLRP3 inflammasome include the reactive oxygen species theory, the lysosomal damage theory and the mitochondrial DNA theory. The NLRP3 activation promotes occurrence of AD by producing IL-1β, IL-18 and other cytokines, and then by affecting the deposition of Aβ and tau proteins. Over-activated NLRP3 inflammasome often impair cell function and induces immune-related diseases. Some mechanisms have been found to negatively regulate activation of the NLRP3 inflammasome, which may be through receptor binding blocking mechanism, autophagy related mechanism, abnormal cytokine secretion mechanism, or interference related gene expression regulation mechanism. In this review, we summarize the possible mechanisms by which the activation of NLRP3 inflammasomes affects the pathogenesis of AD, and the recent advances in the prevention and treatment of AD by controlling the activation of NLRP3 inflammasomes. By researching the activation or inactivation of NLRP3 inflammasome, it is possible to reveal the pathogenesis of AD from a new perspective and provide a new idea for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Hua Bai
- Department of Neurology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, China.,Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Medical Experimental Center of the Third Affiliated Hospital of Guizhou Medical University, Duyun, China
| | - Qifang Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China.,Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
26
|
Popov SV, Maslov LN, Naryzhnaya NV, Mukhomezyanov AV, Krylatov AV, Tsibulnikov SY, Ryabov VV, Cohen MV, Downey JM. The Role of Pyroptosis in Ischemic and Reperfusion Injury of the Heart. J Cardiovasc Pharmacol Ther 2021; 26:562-574. [PMID: 34264787 DOI: 10.1177/10742484211027405] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
While ischemia itself can kill heart muscle, much of the infarction after a transient period of coronary artery occlusion has been found to result from injury during reperfusion. Here we review the role of inflammation and possible pyroptosis in myocardial reperfusion injury. Current evidence suggests pyroptosis's contribution to infarction may be considerable. Pyroptosis occurs when inflammasomes activate caspases that in turn cleave off an N-terminal fragment of gasdermin D. This active fragment makes large pores in the cell membrane thus killing the cell. Inhibition of inflammation enhances cardiac tolerance to ischemia and reperfusion injury. Stimulation of the purinergic P2X7 receptor and the β-adrenergic receptor and activation of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) by toll-like receptor (TLR) agonists are all known to contribute to ischemia/reperfusion (I/R) cardiac injury through inflammation, potentially by pyroptosis. In contrast, stimulation of the cannabinoid CB2 receptor reduces I/R cardiac injury and inhibits this pathway. MicroRNAs, Akt, the phosphate and tension homology deleted on chromosome 10 protein (PTEN), pyruvate dehydrogenase and sirtuin-1 reportedly modulate inflammation in cardiomyocytes during I/R. Cryopyrin and caspase-1/4 inhibitors are reported to increase cardiac tolerance to ischemic and reperfusion cardiac injury, presumably by suppressing inflammasome-dependent inflammation. The ambiguity surrounding the role of pyroptosis in reperfusion injury arises because caspase-1 also activates cytotoxic interleukins and proteolytically degrades a surprisingly large number of cytosolic enzymes in addition to activating gasdermin D.
