1
|
Ali MA, Khalifa AA, Elblehi SS, Elsokkary NH, El-Mas MM. Effects of remote ischemic preconditioning and/or erythropoietin on lung injury induced by skeletal ischemia reperfusion: role of the NLRP3 inflammasome. Inflamm Res 2025; 74:67. [PMID: 40272513 DOI: 10.1007/s00011-025-02033-4] [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: 11/23/2024] [Revised: 03/20/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Remote ischemic preconditioning (RIPC) diminishes multi-organ failure induced by skeletal muscle ischemia and reperfusion (S-I/R). The current study investigated whether skeletal RIPC protection against S-I/R-induced acute lung injury (ALI) could be facilitated following simultaneous exposure to the glycoprotein hormone erythropoietin (EPO) in rats and whether this interaction is modulated by the NLRP3 inflammasome. METHODS S-I/R challenge was performed by 3-h ischemia followed by 3-h reperfusion of the right hindlimb, whereas RIPC involved three 20-min brief consecutive I/R cycles of the contralateral hindlimb. RESULTS The lung injurious response to S-I/R was verified by: (i) decreases in minute respiratory volume (MRV), forced expiratory volume 1 (FEV1) and functional vital capacity (FVC), (ii) increases in respiratory rate (RR), (iii) falls in lung surfactant protein-D (SP-D) and rises in of lung plasminogen activator inhibitor-1 (PAI-1) and intercellular adhesion molecule-1 (ICAM-1), and (iv) disruption of alveolar architecture. These lung defects were partially amended by RIPC or EPO (500 or 5000 IU/kg). Further, the prior exposure to RIPC plus EPO-500 was more effective than separate interventions in rectifying ALI damages. Molecularly, the dual RIPC/EPO-500 regimen was also more effective in reversing the S-I/R-associated increments in pulmonary expressions of NLRP3 and related inflammatory (TLR4, MyD88, TRAF, NF-κB, TNF-α, IL-1β, and IL-18), apoptotic (ASC, procaspse-1, caspase-1), and microRNA signals (increases in miR-21 and decreases miR-495). CONCLUSION These findings suggest a pivotal role for the suppression of NLRP3 inflammasome and interconnected cellular offenses in the augmented therapeutic potential of the RIPC/EPO-500 regimen against S-I/R-induced ALI.
Collapse
Affiliation(s)
- Mennatallah A Ali
- PharmD Program, Department of Pharmacology and Toxicology, Egypt-Japan University of Science and Technology (E-JUST), Alexandria, Egypt
| | - Asmaa A Khalifa
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Nahed H Elsokkary
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Al-Jabriyah Block 4, Hawally, Kuwait.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
2
|
Hanifa M, Suri M, Singh H, Gagnani R, Jaggi AS, Bali A. Dual Role of TRPV1 Channels in Cerebral Stroke: An Exploration from a Mechanistic and Therapeutic Perspective. Mol Neurobiol 2024; 61:10574-10592. [PMID: 38760620 DOI: 10.1007/s12035-024-04221-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/05/2024] [Indexed: 05/19/2024]
Abstract
Transient receptor potential vanilloid subfamily member 1 (TRPV1) has been strongly implicated in the pathophysiology of cerebral stroke. However, the exact role and mechanism remain elusive. TPRV1 channels are exclusively present in the neurovascular system and involve many neuronal processes. Numerous experimental investigations have demonstrated that TRPV1 channel blockers or the lack of TRPV1 channels may prevent harmful inflammatory responses during ischemia-reperfusion injury, hence conferring neuroprotection. However, TRPV1 agonists such as capsaicin and some other non-specific TRPV1 activators may induce transient/slight degree of TRPV1 channel activation to confer neuroprotection through a variety of mechanisms, including hypothermia induction, improving vascular functions, inducing autophagy, preventing neuronal death, improving memory deficits, and inhibiting inflammation. Another factor in capsaicin-mediated neuroprotection could be the desensitization of TRPV1 channels. Based on the summarized evidence, it may be plausible to suggest that TPRV1 channels have a dual role in ischemia-reperfusion-induced cerebral injury, and thus, both agonists and antagonists may produce neuroprotection depending upon the dose and duration. The current review summarizes the dual function of TRPV1 in ischemia-reperfusion-induced cerebral injury models, explains its mechanism, and predicts the future.
Collapse
Affiliation(s)
- Mohd Hanifa
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Manisha Suri
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Harshita Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Riya Gagnani
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | | | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
3
|
Sütő B, Kun J, Bagoly T, Németh T, Pintér E, Kardos D, Helyes Z. Plasma Somatostatin Levels Are Lower in Patients with Coronary Stenosis and Significantly Increase after Stent Implantation. J Clin Med 2024; 13:4727. [PMID: 39200869 PMCID: PMC11355572 DOI: 10.3390/jcm13164727] [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: 06/06/2024] [Revised: 07/22/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Background/Objectives: Stimulated capsaicin-sensitive peptidergic sensory nerves release somatostatin (SST), which has systemic anti-inflammatory and analgesic effects, correlating with the severity of tissue injury. Previous studies suggest that SST release into the systemic circulation is likely to serve as a protective mechanism during thoracic and orthopedic surgeries, scoliosis operations, and septic conditions, all involving significant tissue damage, pain, and inflammation. In a severe systemic inflammation rat model, SST released from sensory nerves into the bloodstream enhanced innate defense, reducing mortality. Inflammation is the key pathophysiological process responsible for the formation, progression, instability, and healing of atherosclerotic plaques. Methods: We measured SST-like immunoreactivity (SST-LI) in the plasma of healthy volunteers in different age groups and also that of stable angina patients with coronary heart disease (CHD) using ELISA and tracked changes during invasive coronary interventions (coronarography) with and without stent implantation. Samples were collected at (1) pre-intervention, (2) after coronarography, (3) 2 h after coronarography initiation and coronary stent placement, and (4) the next morning. Results: There was a strong negative correlation between SST-LI concentrations and age; the plasma SST-LI of older healthy volunteers (47-73 years) was significantly lower than in young ones (24-27 years). Baseline SST-LI in CHD patients who needed stents was significantly reduced compared to those not requiring stents. Plasma SST-LI significantly increased two hours post stent insertion and the next morning compared to pre-intervention levels. Conclusions: Age-related SST decrease might be a consequence of lower gene expression within specific hypo-thalamic nuclei as has been previously demonstrated in rodent animals. Reperfusion of ischemic myocardium post-stent implantation may trigger SST release, potentially offering protective benefits in coronary heart disease. Investigating this SST-mediated mechanism could offer valuable insights for future therapies.
Collapse
Affiliation(s)
- Balázs Sütő
- Department of Anaesthesia and Intensive Therapy, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - József Kun
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary
- Hungarian Centre for Genomics and Bioinformatics, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, 1117 Budapest, Hungary
| | - Teréz Bagoly
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Timea Németh
- Department of Languages for Biomedical Purposes and Communication, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, 1117 Budapest, Hungary
- Hungarian Research Network (HUN-REN-PTE), Chronic Pain Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Dorottya Kardos
- Department of Anaesthesia and Intensive Therapy, General District Hospital Szekszárd, 7100 Szekszárd, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, 1117 Budapest, Hungary
- Hungarian Research Network (HUN-REN-PTE), Chronic Pain Research Group, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
4
|
Sun C, Fan Q, Xie R, Luo C, Hu B, Wang Q. Tetherless Optical Neuromodulation: Wavelength from Orange-red to Mid-infrared. Neurosci Bull 2024; 40:1173-1188. [PMID: 38372931 PMCID: PMC11306867 DOI: 10.1007/s12264-024-01179-1] [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: 07/06/2023] [Accepted: 11/11/2023] [Indexed: 02/20/2024] Open
Abstract
Optogenetics, a technique that employs light for neuromodulation, has revolutionized the study of neural mechanisms and the treatment of neurological disorders due to its high spatiotemporal resolution and cell-type specificity. However, visible light, particularly blue and green light, commonly used in conventional optogenetics, has limited penetration in biological tissue. This limitation necessitates the implantation of optical fibers for light delivery, especially in deep brain regions, leading to tissue damage and experimental constraints. To overcome these challenges, the use of orange-red and infrared light with greater tissue penetration has emerged as a promising approach for tetherless optical neuromodulation. In this review, we provide an overview of the development and applications of tetherless optical neuromodulation methods with long wavelengths. We first discuss the exploration of orange-red wavelength-responsive rhodopsins and their performance in tetherless optical neuromodulation. Then, we summarize two novel tetherless neuromodulation methods using near-infrared light: upconversion nanoparticle-mediated optogenetics and photothermal neuromodulation. In addition, we discuss recent advances in mid-infrared optical neuromodulation.
Collapse
Affiliation(s)
- Chao Sun
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Qi Fan
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Rougang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Bingliang Hu
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Quan Wang
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China.
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China.
| |
Collapse
|
5
|
Wang XM, Li XX, Jiang B, Wang TQ, Guo Z. Morphine timing-dependent modulation of TRPV1 phosphorylation correlates with differential morphine effects on myocardial ischemia/reperfusion injury. Eur J Pharmacol 2024; 975:176648. [PMID: 38759706 DOI: 10.1016/j.ejphar.2024.176648] [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: 02/17/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Opioids are used for pain relief in patients suffering from acute myocardial ischemia or infarction. Clinical and laboratory studies demonstrate that morphine treated patients or the experimental animal model suffering acute myocardial ischemia and reperfusion, may worsen myocardial viability. As transient receptor potential vanilloid 1 (TRPV1) plays important roles in pain sensation and cardio-protection, we query whether opioids may exacerbate myocardial viability via interaction with TRPV1 activity in the pain relief. We found the co-expressions of TRPV1 and opioid μ, δ and κ receptors in adult rat cardiomyocytes. Intravenous injection of morphine (0.3 mg/kg) at 20 min after induction of myocardial ischemia, in the rat model of acute myocardial ischemia and reperfusion, induced significant reduction of phosphorylated TRPV1 (p-TRPV1) in the ventricular myocardium and increase in serum cardiac troponin I (cTnI), compared with the ischemia/reperfusion controls (all P < 0.05). The effects of morphine were completely reversed by selective opioid μ, δ and κ receptor antagonists. While significant upregulation of p-TRPV1 (P < 0.05) and improvement of ±dP/dt max (all P < 0.05) were detected in the animals giving the same dose of morphine before induction of myocardial ischemia. The changes in p-TRPV1 correlate with the alterations of cTnI (r = -0.5840, P = 0.0283) and ±dP/dt max (r = 0.8084, P = 0.0005 and r = -0.8133, P = 0.0004, respectively). The findings of this study may indicate that potentiation and attenuation of TRPV1 sensitivity correlate with the improvement of the cardiac performance and the aggravation of myocardial viability, respectively, by giving morphine before and during myocardial ischemia and reperfusion.
Collapse
Affiliation(s)
- Xin-Meng Wang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Xiao-Xi Li
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Bo Jiang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Tian-Qi Wang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Zheng Guo
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
6
|
Ma X, Guo Z, Li MR, Chen L, Zhao X, Wang TQ, Sun T. Epidural administration of large dose of opioid μ receptor agonist may impair cardiac functions and myocardial viability via desensitizing transient receptor potential vanilloid 1. Toxicol Appl Pharmacol 2024; 483:116802. [PMID: 38184280 DOI: 10.1016/j.taap.2023.116802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/06/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
The incidence of postoperative myocardial injury remains high as the underlying pathogenesis is still unknown. The dorsal root ganglion (DRG) neurons express transient receptor potential vanilloid 1 (TRPV1) and its downstream effector, calcitonin gene-related peptide (CGRP) participating in transmitting pain signals and cardiac protection. Opioids remain a mainstay therapeutic option for moderate-to-severe pain relief clinically, as a critical component of multimodal postoperative analgesia via intravenous and epidural delivery. Evidence indicates the interaction of opioids and TRPV1 activities in DRG neurons. Here, we verify the potential impairment of myocardial viability by epidural usage of opioids in postoperative analgesia. We found that large dose of epidural morphine (50 μg) significantly worsened the cardiac performance (+dP/dtmax reduction by 11% and -dP/dtmax elevation by 24%, all P < 0.001), the myocardial infarct size (morphine vs Control, 0.54 ± 0.09 IS/AAR vs. 0.23 ± 0.06 IS/AAR, P < 0.001) and reduced CGRP in the myocardium (morphine vs. Control, 9.34 ± 2.24 pg/mg vs. 21.23 ± 4.32 pg/mg, P < 0.001), while induced definite suppression of nociception in the postoperative animals. It was demonstrated that activation of μ-opioid receptor (μ-OPR) induced desensitization of TRPV1 by attenuating phosphorylation of the channel in the dorsal root ganglion neurons, via inhibiting the accumulation of cAMP. CGRP may attenuated the buildup of ROS and the reduction of mitochondrial membrane potential in cardiomyocytes induced by hypoxia/reoxygenation. The findings of this study indicate that epidurally giving large dose of μ-OPR agonist may aggravate myocardial injury by inhibiting the activity of TRPV1/CGRP pathway.
Collapse
Affiliation(s)
- Xiang Ma
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China
| | - Zheng Guo
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, Shanxi, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China.
| | - Mu-Rong Li
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China
| | - Lu Chen
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China
| | - Xing Zhao
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China
| | - Tian-Qi Wang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China
| | - Tao Sun
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan 030001, Shanxi, China
| |
Collapse
|
7
|
van Weperen VYH, Vaseghi M. Cardiac vagal afferent neurotransmission in health and disease: review and knowledge gaps. Front Neurosci 2023; 17:1192188. [PMID: 37351426 PMCID: PMC10282187 DOI: 10.3389/fnins.2023.1192188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 06/24/2023] Open
Abstract
The meticulous control of cardiac sympathetic and parasympathetic tone regulates all facets of cardiac function. This precise calibration of cardiac efferent innervation is dependent on sensory information that is relayed from the heart to the central nervous system. The vagus nerve, which contains vagal cardiac afferent fibers, carries sensory information to the brainstem. Vagal afferent signaling has been predominantly shown to increase parasympathetic efferent response and vagal tone. However, cardiac vagal afferent signaling appears to change after cardiac injury, though much remains unknown. Even though subsequent cardiac autonomic imbalance is characterized by sympathoexcitation and parasympathetic dysfunction, it remains unclear if, and to what extent, vagal afferent dysfunction is involved in the development of vagal withdrawal. This review aims to summarize the current understanding of cardiac vagal afferent signaling under in health and in the setting of cardiovascular disease, especially after myocardial infarction, and to highlight the knowledge gaps that remain to be addressed.
Collapse
Affiliation(s)
- Valerie Y. H. van Weperen
- Division of Cardiology, Department of Medicine, UCLA Cardiac Arrhythmia Center, Los Angeles, CA, United States
| | - Marmar Vaseghi
- Division of Cardiology, Department of Medicine, UCLA Cardiac Arrhythmia Center, Los Angeles, CA, United States
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
8
|
Cao Y, Redd MA, Fang C, Mizikovsky D, Li X, Macdonald PS, King GF, Palpant NJ. New Drug Targets and Preclinical Modelling Recommendations for Treating Acute Myocardial Infarction. Heart Lung Circ 2023:S1443-9506(23)00139-7. [PMID: 37230806 DOI: 10.1016/j.hlc.2022.12.015] [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: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 05/27/2023]
Abstract
Acute myocardial infarction (AMI) is the leading cause of morbidity and mortality worldwide and the primary underlying risk factor for heart failure. Despite decades of research and clinical trials, there are no drugs currently available to prevent organ damage from acute ischaemic injuries of the heart. In order to address the increasing global burden of heart failure, drug, gene, and cell-based regeneration technologies are advancing into clinical testing. In this review we highlight the burden of disease associated with AMI and the therapeutic landscape based on market analyses. New studies revealing the role of acid-sensitive cardiac ion channels and other proton-gated ion channels in cardiac ischaemia are providing renewed interest in pre- and post-conditioning agents with novel mechanisms of action that may also have implications for gene- and cell-based therapeutics. Furthermore, we present guidelines that couple new cell technologies and data resources with traditional animal modelling pipelines to help de-risk drug candidates aimed at treating AMI. We propose that improved preclinical pipelines and increased investment in drug target identification for AMI is critical to stem the increasing global health burden of heart failure.