Collapse
Affiliation(s)
- Sergey V Popov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Leonid N Maslov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Natalia V Naryzhnaya
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Alexandr V Mukhomezyanov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Andrey V Krylatov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Sergey Y Tsibulnikov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Vyacheslav V Ryabov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Michael V Cohen
- Department of Physiology and Cell Biology, 12214University of South Alabama College of Medicine, Mobile, AL, USA
| | - James M Downey
- Department of Physiology and Cell Biology, 12214University of South Alabama College of Medicine, Mobile, AL, USA
| |
Collapse
|
27
|
P2X7 receptor in multifaceted cellular signalling and its relevance as a potential therapeutic target in different diseases. Eur J Pharmacol 2021; 906:174235. [PMID: 34097884 DOI: 10.1016/j.ejphar.2021.174235] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023]
Abstract
P2X7 receptor, a purinergic receptor family member, is abundantly expressed on many cells, including immune, muscle, bone, neuron, and glia. It acts as an ATP-activated cation channel that permits the influx of Ca2+, Na+ and efflux of K+ ions. The P2X7 receptor plays crucial roles in many physiological processes including cytokine and chemokine secretion, NLRP3 inflammasome activation, cellular growth and differentiation, locomotion, wound healing, transcription factors activation, cell death and T-lymphocyte survival. Past studies have demonstrated the up-regulation and direct association of this receptor in many pathophysiological conditions such as cancer, diabetics, arthritis, tuberculosis (TB) and inflammatory diseases. Hence, targeting this receptor is considered a worthwhile approach to lessen the afflictions associated with the disorders mentioned above by understanding the receptor architecture and downstream signalling processes. Here, in the present review, we have dissected the structural and functional aspects of the P2X7 receptor, emphasizing its role in various diseased conditions. This information will provide in-depth knowledge about the receptor and help to develop apt curative methodologies for the betterment of humanity in the coming years.
Collapse
|
28
|
An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol Immunol 2021; 18:1141-1160. [PMID: 33850310 PMCID: PMC8093260 DOI: 10.1038/s41423-021-00670-3] [Citation(s) in RCA: 396] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/25/2021] [Indexed: 02/08/2023] Open
Abstract
The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a multiprotein complex involved in the release of mature interleukin-1β and triggering of pyroptosis, which is of paramount importance in a variety of physiological and pathological conditions. Over the past decade, considerable advances have been made in elucidating the molecular mechanisms underlying the priming/licensing (Signal 1) and assembly (Signal 2) involved in NLRP3 inflammasome activation. Recently, a number of studies have indicated that the priming/licensing step is regulated by complicated mechanisms at both the transcriptional and posttranslational levels. In this review, we discuss the current understanding of the mechanistic details of NLRP3 inflammasome activation with a particular emphasis on protein-protein interactions, posttranslational modifications, and spatiotemporal regulation of the NLRP3 inflammasome machinery. We also present a detailed summary of multiple positive and/or negative regulatory pathways providing upstream signals that culminate in NLRP3 inflammasome complex assembly. A better understanding of the molecular mechanisms underlying NLRP3 inflammasome activation will provide opportunities for the development of methods for the prevention and treatment of NLRP3 inflammasome-related diseases.
Collapse
|
29
|
Sun Y, Lu M, Sun T, Wang H. Astragaloside IV attenuates inflammatory response mediated by NLRP-3/calpain-1 is involved in the development of pulmonary hypertension. J Cell Mol Med 2021; 25:586-590. [PMID: 33295020 PMCID: PMC7810938 DOI: 10.1111/jcmm.15671] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/16/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022] Open
Abstract
Inflammation eventually leads to pulmonary arterial hypertension (PAH). Astragaloside IV(AS-IV) has a protective effect on pulmonary hypertension, but the specific protective mechanism has been unclear until now. Therefore, in this study, our aim was to investigate the mechanisms underlying the effects of AS-IV on PAH. In vivo, male Sprague-Dawley (SD) rats were injected intraperitoneally with monocrotaline (MCT, 60 mg/kg) and treated with AS-IV (40 mg/kg, 80 mg/kg), MCC950 and MDL-28170. In vitro, human pulmonary artery endothelial cells (HPAECs) were treated with monocrotaline pyrrole (MCTP, 60 μg/mL). The protein expression levels of NLRP-3, caspase-1, ASC, IL-18, IL-1β and calpain-1 were measured in vivo and/or in vitro. The results showed that AS-IV decreased the protein expression levels of NLRP-3, caspase-1, ASC, IL-18, IL-1β and calpain-1 in vivo and/or vitro. In conclusion, in this study the results suggested that AS-IV could inhibit monocrotaline-induced pulmonary arterial hypertension via the NLRP-3/calpain-1 pathway.