Collapse
Affiliation(s)
- Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Meredith A Redd
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Chen Fang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Xichun Li
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Peter S Macdonald
- Cardiopulmonary Transplant Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
9
|
Maddah M, Hoseinian N, Pourfath M. An ensemble docking-based virtual screening according to different TRPV1 pore states toward identifying phytochemical activators. NEW J CHEM 2023. [DOI: 10.1039/d2nj04918j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Identifying phytochemical activators for TRPV1 using ensemble-based virtual screening, machine learning, and MD simulation.
Collapse
Affiliation(s)
- Mina Maddah
- School of Electrical and Computer Engineering, University College of Engineering, University of Tehran, Tehran, Iran
| | - Nadia Hoseinian
- School of Electrical and Computer Engineering, University College of Engineering, University of Tehran, Tehran, Iran
| | - Mahdi Pourfath
- School of Electrical and Computer Engineering, University College of Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
10
|
Xu Y, Zhao Y, Gao B. Role of TRPV1 in High Temperature-Induced Mitochondrial Biogenesis in Skeletal Muscle: A Mini Review. Front Cell Dev Biol 2022; 10:882578. [PMID: 35450292 PMCID: PMC9017999 DOI: 10.3389/fcell.2022.882578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/22/2022] [Indexed: 12/23/2022] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a protein that is susceptible to cell environment temperature. High temperatures of 40–45°C can activate the TRPV1 channel. TRPV1 is highly expressed in skeletal muscle and located on the sarcoplasmic reticulum (SR). Therefore, TRPV1 activated by high-temperature stress releases Ca2+ from the SR to the cytoplasm. Cellular Ca2+ accumulation is a key event that enhances TRPV1 activity by directly binding to the N-terminus and C-terminus. Moreover, Ca2+ is the key messenger involved in regulating mitochondrial biogenesis in skeletal muscle. Long-term activation of TRPV1 may promote mitochondrial biogenesis in skeletal muscle through the Ca2+-CaMKII-p38 MAPK-PGC-1α signaling axis. The discovery of the TRPV1 channel highlights the potential mechanism for high-temperature stress improving muscle mitochondrial biogenesis. The appropriate hot stimulus in thermal environments might be beneficial to the muscular mitochondrial adaptation for aerobic capacity. However, the investigation of TRPV1 on mitochondrial biogenesis is at an early stage. Further investigations need to examine the role of TRPV1 in response to mitochondrial biogenesis in skeletal muscle induced by different thermal environments.
Collapse
Affiliation(s)
- Yixiao Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yongcai Zhao
- College of Social Sport and Health Sciences, Tianjin University of Sport, Tianjin, China
| | - Binghong Gao
- School of Physical Education and Training, Shanghai University of Sport, Shanghai, China
- *Correspondence: Binghong Gao,
| |
Collapse
|
11
|
TRPV1 Contributes to Modulate the Nitric Oxide Pathway and Oxidative Stress in the Isolated and Perfused Rat Heart during Ischemia and Reperfusion. Molecules 2022; 27:molecules27031031. [PMID: 35164296 PMCID: PMC8839190 DOI: 10.3390/molecules27031031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
The transient vanilloid receptor potential type 1 (TRPV1) regulates neuronal and vascular functions mediated by nitric oxide (NO) and by the calcitonin gene-related peptide (CGRP). Here, we study the participation of TRPV1 in the regulation of myocardial injury caused by ischemia-reperfusion and in the control of NO, tetrahydrobiopterin (BH4), the cGMP pathway, CGRP, total antioxidant capacity (TAC), malondialdehyde (MDA) and phosphodiesterase-3 (PDE-3). Isolated hearts of Wistar rats perfused according to the Langendorff technique were used to study the effects of an agonist of TRPV1, capsaicin (CS), an antagonist, capsazepine (CZ), and their combination CZ+CS. The hearts were subjected to three conditions: (1) control, (2) ischemia and (3) ischemia-reperfusion. We determined cardiac mechanical activity and the levels of NO, cGMP, BH4, CGRP, TAC, MDA and PDE-3 in ventricular tissue after administration of CS, CZ and CZ+CS. Western blots were used to study the expressions of eNOS, iNOS and phosphorylated NOS (pNOS). Structural changes were determined by histological evaluation. CS prevented damage caused by ischemia-reperfusion by improving cardiac mechanical activity and elevating the levels of NO, cGMP, BH4, TAC and CGRP. TRPV1 and iNOS expression were increased under ischemic conditions, while eNOS and pNOS were not modified. We conclude that the activation of TRPV1 constitutes a therapeutic possibility to counteract the damage caused by ischemia and reperfusion by regulating the NO pathway through CGRP.
Collapse
|
12
|
Vörös I, Sághy É, Pohóczky K, Makkos A, Onódi Z, Brenner GB, Baranyai T, Ágg B, Váradi B, Kemény Á, Leszek P, Görbe A, Varga ZV, Giricz Z, Schulz R, Helyes Z, Ferdinandy P. Somatostatin and Its Receptors in Myocardial Ischemia/Reperfusion Injury and Cardioprotection. Front Pharmacol 2021; 12:663655. [PMID: 34803662 PMCID: PMC8602362 DOI: 10.3389/fphar.2021.663655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022] Open
Abstract
Little is known about the role of the neuropeptide somatostatin (SST) in myocardial ischemia/reperfusion injury and cardioprotection. Here, we investigated the direct cardiocytoprotective effect of SST on ischemia/reperfusion injury in cardiomyocyte cultures, as well as the expression of SST and its receptors in pig and human heart tissues. SST induced a bell-shaped, concentration-dependent cardiocytoprotection in both adult rat primary cardiomyocytes and H9C2 cells subjected to simulated ischemia/reperfusion injury. Furthermore, in a translational porcine closed-chest acute myocardial infarction model, ischemic preconditioning increased plasma SST-like immunoreactivity. Interestingly, SST expression was detectable at the protein, but not at the mRNA level in the pig left ventricles. SSTR1 and SSTR2, but not the other SST receptors, were detectable at the mRNA level by PCR and sequencing in the pig left ventricle. Moreover, remote ischemic conditioning upregulated SSTR1 mRNA. Similarly, SST expression was also detectable in healthy human interventricular septum samples at the protein level. Furthermore, SST-like immunoreactivity decreased in interventricular septum samples of patients with ischemic cardiomyopathy. SSTR1, SSTR2, and SSTR5 but not SST and the other SST receptors were detectable at the mRNA level by sequencing in healthy human left ventricles. In addition, in healthy human left ventricle samples, SSTR1 and SSTR2 mRNAs were expressed especially in vascular endothelial and some other cell types as detected by RNA Scope® in situ hybridization. This is the first demonstration that SST exerts a direct cardiocytoprotective effect against simulated ischemia/reperfusion injury. Moreover, SST is expressed in the heart tissue at the peptide level; however, it is likely to be of sensory neural origin since its mRNA is not detectable. SSTR1 and SSTR2 might be involved in the cardioprotective action of SST, but other mechanisms cannot be excluded.