Collapse
Affiliation(s)
- Yang Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning ProvinceJinzhou Medical UniversityJinzhouChina
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning ProvinceJinzhou Medical UniversityJinzhouChina
| | - Tairan Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning ProvinceJinzhou Medical UniversityJinzhouChina
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning ProvinceJinzhou Medical UniversityJinzhouChina
| |
Collapse
|
30
|
He Z, Davidson SM, Yellon DM. The importance of clinically relevant background therapy in cardioprotective studies. Basic Res Cardiol 2020; 115:69. [PMID: 33188438 PMCID: PMC7666584 DOI: 10.1007/s00395-020-00830-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022]
Abstract
Treatment of acute myocardial infarct patients (AMI) includes rapid restoration of coronary blood flow and pharmacological therapy aimed to prevent pain and maintain vessel patency. Many interventions have been investigated to offer additional protection. One such intervention is remote ischaemic conditioning (RIC) involving short-episodes of ischaemia of the arm with a blood pressure cuff, followed by reperfusion to protect the heart organs from subsequent severe ischaemia. However, the recent CONDI2-ERIC-PPCI multicentre study of RIC in STEMI showed no benefit in clinical outcome in low risk patients. It could also be argued that these patients were already in a partially protected state, highlighting the disconnect between animal- and clinical-based outcome studies. To improve potential translatability, we developed an animal model using pharmacological agents similar to those given to patients presenting with an AMI, prior to PPCI. Rats underwent MI on a combined background of an opioid agonist, heparin and a platelet-inhibitor thereby allowing us to assess whether additional cardioprotective strategies had any effect over and above this “cocktail”. We demonstrated that the “background drugs” were protective in their own right, reducing MI from 57.5 ± 3.7% to 37.3 ± 2.9% (n = 11, p < 0.001). On this background of drugs, RIC did not add any further protection (38.0 ± 3.4%). However, using a caspase inhibitor, which acts via a different mechanistic pathway to RIC, we were able to demonstrate additional protection (20.6 ± 3.3%). This concept provides initial evidence to develop models which can be used to evaluate future animal-to-clinical translation in cardioprotective studies.
Collapse
Affiliation(s)
- Zhenhe He
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
31
|
Rocha M, Apostolova N, Diaz-Rua R, Muntane J, Victor VM. Mitochondria and T2D: Role of Autophagy, ER Stress, and Inflammasome. Trends Endocrinol Metab 2020; 31:725-741. [PMID: 32265079 DOI: 10.1016/j.tem.2020.03.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/08/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes (T2D) is one of the main current threats to human health. Both T2D and its numerous clinical complications are related to mitochondrial dysfunction and oxidative stress. Over the past decade, great progress has been made in extending our knowledge about the signaling events regulated by mitochondria. However, the links among mitochondrial impairment, oxidative stress, autophagy, endoplasmic reticulum (ER) stress, and activation of the inflammasome still need to be clarified. In light of this deficit, we aim to provide a review of the existing literature concerning the complicated crosstalk between mitochondrial impairment, autophagy, ER stress, and the inflammasome in the molecular pathogenesis of T2D.