Collapse
Affiliation(s)
- Imre Vörös
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Éva Sághy
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Krisztina Pohóczky
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentágothai János Research Center, University of Pécs, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - András Makkos
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zsófia Onódi
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Gábor B. Brenner
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Tamás Baranyai
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Barnabás Váradi
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Ágnes Kemény
- Szentágothai János Research Center, University of Pécs, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Department of Medical Biology, University of Pécs, Pécs, Hungary
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Cardinal Stefan Wyszyński National Institute of Cardiology, Warszawa, Poland
| | - Anikó Görbe
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V. Varga
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Zoltán Giricz
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Zsuzsanna Helyes
- Szentágothai János Research Center, University of Pécs, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Ferdinandy
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
13
|
Affiliation(s)
- Romain Guinamard
- Normandie UniversitéEA 4650, Signalisation, Electrophysiologie et Imagerie des Lésions d’Ischémie‐Reperfusion MyocardiqueGIP CyceronUNICAEN Caen France
| | - Thomas Hof
- Translational Cardiology Department of Cardiology, Inselspital Bern University Hospital, and Institute of PhysiologyUniversity of Bern Bern Switzerland
| |
Collapse
|
14
|
Yamada H, Sakata N, Tanaka T, Tagashira H, Yoshimatsu G, Kawakami R, Wada H, Iwamoto T, Kodama S. Lymphangiogenesis and angiogenesis rescue murine ischemic hindlimb via transient receptor potential vanilloid 4. J Pharmacol Sci 2021; 146:244-248. [PMID: 34116738 DOI: 10.1016/j.jphs.2021.04.009] [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: 01/21/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022] Open
Abstract
In this study, we assessed the regulation of transient receptor potential vanilloid 4 (TRPV4) promoting lymphangio/angiogenesis to improve the ischemic hindlimb animal model, and revealed that (1) a TRPV4 agonist improved the blood flow of ischemic hindlimbs by inducing both angiogenesis and lymphangiogenesis; (2) excessive TRPV4 expression was detected on lymphatic endothelial cells (LECs) in the ischemic hindlimb; and (3) hypoxic conditions promoted Ca2+ influx into LECs via TRPV4. It is considered that the upregulation of both lymphatic and blood vessels by activating TRPV4 would be a promising therapeutic strategy for peripheral artery disease.
Collapse
Affiliation(s)
- Hideaki Yamada
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Department of Cardiovascular Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Tomoko Tanaka
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Hideaki Tagashira
- Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Department of Pharmacology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Ryo Kawakami
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Hideichi Wada
- Department of Cardiovascular Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan.
| | - Takahiro Iwamoto
- Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Department of Pharmacology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan; Research Institute for Regenerative Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan, Fukuoka 814-0180, Japan.
| |
Collapse
|
15
|
Liu L, Guo M, Lv X, Wang Z, Yang J, Li Y, Yu F, Wen X, Feng L, Zhou T. Role of Transient Receptor Potential Vanilloid 4 in Vascular Function. Front Mol Biosci 2021; 8:677661. [PMID: 33981725 PMCID: PMC8107436 DOI: 10.3389/fmolb.2021.677661] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels are widely expressed in systemic tissues and can be activated by many stimuli. TRPV4, a Ca2+-permeable cation channel, plays an important role in the vasculature and is implicated in the regulation of cardiovascular homeostasis processes such as blood pressure, vascular remodeling, and pulmonary hypertension and edema. Within the vasculature, TRPV4 channels are expressed in smooth muscle cells, endothelial cells, and perivascular nerves. The activation of endothelial TRPV4 contributes to vasodilation involving nitric oxide, prostacyclin, and endothelial-derived hyperpolarizing factor pathways. TRPV4 activation also can directly cause vascular smooth muscle cell hyperpolarization and vasodilation. In addition, TRPV4 activation can evoke constriction in some specific vascular beds or under some pathological conditions. TRPV4 participates in the control of vascular permeability and vascular damage, particularly in the lung capillary endothelial barrier and lung injury. It also participates in vascular remodeling regulation mainly by controlling vasculogenesis and arteriogenesis. This review examines the role of TRPV4 in vascular function, particularly in vascular dilation and constriction, vascular permeability, vascular remodeling, and vascular damage, along with possible mechanisms, and discusses the possibility of targeting TRPV4 for therapy.
Collapse
Affiliation(s)
- Liangliang Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Mengting Guo
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaowang Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jigang Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yanting Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xin Wen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
16
|
Jia X, Yu T, Xiao C, Sheng D, Yang M, Cheng Q, Wu J, Lian T, Zhao Y, Zhang S. Expression of transient receptor potential vanilloid genes and proteins in diabetic rat heart. Mol Biol Rep 2021; 48:1217-1223. [PMID: 33523372 DOI: 10.1007/s11033-021-06182-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 11/27/2022]
Abstract
Cardiac complications are leading causes of death in diabetic patients. Imbalance of Ca2+ homeostasis is a hallmark of cardiac dysfunction in diabetes, while TRPV channels are non-selective for cations and are permeable to Ca2+. Our aim was to evaluate the expression levels of TRPV1, TRPV2, TRPV3, TRPV4, TRPV5, and TRPV6 genes and proteins in cardiac tissue at 3 days and 4, 8, and 12 weeks after induction of diabetes. Sprague-Dawley rats were assigned to control and DM groups. DM was induced by intraperitoneal injection of streptozotocin (60 mg/kg). The expression levels of TRPV genes were analyzed by the quantitative reverse transcription polymerase chain reaction, and TRPV proteins were determined by western blotting. Compared to controls, the expression levels of TRPV2, TRPV3, and TRPV6 in diabetic myocardium did not change, while TRPV1 decreased at 4, 8, and 12 weeks, TRPV4 was upregulated at 3 days and 4, 8, and 12 weeks, TRPV5 mRNA increased at 8 and 12 weeks, and TRPV5 protein increased at 4, 8, and 12 weeks. Our findings showed that TRPV1, TRPV4, and TRPV5 are associated with the diabetic heart.
Collapse
Affiliation(s)
- Xiaoli Jia
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Tao Yu
- Renhe Hospital of China Three Gorges University, Yichang, China
| | - Chao Xiao
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Deqiao Sheng
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Mengcheng Yang
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Quanyi Cheng
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Jing Wu
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Ting Lian
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China
| | - Yun Zhao
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China.
| | - Shizhong Zhang
- Department of Physiology, Medical Science College of China Three Gorges University, Yichang, China.
| |
Collapse
|
17
|
Bencsik P, Gömöri K, Szabados T, Sántha P, Helyes Z, Jancsó G, Ferdinandy P, Görbe A. Myocardial ischaemia reperfusion injury and cardioprotection in the presence of sensory neuropathy: Therapeutic options. Br J Pharmacol 2020; 177:5336-5356. [PMID: 32059259 PMCID: PMC7680004 DOI: 10.1111/bph.15021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
During the last decades, mortality from acute myocardial infarction has been dramatically reduced. However, the incidence of post-infarction heart failure is still increasing. Cardioprotection by ischaemic conditioning had been discovered more than three decades ago. Its clinical translation, however, is still an unmet need. This is mainly due to the disrupted cardioprotective signalling pathways in the presence of different cardiovascular risk factors, co-morbidities and the medication being taken. Sensory neuropathy is one of the co-morbidities that has been shown to interfere with cardioprotection. In the present review, we summarize the diverse aetiology of sensory neuropathies and the mechanisms by which these neuropathies may interfere with ischaemic heart disease and cardioprotective signalling. Finally, we suggest future therapeutic options targeting both ischaemic heart and sensory neuropathy simultaneously. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
| | - Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
| | - Péter Sántha
- Department of Physiology, Faculty of MedicineUniversity of SzegedSzegedHungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical SchoolUniversity of PécsPécsHungary
- Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research CentreUniversity of PécsPécsHungary
| | - Gábor Jancsó
- Department of Physiology, Faculty of MedicineUniversity of SzegedSzegedHungary
| | - Péter Ferdinandy
- Pharmahungary GroupSzegedHungary
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| |
Collapse
|
18
|
Liu T, Zhang G, Wang Y, Rao M, Zhang Y, Guo A, Wang M. Identification of Circular RNA-MicroRNA-Messenger RNA Regulatory Network in Atrial Fibrillation by Integrated Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8037273. [PMID: 33062700 PMCID: PMC7545447 DOI: 10.1155/2020/8037273] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Circular RNA (circRNA) is a noncoding RNA that forms a closed-loop structure, and its abnormal expression may cause disease. We aimed to find potential network for circRNA-related competitive endogenous RNA (ceRNA) in atrial fibrillation (AF). METHODS The circRNA, miRNA, and mRNA expression profiles in the heart tissue from AF patients were retrieved from the Gene Expression Omnibus database and analyzed comprehensively. Differentially expressed circRNAs (DEcircRNAs), differentially expressed miRNAs (DEmiRNAs), and differentially expressed mRNAs (DEmRNAs) were identified, followed by the establishment of DEcircRNA-DEmiRNA-DEmRNA regulatory network. Functional annotation analysis of host gene of DEcircRNAs and DEmRNAs in ceRNA regulatory network was performed. In vitro experiment and electronic validation were used to validate the expression of DEcircRNAs, DEmiRNAs, and DEmRNAs. RESULTS A total of 1611 DEcircRNAs, 51 DEmiRNAs, and 1250 DEmRNAs were identified in AF. The DEcircRNA-DEmiRNA-DEmRNA network contained 62 circRNAs, 14 miRNAs, and 728 mRNAs. Among which, two ceRNA regulatory pairs of hsa-circRNA-100053-hsa-miR-455-5p-TRPV1 and hsa-circRNA-005843-hsa-miR-188-5p-SPON1 were identified. In addition, six miRNA-mRNA regulatory pairs including hsa-miR-34c-5p-INMT, hsa-miR-1253-DDIT4L, hsa-miR-508-5p-SMOC2, hsa-miR-943-ACTA1, hsa-miR-338-3p-WIPI1, and hsa-miR-199a-3p-RAP1GAP2 were also obtained. MTOR was a significantly enriched signaling pathway of host gene of DEcircRNAs. In addition, arrhythmogenic right ventricular cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy were remarkably enriched signaling pathways of DEmRNAs in DEcircRNA-DEmiRNA-DEmRNA regulatory network. The expression validation of hsa-circRNA-402565, hsa-miR-34c-5p, hsa-miR-188-5p, SPON1, DDIT4L, SMOC2, and WIPI1 was consistent with the integrated analysis. CONCLUSION We speculated that hsa-circRNA-100053-hsa-miR-455-5p-TRPV1 and hsa-circRNA-005843-hsa-miR-188-5p-SPON1 interaction pairs may be involved in AF.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Guoru Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yaling Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Mingyue Rao
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yang Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Anjun Guo
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Mei Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
19
|
Hong KS, Lee MG. Endothelial Ca 2+ signaling-dependent vasodilation through transient receptor potential channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:287-298. [PMID: 32587123 PMCID: PMC7317173 DOI: 10.4196/kjpp.2020.24.4.287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/03/2020] [Accepted: 04/14/2020] [Indexed: 01/18/2023]
Abstract
Ca2+ signaling of endothelial cells plays a critical role in controlling blood flow and pressure in small arteries and arterioles. As the impairment of endothelial function is closely associated with cardiovascular diseases (e.g., atherosclerosis, stroke, and hypertension), endothelial Ca2+ signaling mechanisms have received substantial attention. Increases in endothelial intracellular Ca2+ concentrations promote the synthesis and release of endothelial-derived hyperpolarizing factors (EDHFs, e.g., nitric oxide, prostacyclin, or K+ efflux) or directly result in endothelial-dependent hyperpolarization (EDH). These physiological alterations modulate vascular contractility and cause marked vasodilation in resistance arteries. Transient receptor potential (TRP) channels are nonselective cation channels that are present in the endothelium, vascular smooth muscle cells, or perivascular/sensory nerves. TRP channels are activated by diverse stimuli and are considered key biological apparatuses for the Ca2+ influx-dependent regulation of vasomotor reactivity in resistance arteries. Ca2+-permeable TRP channels, which are primarily found at spatially restricted microdomains in endothelial cells (e.g., myoendothelial projections), have a large unitary or binary conductance and contribute to EDHFs or EDH-induced vasodilation in concert with the activation of intermediate/small conductance Ca2+-sensitive K+ channels. It is likely that endothelial TRP channel dysfunction is related to the dysregulation of endothelial Ca2+ signaling and in turn gives rise to vascular-related diseases such as hypertension. Thus, investigations on the role of Ca2+ dynamics via TRP channels in endothelial cells are required to further comprehend how vascular tone or perfusion pressure are regulated in normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Kwang-Seok Hong
- Department of Physical Education, College of Education, Chung-Ang University, Seoul 06974, Korea
| | - Man-Gyoon Lee
- Sports Medicine and Science, Graduate School of Physical Education, Kyung Hee University, Yongin 17104, Korea
| |
Collapse
|
20
|
Capsaicin-Sensitive Sensory Nerves and the TRPV1 Ion Channel in Cardiac Physiology and Pathologies. Int J Mol Sci 2020; 21:ijms21124472. [PMID: 32586044 PMCID: PMC7352834 DOI: 10.3390/ijms21124472] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/20/2020] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases, including coronary artery disease, ischemic heart diseases such as acute myocardial infarction and postischemic heart failure, heart failure of other etiologies, and cardiac arrhythmias, belong to the leading causes of death. Activation of capsaicin-sensitive sensory nerves by the transient receptor potential vanilloid 1 (TRPV1) capsaicin receptor and other receptors, as well as neuropeptide mediators released from them upon stimulation, play important physiological regulatory roles. Capsaicin-sensitive sensory nerves also contribute to the development and progression of some cardiac diseases, as well as to mechanisms of endogenous stress adaptation leading to cardioprotection. In this review, we summarize the role of capsaicin-sensitive afferents and the TRPV1 ion channel in physiological and pathophysiological functions of the heart based mainly on experimental results and show their diagnostic or therapeutic potentials. Although the actions of several other channels or receptors expressed on cardiac sensory afferents and the effects of TRPV1 channel activation on different non-neural cell types in the heart are not precisely known, most data suggest that stimulation of the TRPV1-expressing sensory nerves or stimulation/overexpression of TRPV1 channels have beneficial effects in cardiac diseases.
Collapse
|
21
|
Late Phases of Cardioprotection During Remote Ischemic Preconditioning and Adenosine Preconditioning Involve Activation of Neurogenic Pathway. J Cardiovasc Pharmacol 2020; 73:63-69. [PMID: 30422893 DOI: 10.1097/fjc.0000000000000634] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The role of the neurogenic pathway in early phases of cardioprotection during remote ischemic preconditioning (RIPC) and adenosine preconditioning is reported. AIM This study was designed to explore the involvement of the neurogenic pathway in late phases of cardioprotection during RIPC and adenosine preconditioning. MATERIAL AND METHODS Fifty-four Wistar rats were used and divided into 9 experimental groups. RIPC was induced by tying the blood pressure cuff around the hind limb and subjecting to 4 cycles of inflation and deflation of 5 minutes each. In early RIPC, the heart was isolated immediately after the last episode of RIPC, whereas in late RIPC, the heart was isolated 24 hours after the last cycle of RIPC. In a similar way, adenosine preconditioning was instituted in early and late phases by either isolating the heart 40 minutes or 24 hours after adenosine (4 mg/kg, intraperitoneally [i.p.]) administration. Isolated hearts were subjected to ischemia-reperfusion (I/R) injury on the Langendorff's system. RESULTS Both early and late phases of RIPC and adenosine preconditioning significantly abrogated I/R-induced myocardial injury in terms of decrease in the release of lactate dehydrogenase, creatine kinase, and decrease in infarct size. Pretreatment with hexamethonium, a ganglion blocker (20 mg/kg, i.p.), significantly abolished the cardioprotective effects of both early and late phases of RIPC and adenosine preconditioning. CONCLUSION Apart from the involvement of the neurogenic pathway in the early phases, there is a critical role of the neurogenic pathway in the late phase of cardioprotection during RIPC and adenosine preconditioning.
Collapse
|
22
|
Hof T, Chaigne S, Récalde A, Sallé L, Brette F, Guinamard R. Transient receptor potential channels in cardiac health and disease. Nat Rev Cardiol 2020; 16:344-360. [PMID: 30664669 DOI: 10.1038/s41569-018-0145-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transient receptor potential (TRP) channels are nonselective cationic channels that are generally Ca2+ permeable and have a heterogeneous expression in the heart. In the myocardium, TRP channels participate in several physiological functions, such as modulation of action potential waveform, pacemaking, conduction, inotropy, lusitropy, Ca2+ and Mg2+ handling, store-operated Ca2+ entry, embryonic development, mitochondrial function and adaptive remodelling. Moreover, TRP channels are also involved in various pathological mechanisms, such as arrhythmias, ischaemia-reperfusion injuries, Ca2+-handling defects, fibrosis, maladaptive remodelling, inherited cardiopathies and cell death. In this Review, we present the current knowledge of the roles of TRP channels in different cardiac regions (sinus node, atria, ventricles and Purkinje fibres) and cells types (cardiomyocytes and fibroblasts) and discuss their contribution to pathophysiological mechanisms, which will help to identify the best candidates for new therapeutic targets among the cardiac TRP family.