Collapse
Affiliation(s)
- Milagros Rocha
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| | | | - Ruben Diaz-Rua
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Jordi Muntane
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Biomedicine of Seville (IBiS), University Hospital 'Virgen del Rocío'/CSIC/University of Seville, Seville, Spain; Department of General Surgery, University Hospital 'Virgen del Rocío'/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - Victor M Victor
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Physiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
32
|
Activation of the NLRP3 Inflammasome by Particles from the Echinococcus granulosus Laminated Layer. Infect Immun 2020; 88:IAI.00190-20. [PMID: 32571988 PMCID: PMC7440765 DOI: 10.1128/iai.00190-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022] Open
Abstract
The interaction of dendritic cells and macrophages with a variety of rigid noncellular particles triggers activation of the NLRP3 inflammasome and consequent secretion of interleukin 1β (IL-1β). Noncellular particles can also be generated in the context of helminth infection, since these large pathogens often shed their outermost structures during growth and/or molting. One such structure is the massive, mucin-based, soft, flexible laminated layer (LL), which protects the larval stages of cestodes of the genus Echinococcus. The interaction of dendritic cells and macrophages with a variety of rigid noncellular particles triggers activation of the NLRP3 inflammasome and consequent secretion of interleukin 1β (IL-1β). Noncellular particles can also be generated in the context of helminth infection, since these large pathogens often shed their outermost structures during growth and/or molting. One such structure is the massive, mucin-based, soft, flexible laminated layer (LL), which protects the larval stages of cestodes of the genus Echinococcus. We show that particles from the Echinococcus granulosus LL (pLL) trigger NLRP3- and caspase-1-dependent IL-1β in lipopolysaccharide (LPS)-primed mouse bone marrow-derived dendritic cells (BMDC). This response can be elicited by pLL too large for phagocytosis and nonetheless requires actin dynamics, Syk, and phosphatidylinositol 3-kinase (PI3K). These three requirements had already been observed in our previous study on the alteration by pLL of CD86, CD40, IL-10, and IL-12 responses to LPS in BMDC; however, we now show that these alterations are independent of NLRP3 and caspase-1. In other words, an initial interaction with particles requiring actin dynamics, Syk, and PI3K, but not phagocytosis, elicits both NLRP3-dependent and NLRP3-independent responses. Intraperitoneal injection of pLL induced IL-1β, suggesting that contact with LL materials induces IL-1β in the E. granulosus infection setting. Our results extend our understanding of NLRP3 inflammasome activation by noncellular particulate materials both to helminth-derived materials and to flexible/soft materials.
Collapse
|
33
|
Abstract
Acute occlusion of a coronary artery can result in myocardial infarction-a leading cause of premature death. Prompt restoration of blood flow to the myocardium can prevent excessive death of cardiomyocytes and improve clinical outcome. Although the major mechanism of cell death after reperfusion is necrosis, it is now recognized that many other cell death pathways may be involved in ischemia-reperfusion (I/R) injury. Pyroptosis is one such cell death pathway that is caspase-1-dependent and induced in response to cellular insult. The activated caspase-1 protease cleaves and activates specific cellular targets including gasdermin D and the proinflammatory cytokines interleukin-1β and interleukin-18. The N-terminal fragment of gasdermin D forms plasma membrane pores resulting in cytosolic leakage and cell rupture, releasing interleukin-1β and interleukin-18. Evidence suggests that inflammation induced by I/R through the pyroptotic pathway contributes to cardiomyocyte death, excessive scar formation, and poor ventricular remodeling. For this reason, there is growing interest in targeting components of the pyroptotic pathway as a means of reducing I/R injury.
Collapse
|
34
|