Collapse
Affiliation(s)
- Thomas Hof
- IHU-Liryc, Electrophysiology and Heart Modeling Institute, Foundation Bordeaux Université, Pessac-Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Sébastien Chaigne
- IHU-Liryc, Electrophysiology and Heart Modeling Institute, Foundation Bordeaux Université, Pessac-Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Alice Récalde
- IHU-Liryc, Electrophysiology and Heart Modeling Institute, Foundation Bordeaux Université, Pessac-Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Laurent Sallé
- Normandie Université, UNICAEN, EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, Caen, France
| | - Fabien Brette
- IHU-Liryc, Electrophysiology and Heart Modeling Institute, Foundation Bordeaux Université, Pessac-Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Romain Guinamard
- Normandie Université, UNICAEN, EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, Caen, France.
| |
Collapse
|
23
|
Tyagi S, Kaur S, Singh N, Jaggi AS. Investigating the role of acute and repeated stress on remote ischemic preconditioning-induced cardioprotection. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:111-116. [PMID: 32395208 PMCID: PMC7206844 DOI: 10.22038/ijbms.2019.36416.8678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 07/31/2019] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To study the effect of acute and repeated stress on cardioprotection-induced by remote ischemic preconditioning (RIPC). MATERIALS AND METHODS RIPC was induced by giving 4 short cycles of ischemia and reperfusion, each consisting of five min. The Langendorff's apparatus was used to perfuse the isolated rat hearts by subjecting the hearts to global ischemia of 30 min and reperfusion of 120 min. The coronary effluent was collected to measure the levels of lactate dehydrogenase (LDH) and creatine kinase (CK) for the assessment of injury to the myocardium. Myocardial infarct size was measured by the use of triphenyl tetrazolium chloride. Acute stress was induced by subjecting the animals to cold immersion stress for 5 min. However, in the case of stress adaptation, rats were exposed to a homotypic stressor (cold-water immersion stress) each of 5 min duration for five consecutive days. RESULTS RIPC demonstrated a significant decrease in ischemia-reperfusion-induced myocardial injury in terms of decrease in LDH, CK, and infarct size. However, acute stress for five minutes prior to RIPC significantly abolished its cardioprotective effects. Exogenous administration of adenosine restored RIPC-induced cardioprotective effects in the presence of acute stress. On repeated stress exposure for 5 days, stress adaptation was noted, and there was no effect of repeated stress exposure on RIPC-induced cardioprotection. However, the cardioprotective effects of adenosine were absent in the case of rats subjected to repeated episodes of stress. CONCLUSION Acute stress, but not repeated stress exposure, may alter the release of adenosine during RIPC, which may be manifested in the form of reduced cardioprotection during ischemic-reperfusion injury.
Collapse
Affiliation(s)
- Sakshi Tyagi
- Department of Pharmaceutical Sciences and Drug Research Punjabi University, Patiala, India
| | - Simranjot Kaur
- Department of Pharmaceutical Sciences and Drug Research Punjabi University, Patiala, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research Punjabi University, Patiala, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research Punjabi University, Patiala, India
| |
Collapse
|
24
|
Yu J, Chen K, Wu L, Liu X, Lu Y. Anesthetic propofol blunts remote preconditioning of trauma-induced cardioprotection via the TRPV1 receptor. Biomed Pharmacother 2019; 118:109308. [PMID: 31401396 DOI: 10.1016/j.biopha.2019.109308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/28/2019] [Accepted: 07/31/2019] [Indexed: 01/07/2023] Open
Abstract
Remote preconditioning of trauma (RPCT) by surgical incision is an effective cardioprotective strategy via the transient receptor potential vanilloid 1 (TRPV1) channel as a form of remote ischemic preconditioning (RIPC). However, cardioprotection by RIPC has been shown to be completely blocked by propofol. We thus hypothesized that propofol may interfere with RPCT induced cardioprotection, and that RPCT induces cardioprotection via the cardiac TRPV1 channel. Male Sprague-Dawley rats were subjected to 30 min of myocardial ischemia followed by 2 h of reperfusion. RPCT was achieved by a transverse abdominal incision. Additionally, propofol or the TRPV1 receptor inhibitor capsazepine (CPZ) was given before RPCT. Infarct size was assessed by triphenyltetrazolium staining. Heart TRPV1 expression was detected by Western blot and immunofluorescence. RPCT significantly reduced infarct size compared to control treatment (45.6 ± 4% versus 65.4 ± 2%, P < 0.01). This protective effect of RPCT was completely abolished by propofol and CPZ. TRPV1 channels are present in the heart. Therefore, cardioprotection by RPCT is also abolished by propofol, and cardiac TRPV1 mediates this cardioprotection.
Collapse
Affiliation(s)
- Junma Yu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, 230027, PR China; Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, PR China
| | - Ke Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Lining Wu
- Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, PR China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Yao Lu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China.
| |
Collapse
|
25
|
Singh L, Kulshrestha R, Singh N, Jaggi AS. Mechanisms involved in adenosine pharmacological preconditioning-induced cardioprotection. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:225-234. [PMID: 29719445 PMCID: PMC5928336 DOI: 10.4196/kjpp.2018.22.3.225] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/05/2018] [Accepted: 02/27/2018] [Indexed: 01/11/2023]
Abstract
Adenosine is a naturally occurring breakdown product of adenosine triphosphate and plays an important role in different physiological and pathological conditions. Adenosine also serves as an important trigger in ischemic and remote preconditioning and its release may impart cardioprotection. Exogenous administration of adenosine in the form of adenosine preconditioning may also protect heart from ischemia-reperfusion injury. Endogenous release of adenosine during ischemic/remote preconditioning or exogenous adenosine during pharmacological preconditioning activates adenosine receptors to activate plethora of mechanisms, which either independently or in association with one another may confer cardioprotection during ischemia-reperfusion injury. These mechanisms include activation of KATP channels, an increase in the levels of antioxidant enzymes, functional interaction with opioid receptors; increase in nitric oxide production; decrease in inflammation; activation of transient receptor potential vanilloid (TRPV) channels; activation of kinases such as protein kinase B (Akt), protein kinase C, tyrosine kinase, mitogen activated protein (MAP) kinases such as ERK 1/2, p38 MAP kinases and MAP kinase kinase (MEK 1) MMP. The present review discusses the role and mechanisms involved in adenosine preconditioning-induced cardioprotection.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | | | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| |
Collapse
|
26
|
Randhawa PK, Jaggi AS. Investigating the involvement of glycogen synthase kinase-3β and gap junction signaling in TRPV 1 and remote hind preconditioning-induced cardioprotection. Eur J Pharmacol 2017; 814:9-17. [PMID: 28755986 DOI: 10.1016/j.ejphar.2017.07.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/26/2017] [Accepted: 07/26/2017] [Indexed: 01/10/2023]
Abstract
Remote ischemic preconditioning (RIPC) is the phenomenon that harnesses the body's endogenous protective mechanisms against prolonged ischemia-reperfusion-induced injury. The present study aimed to explore the involvement of glycogen synthase kinase-3β and gap junction signaling in TRPV1 and remote hind preconditioning-induced cardioprotection. In the present study, four consecutive cycles (5min of ischemia-reperfusion) of remote hind limb preconditioning stimulus were delivered using a blood pressure cuff fastened at the inguinal level of the rat. The isolated rat hearts were mounted on the Langendorff's apparatus and were exposed to 30min of global ischemia-120min of reperfusion. Sustained ischemia-reperfusion led to cardiac injury that was assessed in terms of infarct size, LDH release, CK release, LVDP, +dp/dtmax, -dp/dtmin, heart rate and coronary flow rate. The pharmacological agents employed in the present study included capsaicin (10mg/kg) as TRPV1 channel activator, AR-A014418 (1 and 3mg/kg) as glycogen synthase kinase-3β inhibitor and carbenoxolone disodium (50 and 100mg/kg) as gap junction blocker. Remote hind limb, capsaicin and AR-A014418 preconditioning led to significant reduction in the infarct size, LDH release, CK release and improved LVDP, +dp/dtmax, -dp/dtmin, heart rate and coronary flow rate. However, remote hind limb, capsaicin and AR-A014418 preconditioning-induced cardioprotective effects were remarkably reduced in the presence of carbenoxolone (100mg/kg). This indicates that remote preconditioning stimulus probably activates TRPV1 channels that may inhibit glycogen synthase kinase-3β activity which subsequently enhances gap junction coupling to produce cardioprotective effects.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002 India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002 India.
| |
Collapse
|
27
|
Chen CK, Hsu PY, Wang TM, Miao ZF, Lin RT, Juo SHH. TRPV4 Activation Contributes Functional Recovery from Ischemic Stroke via Angiogenesis and Neurogenesis. Mol Neurobiol 2017; 55:4127-4135. [PMID: 28597396 DOI: 10.1007/s12035-017-0625-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
The endothelial transient receptor potential cation channel subfamily V member 4 (TRPV4) plays a crucial role in vascular remodeling; however, TRPV4-mediated angiogenesis after ischemic neuronal death as a neurorestorative strategy has not yet been thoroughly examined. In this study, we first tested whether TRPV4 activation can improve functional recovery in rats subjected to transient brain ischemia. The possible mechanisms for TRPV4 activation-promoted functional recovery were explored. A TRPV4 agonist, 4α-phorbol 12,13-didecanoate (4α-PDD), was intravenously injected via the tail vein at 6 h and 1, 2, 3, 4 days after ischemic stroke. The treatment reduced infarct volume by almost 50% (14.7 ± 3.7 vs. 29.2 ± 6.2%; p < 0.0001) and improved functional outcomes (p = 0.03) on day 5. To explore the therapeutic mechanism, we measured endothelial nitric oxide synthase (eNOS) expression and phosphorylation, vascular endothelial growth factor A (VEGFA) signaling, and neural stem/progenitor cells (NPCs). TRPV4 activation significantly increased eNOS expression and phosphorylation (serine 1177) by more than 2-fold in the ischemic region. The expressions of VEGFA and VEGF receptor-2 were significantly higher in the treated animals, especially an increase of the proangiogenic VEGFA164a isoform while a decrease of the antiangiogenic VEGFA165b isoform. We evaluated angiogenesis by detecting microvessel density in ischemic region. Using the immunohistochemistry staining, we found that 4α-PDD treatment caused a 3.4-fold increase of microvessel density (p < 0.0001). In addition, NPC proliferation and migration in the ischemic hemisphere were increased by 3-fold and 5-fold, respectively. In conclusion, our data suggest that TRPV4 activation by 4α-PDD may improve poststroke functional improvement through angiogenesis and neurogenesis.
Collapse
Affiliation(s)
- Chun-Kai Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Collage of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Yuan Hsu
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Ming Wang
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Zhi-Feng Miao
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ruey-Tay Lin
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Suh-Hang H Juo
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan. .,Institute of New Drug Development, China Medical University, Taichung, Taiwan. .,Brain Disease Research Center, China Medical University, Taichung, Taiwan. .,Center for Myopia and Eye Disease, China Medical University, Taichung, Taiwan.
| |
Collapse
|
28
|
Randhawa PK, Jaggi AS. A Review on Potential Involvement of TRPV1 Channels in Ischemia–Reperfusion Injury. J Cardiovasc Pharmacol Ther 2017; 23:38-45. [DOI: 10.1177/1074248417707050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Besides functioning as thermosensors, transient receptor potential vanilloid 1 (TRPV1) channels play a pivotal role in ischemia–reperfusion injury. Transient receptor potential vanilloid 1 channel activation attenuates ischemia–reperfusion-induced injury in various organs including the heart, lungs, kidneys, and the brain. Transient receptor potential vanilloid 1 channels are expressed on the sensory neurons innervating the myocardium, ventricles of the heart, epicardial surface of the heart, endothelial cells, and the vascular smooth muscle cells. During ischemic conditions, activation of TRPV1 channels on the perivascular nerves stimulates the release of calcitonin gene-related peptide and substance P to produce cardioprotection. Furthermore, TRPV1 channel activation reduces the generation of free radicals and inflammatory cytokines, inhibits neutrophil infiltration, and enhances the production of anti-inflammatory cytokines to reduce ischemia–reperfusion-induced tissue injury. The present review describes the potential involvement of TRPV1 channels and the signaling cascade in attenuating ischemia–reperfusion injury in various organs.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, India
| |
Collapse
|
29
|
Blockage of transient receptor potential vanilloid 4 alleviates myocardial ischemia/reperfusion injury in mice. Sci Rep 2017; 7:42678. [PMID: 28205608 PMCID: PMC5311718 DOI: 10.1038/srep42678] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 01/13/2017] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable nonselective cation channel and can be activated during ischemia/reperfusion (I/R). This study tested whether blockade of TRPV4 can alleviate myocardial I/R injury in mice. TRPV4 expression began to increase at 1 h, reached statistically at 4 h, and peaked at 24–72 h. Treatment with the selective TRPV4 antagonist HC-067047 or TRPV4 knockout markedly ameliorated myocardial I/R injury as demonstrated by reduced infarct size, decreased troponin T levels and improved cardiac function at 24 h after reperfusion. Importantly, the therapeutic window for HC-067047 lasts for at least 12 h following reperfusion. Furthermore, treatment with HC-067047 reduced apoptosis, as evidenced by the decrease in TUNEL-positive myocytes, Bax/Bcl-2 ratio, and caspase-3 activation. Meanwhile, treatment with HC-067047 attenuated the decrease in the activation of reperfusion injury salvage kinase (RISK) pathway (phosphorylation of Akt, ERK1/2, and GSK-3β), while the activation of survival activating factor enhancement (SAFE) pathway (phosphorylation of STAT3) remained unchanged. In addition, the anti-apoptotic effects of HC-067047 were abolished by the RISK pathway inhibitors. We conclude that blockade of TRPV4 reduces apoptosis via the activation of RISK pathway, and therefore might be a promising strategy to prevent myocardial I/R injury.
Collapse
|
30
|
Cardioprotection by remote ischemic conditioning and its signal transduction. Pflugers Arch 2016; 469:159-181. [DOI: 10.1007/s00424-016-1922-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/23/2022]
|
31
|
Investigating the involvement of TRPV 1 ion channels in remote hind limb preconditioning-induced cardioprotection in rats. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:117-126. [PMID: 27752734 DOI: 10.1007/s00210-016-1311-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/09/2016] [Indexed: 01/03/2023]
Abstract
Remote ischemic preconditioning (RIPC) treatment strategy is a breakthrough in the field of cardiovascular pharmacology as it has the potential to attenuate myocardial ischemia-reperfusion injury. However, the underlying intracellular pathways have not been widely explored. The present study intends to explore the possible role of TRPV1 channels in mediating remote hind limb preconditioning-induced cardioprotection. Remote hind limb preconditioning stimulus (4 cycles in succession) was delivered by tying the blood pressure cuff at the inguinal level of the rat. The Langendorff system was used to perfuse the isolated heart and afterward was subjected to 30 min of global ischemia and 120 min of reperfusion. Sustained ischemia and, thereafter, reperfusion led to cardiac injury that was assessed in terms of infarct size, lactate dehydrogenase (LDH) release, creatine kinase (CK) release, left ventricular end diastolic pressure (LVEDP), left ventricular developed pressure (LVDP), +dp/dtmax, -dp/dtmin, heart rate, rate pressure product, and coronary flow rate. The pharmacological modulators employed included capsaicin as TRPV1 agonist and capsazepine as TRPV1 antagonist. Remote hind limb preconditioning stimulus and capsaicin preconditioning (5 and 10 mg/kg) led to significant reduction in infarct size, LVEDP, LDH release, CK release, and significant improvement in LVDP, +dp/dtmax, -dp/dtmin, heart rate, rate pressure product, and coronary flow rate. However, remote hind limb preconditioning-induced cardioprotective effects were considerably abolished in the presence of capsazepine (2.5 and 5 mg/kg). This indicates that remote hind limb preconditioning stimulus possibly activates TRPV1 channels to produce cardioprotective effects.