Zuurbier CJ, Abbate A, Cabrera-Fuentes HA, Cohen MV, Collino M, De Kleijn DPV, Downey JM, Pagliaro P, Preissner KT, Takahashi M, Davidson SM. Innate immunity as a target for acute cardioprotection. Cardiovasc Res 2020; 115:1131-1142. [PMID: 30576455 DOI: 10.1093/cvr/cvy304] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/07/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Acute obstruction of a coronary artery causes myocardial ischaemia and if prolonged, may result in an ST-segment elevation myocardial infarction (STEMI). First-line treatment involves rapid reperfusion. However, a highly dynamic and co-ordinated inflammatory response is rapidly mounted to repair and remove the injured cells which, paradoxically, can further exacerbate myocardial injury. Furthermore, although cardiac remodelling may initially preserve some function to the heart, it can lead over time to adverse remodelling and eventually heart failure. Since the size of the infarct corresponds to the subsequent risk of developing heart failure, it is important to find ways to limit initial infarct development. In this review, we focus on the role of the innate immune system in the acute response to ischaemia-reperfusion (IR) and specifically its contribution to cell death and myocardial infarction. Numerous danger-associated molecular patterns are released from dying cells in the myocardium, which can stimulate pattern recognition receptors including toll like receptors and NOD-like receptors (NLRs) in resident cardiac and immune cells. Activation of the NLRP3 inflammasome, caspase 1, and pyroptosis may ensue, particularly when the myocardium has been previously aggravated by the presence of comorbidities. Evidence will be discussed that suggests agents targeting innate immunity may be a promising means of protecting the hearts of STEMI patients against acute IR injury. However, the dosing and timing of such agents should be carefully determined because innate immunity pathways may also be involved in cardioprotection. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Antonio Abbate
- VCU Pauley Heart Center and Wright Center for Clinical and Translational Research, Richmond, VA, USA
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Centro de Biotecnología-FEMSA, Monterrey, Nuevo León, México.,Department of Microbiology, Kazan Federal University, Kazan, Russian Federation.,Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Michael V Cohen
- Department of Medicine, University of South Alabama College of Medicine, Mobile, AL, USA.,Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Torino, Italy
| | - Dominique P V De Kleijn
- Department of Vascular Surgery, UMC Utrecht, Utrecht University, Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands
| | - James M Downey
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Pasquale Pagliaro
- Department of Biological and Clinical Sciences, University of Turin, Torino, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| | - Klaus T Preissner
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, UK
| |
Collapse
|
35
|
Erndt-Marino J, Yeisley DJ, Chen H, Levin M, Kaplan DL, Hahn MS. Interferon-Gamma Stimulated Murine Macrophages In Vitro: Impact of Ionic Composition and Osmolarity and Therapeutic Implications. Bioelectricity 2020; 2:48-58. [PMID: 32292895 PMCID: PMC7107958 DOI: 10.1089/bioe.2019.0032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Injections of osmolytes are promising immunomodulatory treatments for medical benefit, although the rationale and underlying mechanisms are often lacking. The goals of the present study were twofold: (1) to clarify the anti-inflammatory role of the potassium ion and (2) to begin to decouple the effects that ionic strength, ionic species, and osmolarity have on macrophage biology. Materials and Methods: RAW 264.7 murine macrophages were encapsulated in three-dimensional, poly(ethylene glycol) diacrylate hydrogels and activated with interferon-gamma to yield M(IFN). Gene and protein profiles were made of M(IFN) exposed to different hyperosmolar treatments (80 mM potassium gluconate, 80 mM sodium gluconate, and 160 mM sucrose). Results: Relative to M(IFN), all hyperosmolar treatments suppressed expression of pro-inflammatory markers (nitric oxide synthase-2 [NOS-2], tumor necrosis factor-alpha, monocyte chemoattractant protein-1 [MCP-1]) and increased messenger RNA (mRNA) expression of the pleiotropic and angiogenic markers interleukin-6 (IL-6) and vascular endothelial growth factor-A (VEGF), respectively. Ionic osmolytes also demonstrated a greater level of change compared to the nonionic treatments, with mRNA levels of IL-6 the most significantly affected. M(IFN) exposed to K+ exhibited the lowest levels of NOS-2 and MCP-1, and this ion limited IL-6 release induced by osmolarity. Conclusion: Cumulatively, these data suggest that osmolyte composition, ionic strength, and osmolarity are all parameters that can influence therapeutic outcomes. Future work is necessary to further decouple and mechanistically understand the influence that these biophysical parameters have on cell biology, including their impact on other macrophage functions, intracellular osmolyte composition, and cellular and organellular membrane potentials.