Collapse
|
32
|
Affiliation(s)
- Friedrich C Luft
- Experimental and Clinical Research Center, Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Lindenbergerweg 80, Berlin, 13125, Germany.
| |
Collapse
|
33
|
Abstract
Remote ischemic preconditioning (RIPC) is an intriguing process whereby transient regional ischemia and reperfusion episodes to remote tissues including skeletal, renal, mesenteric provide protection to the heart against sustained ischemia-reperfusion-induced injury. Clinically, this technique has been used in patients undergoing various surgical interventions including coronary artery bypass graft surgery, abdominal aortic aneurysm repair, percutaneous coronary intervention, and heart valve surgery. The endogenous opioid system is extensively expressed in the brain to modulate pain sensation. Besides the role of opioids in relieving pain, numerous researchers have found their critical involvement in evoking cardioprotective effects. Endogenous opioids including endorphins, enkephalins, and dynorphins are released during RIPC and are critically involved in mediating RIPC-induced cardioprotective effects. It has been suggested that during RIPC, the endogenous opioids may be released into the systemic circulation and may travel via bloodstream that act on the myocardial opioid receptors to induce cardioprotection. The present review describes the potential role of opioids in mediating RIPC-induced cardioprotection.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- 1 Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Patiala, Punjab, India
| | - Amteshwar Singh Jaggi
- 1 Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Patiala, Punjab, India
| |
Collapse
|
34
|
Randhawa PK, Jaggi AS. Unraveling the role of adenosine in remote ischemic preconditioning-induced cardioprotection. Life Sci 2016; 155:140-6. [PMID: 27157518 DOI: 10.1016/j.lfs.2016.05.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/25/2022]
Abstract
Remote ischemic preconditioning (RIPC) induced by alternate cycles of preconditioning ischemia and reperfusion protects the heart against sustained ischemia-reperfusion-induced injury. This technique has been translated to clinical levels in patients undergoing various surgical interventions including coronary artery bypass graft surgery, abdominal aortic aneurysm repair, percutaneous coronary intervention and heart valve surgery. Adenosine is a master regulator of energy metabolism and reduces myocardial ischemia-reperfusion-induced injury. Furthermore, adenosine is a critical trigger as well as a mediator in RIPC-induced cardioprotection and scientists have demonstrated the role of adenosine by showing an increase in its levels in the systemic circulation during RIPC delivery. Furthermore, the blockade of cardioprotective effects of RIPC in the presence of specific adenosine receptor blockers and transgenic animals with targeted ablation of A1 receptors has also demonstrated its critical role in RIPC. The studies have shown that adenosine may elicit cardioprotection via activation of neurogenic pathway. The present review describes the possible role and mechanism of adenosine in mediating RIPC-induced cardioprotection.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002, India.
| |
Collapse
|
35
|
Sharma R, Randhawa PK, Singh N, Jaggi AS. Bradykinin in ischemic conditioning-induced tissue protection: Evidences and possible mechanisms. Eur J Pharmacol 2015; 768:58-70. [DOI: 10.1016/j.ejphar.2015.10.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 01/02/2023]
|
36
|
Randhawa PK, Jaggi AS. TRPV4 channels: physiological and pathological role in cardiovascular system. Basic Res Cardiol 2015; 110:54. [PMID: 26415881 DOI: 10.1007/s00395-015-0512-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/11/2015] [Accepted: 09/22/2015] [Indexed: 12/12/2022]
Abstract
TRPV4 channels are non-selective cation channels permeable to Ca(2+), Na(+), and Mg(2+) ions. Recently, TRPV4 channels have received considerable attention as these channels are widely expressed in the cardiovascular system including endothelial cells, cardiac fibroblasts, vascular smooth muscles, and peri-vascular nerves. Therefore, these channels possibly play a pivotal role in the maintenance of cardiovascular homeostasis. TRPV4 channels critically regulate flow-induced arteriogenesis, TGF-β1-induced differentiation of cardiac fibroblasts into myofibroblasts, and heart failure-induced pulmonary edema. These channels also mediate hypoxia-induced increase in proliferation and migration of pulmonary artery smooth muscle cells and progression of pulmonary hypertension. These channels also maintain flow-induced vasodilation and preserve vascular function by directly activating Ca(2+)-dependent KCa channels. Furthermore, these may also induce vasodilation and maintain blood pressure indirectly by evoking the release of NO, CGRP, and substance P. The present review discusses the evidences and the potential mechanisms implicated in diverse responses including arteriogenesis, cardiac remodeling, congestive heart failure-induced pulmonary edema, pulmonary hypertension, flow-induced dilation, regulation of blood pressure, and hypoxic preconditioning.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India.
| |
Collapse
|
37
|
Nakatsuji H, Numata T, Morone N, Kaneko S, Mori Y, Imahori H, Murakami T. Thermosensitive Ion Channel Activation in Single Neuronal Cells by Using Surface-Engineered Plasmonic Nanoparticles. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201505534] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
38
|
Nakatsuji H, Numata T, Morone N, Kaneko S, Mori Y, Imahori H, Murakami T. Thermosensitive Ion Channel Activation in Single Neuronal Cells by Using Surface‐Engineered Plasmonic Nanoparticles. Angew Chem Int Ed Engl 2015; 54:11725-9. [DOI: 10.1002/anie.201505534] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Hirotaka Nakatsuji
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo‐ku, Kyoto 615‐8510 (Japan)
| | - Tomohiro Numata
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo‐ku, Kyoto 615‐8510 (Japan)
| | - Nobuhiro Morone
- Institute for Integrated Cell‐Material Sciences (WPI‐iCeMS), Kyoto University, Sakyo‐ku, Kyoto 606‐8501 (Japan)
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo‐ku, Kyoto 606‐8501 (Japan)
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo‐ku, Kyoto 615‐8510 (Japan)
| | - Hiroshi Imahori
- Institute for Integrated Cell‐Material Sciences (WPI‐iCeMS), Kyoto University, Sakyo‐ku, Kyoto 606‐8501 (Japan)
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo‐ku, Kyoto 615‐8510 (Japan)
| | - Tatsuya Murakami
- Institute for Integrated Cell‐Material Sciences (WPI‐iCeMS), Kyoto University, Sakyo‐ku, Kyoto 606‐8501 (Japan)
| |
Collapse
|
39
|
Martin-Puig S, Tello D, Aragonés J. Novel perspectives on the PHD-HIF oxygen sensing pathway in cardioprotection mediated by IPC and RIPC. Front Physiol 2015; 6:137. [PMID: 26042040 PMCID: PMC4438228 DOI: 10.3389/fphys.2015.00137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/17/2015] [Indexed: 12/31/2022] Open
Abstract
Reperfusion of ischemic cardiac tissue is the standard treatment for improving clinical outcome following myocardial infarction but is inevitably associated with ischemia-reperfusion injury (IRI). Ischemic myocardial injury can be alleviated by exposing the heart to brief episodes of sublethal ischemia-reperfusion prior to the ischemic insult, a phenomenon that has been termed ischemic preconditioning (IPC). Similarly, remote IPC (RIPC) is defined as transient episodes of ischemia at a distant site before a subsequent prolonged injury of the target organ. In this setting, adaptive responses to hypoxia/ischemia in peripheral tissues include the release of soluble factors that have the potential to protect cardiomyocytes remotely. Oxygen fluctuations is a hallmark of insufficient tissue perfusion and ischemic episodes. Emerging evidence indicates that prolyl hydroxylase oxygen sensors (PHDs) and hypoxia-inducible transcription factors (HIFs) are critical regulators of IPC and RIPC. In this review, we discuss recent findings concerning the role of the PHD-HIF axis in IPC and RIPC-mediated cardioprotection and examine molecular pathways and cell types that might be involved. We also appraise the therapeutic value of targeting the PHD-HIF axis to enhance cardiac tolerance against IRI.
Collapse
Affiliation(s)
- Silvia Martin-Puig
- Cell and Developmental Biology Department, Centro Nacional de Investigaciones Cardiovasculares Madrid, Spain
| | - Daniel Tello
- Research Unit, Hospital Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid Madrid, Spain
| | - Julián Aragonés
- Research Unit, Hospital Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid Madrid, Spain
| |
Collapse
|