Collapse
Affiliation(s)
- Joshua Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Department of Biology, Allen Discovery Center at Tufts University, Tufts University, Medford, Massachusetts
| | - Daniel J. Yeisley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Hongyu Chen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Michael Levin
- Department of Biology, Allen Discovery Center at Tufts University, Tufts University, Medford, Massachusetts
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Department of Biology, Allen Discovery Center at Tufts University, Tufts University, Medford, Massachusetts
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
36
|
Wierenga KA, Wee J, Gilley KN, Rajasinghe LD, Bates MA, Gavrilin MA, Holian A, Pestka JJ. Docosahexaenoic Acid Suppresses Silica-Induced Inflammasome Activation and IL-1 Cytokine Release by Interfering With Priming Signal. Front Immunol 2019; 10:2130. [PMID: 31616405 PMCID: PMC6763728 DOI: 10.3389/fimmu.2019.02130] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Occupational exposure to respirable crystalline silica (cSiO2) has been etiologically linked to human autoimmunity. Intranasal instillation with cSiO2 triggers profuse inflammation in the lung and onset of autoimmunity in lupus-prone mice; however, dietary supplementation with the omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) abrogates these responses. Inflammasome activation, IL-1 cytokine release, and death in alveolar macrophages following cSiO2 exposure are early and critical events that likely contribute to triggering premature autoimmune pathogenesis by this particle. Here we tested the hypothesis that DHA suppresses cSiO2-induced NLRP3 inflammasome activation, IL-1 cytokine release, and cell death in the macrophage. The model used was the murine macrophage RAW 264.7 cell line stably transfected with the inflammasome adapter protein ASC (RAW-ASC). Following priming with LPS, both the canonical activator nigericin and cSiO2 elicited robust inflammasome activation in RAW-ASC cells, as reflected by IL-1β release and caspase-1 activation. These responses were greatly diminished or absent in wild-type RAW cells. In contrast to IL-1β, cSiO2 induced IL-1α release in both RAW-ASC and to a lesser extent in RAW-WT cells after LPS priming. cSiO2-driven effects in RAW-ASC cells were confirmed in bone-marrow derived macrophages. Pre-incubating RAW-ASC cells with 10 and 25 μM DHA for 24 h enriched this fatty acid in the phospholipids by 15- and 25-fold, respectively, at the expense of oleic acid. DHA pre-incubation suppressed inflammasome activation and release of IL-1β and IL-1α by nigericin, cSiO2, and two other crystals - monosodium urate and alum. DHA's suppressive effects were linked to inhibition of LPS-induced Nlrp3, Il1b, and Il1a transcription, potentially through the activation of PPARγ. Finally, nigericin-induced death was inflammasome-dependent, indicative of pyroptosis, and could be inhibited by DHA pretreatment. In contrast, cSiO2-induced death was inflammasome-independent and not inhibited by DHA. Taken together, these findings indicate that DHA suppresses cSiO2-induced inflammasome activation and IL-1 cytokine release in macrophages by acting at the level of priming, but was not protective against cSiO2-induced cell death.
Collapse
Affiliation(s)
- Kathryn A Wierenga
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Josephine Wee
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Kristen N Gilley
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Lichchavi D Rajasinghe
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Melissa A Bates
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Mikhail A Gavrilin
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus, OH, United States
| | - Andrij Holian
- Department of Biomedical and Pharmaceutical Sciences, Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States
| | - James J Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
37
|
Mishra PK, Adameova A, Hill JA, Baines CP, Kang PM, Downey JM, Narula J, Takahashi M, Abbate A, Piristine HC, Kar S, Su S, Higa JK, Kawasaki NK, Matsui T. Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol 2019; 317:H891-H922. [PMID: 31418596 DOI: 10.1152/ajpheart.00259.2019] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell death is a fundamental process in cardiac pathologies. Recent studies have revealed multiple forms of cell death, and several of them have been demonstrated to underlie adverse cardiac remodeling and heart failure. With the expansion in the area of myocardial cell death and increasing concerns over rigor and reproducibility, it is important and timely to set a guideline for the best practices of evaluating myocardial cell death. There are six major forms of regulated cell death observed in cardiac pathologies, namely apoptosis, necroptosis, mitochondrial-mediated necrosis, pyroptosis, ferroptosis, and autophagic cell death. In this article, we describe the best methods to identify, measure, and evaluate these modes of myocardial cell death. In addition, we discuss the limitations of currently practiced myocardial cell death mechanisms.
Collapse
Affiliation(s)
- Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University of Bratislava, Bratislava, Slovakia
| | - Joseph A Hill
- Departments of Medicine (Cardiology) and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christopher P Baines
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Peter M Kang
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - James M Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - Masafumi Takahashi
- Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Antonio Abbate
- Virginia Commonwealth University, Pauley Heart Center, Richmond, Virginia
| | - Hande C Piristine
- Department of Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sumit Kar
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Shi Su
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jason K Higa
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Nicholas K Kawasaki
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Takashi Matsui
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| |
Collapse
|
38
|
Yin W, Zhou QL, OuYang SX, Chen Y, Gong YT, Liang YM. Uric acid regulates NLRP3/IL-1β signaling pathway and further induces vascular endothelial cells injury in early CKD through ROS activation and K + efflux. BMC Nephrol 2019; 20:319. [PMID: 31412804 PMCID: PMC6694569 DOI: 10.1186/s12882-019-1506-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) has been considered as a major health problem in the world. Increasing uric acid (UA) could induce vascular endothelial injury, which is closely related to microinflammation, oxidative stress, and disorders of lipids metabolism. However, the specific mechanism that UA induces vascular endothelial cells injury in early CKD remains unknown. METHODS Human umbilical vein endothelial cells (HUVECs) were cultured and subjected to different concentrations of UA for different periods. Early CKD rat model with elevated serum UA was established. Western blotting and quantitative real-time PCR (qPCR) were applied for measuring protein and mRNA expression of different cytokines. The animals were sacrificed and blood samples were collected for measurement of creatinine, UA, IL-1β, TNF-α, and ICAM-1. Renal tissues were pathologically examined by periodic acid-Schiff (PAS) or hematoxylin-eosin (HE) staining. RESULTS The expression of IL-1β, ICAM-1, NLRP3 complexes, and activation of NLRP3 inflammasome could be induced by UA, but the changes induced by UA were partially reversed by siRNA NLRP3 or caspase 1 inhibitor. Furthermore, we identified that UA regulated the activation of NLRP3 inflammasome by activating ROS and K+ efflux. In vivo results showed that UA caused the vascular endothelial injury by activating NLRP3/IL-1β pathway. While allopurinol could reduce UA level and may have protective effects on cardiovascular system. CONCLUSIONS UA could regulate NLRP3/IL-1β signaling pathway through ROS activation and K+ efflux and further induce vascular endothelial cells injury in early stages of CKD.
Collapse
Affiliation(s)
- Wei Yin
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Qiao-Ling Zhou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Sha-Xi OuYang
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Ying Chen
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Yu-Ting Gong
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Yu-Mei Liang
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China.
| |
Collapse
|
39
|
Campden RI, Zhang Y. The role of lysosomal cysteine cathepsins in NLRP3 inflammasome activation. Arch Biochem Biophys 2019; 670:32-42. [PMID: 30807742 DOI: 10.1016/j.abb.2019.02.015] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/22/2019] [Accepted: 02/19/2019] [Indexed: 12/17/2022]
Abstract
Lysosomal cysteine cathepsins are a family of proteases that are involved in a myriad of cellular processes from proteolytic degradation in the lysosome to bone resorption. These proteins mature following the cleavage of a pro-domain in the lysosome to become either exo- or endo-peptidases. The cathepsins B, C, L, S and Z have been implicated in NLRP3 inflammasome activation following their activation with ATP, monosodium urate, silica crystals, or bacterial components, among others. These five cathepsins have both compensatory and independent functions in NLRP3 inflammasome activation. There is much evidence in the literature to support the release of cathepsin B following lysosomal membrane degradation which leads to NLRP3 inflammasome activation. This is likely due to a hitherto unidentified role of this protein in the cytoplasm, although other interactions with autophagy proteins and within lysosomes have been proposed. Cathepsin C is involved in the processing of neutrophil IL-1β through processing of upstream proteases. Cathepsin Z is non-redundantly required for NLRP3 inflammasome activation following nigericin, ATP and monosodium urate activation. Lysosomal cysteine cathepsins are members of a diverse and complementary family, and likely share both overlapping and independent functions in NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Rhiannon I Campden
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Yifei Zhang
- Institute for Immunology, Department of Basic Medical Sciences, School of Medicine, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|