1
|
Gaydarski L, Petrova K, Stanchev S, Pelinkov D, Iliev A, Dimitrova IN, Kirkov V, Landzhov B, Stamenov N. Morphometric and Molecular Interplay in Hypertension-Induced Cardiac Remodeling with an Emphasis on the Potential Therapeutic Implications. Int J Mol Sci 2025; 26:4022. [PMID: 40362262 PMCID: PMC12071960 DOI: 10.3390/ijms26094022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Hypertension-induced cardiac remodeling is a complex process driven by interconnected molecular and cellular mechanisms that culminate in hypertensive myocardium, characterized by ventricular hypertrophy, fibrosis, impaired angiogenesis, and myocardial dysfunction. This review discusses the histomorphometric changes in capillary density, fibrosis, and mast cells in the hypertensive myocardium and delves into the roles of key regulatory systems, including the apelinergic system, vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathways, and nitric oxide (NO)/nitric oxide synthase (NOS) signaling in the pathogenesis of hypertensive heart disease (HHD). Capillary rarefaction, a hallmark of HHD, contributes to myocardial ischemia and fibrosis, underscoring the importance of maintaining vascular integrity. Targeting capillary density (CD) through antihypertensive therapy or angiogenic interventions could significantly improve cardiac outcomes. Myocardial fibrosis, mediated by excessive collagen deposition and influenced by fibroblast growth factor-2 (FGF-2) and transforming growth factor-beta (TGF-β), plays a pivotal role in the structural remodeling of hypertensive myocardium. While renin-angiotensin-aldosterone system (RAAS) inhibitors show anti-fibrotic effects, more targeted therapies are needed to address fibrosis directly. Mast cells, though less studied in humans, emerge as critical regulators of cardiac remodeling through their release of pro-fibrotic mediators such as histamine, tryptase, and FGF-2. The apelinergic system emerges as a promising therapeutic target due to its vasodilatory, anti-fibrotic, and cardioprotective properties. The system counteracts the deleterious effects of the RAAS and has demonstrated efficacy in preclinical models of hypertension-induced cardiac damage. Despite its potential, human studies on apelin analogs remain limited, warranting further exploration to evaluate their clinical utility. VEGF signaling plays a dual role, facilitating angiogenesis and compensatory remodeling during the early stages of arterial hypertension (AH) but contributing to maladaptive changes when dysregulated. Modulating VEGF signaling through exercise or pharmacological interventions has shown promise in improving CD and mitigating hypertensive cardiac damage. However, VEGF inhibitors, commonly used in oncology, can exacerbate AH and endothelial dysfunction, highlighting the need for therapeutic caution. The NO/NOS pathway is essential for vascular homeostasis and the prevention of oxidative stress. Dysregulation of this pathway, particularly endothelial NOS (eNOS) uncoupling and inducible NOS (iNOS) overexpression, leads to endothelial dysfunction and nitrosative stress in hypertensive myocardium. Strategies to restore NO bioavailability, such as tetrahydrobiopterin (BH4) supplementation and antioxidants, hold potential for therapeutic application but require further validation. Future studies should adopt a multidisciplinary approach to integrate molecular insights with clinical applications, paving the way for more personalized and effective treatments for HHD. Addressing these challenges will not only enhance the understanding of hypertensive myocardium but also improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Lyubomir Gaydarski
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Kristina Petrova
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Stancho Stanchev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Dimitar Pelinkov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Alexandar Iliev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Iva N. Dimitrova
- Department of Cardiology, University Hospital “St. Ekaterina”, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Vidin Kirkov
- Department of Health Policy and Management, Faculty of Public Health ‘Prof. Dr. Tzekomir Vodenicharov’, Medical University of Sofia, 1527 Sofia, Bulgaria;
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Nikola Stamenov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| |
Collapse
|
2
|
Kurhaluk N, Tkaczenko H. L-Arginine and Nitric Oxide in Vascular Regulation-Experimental Findings in the Context of Blood Donation. Nutrients 2025; 17:665. [PMID: 40004994 PMCID: PMC11858268 DOI: 10.3390/nu17040665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
This narrative review provides an analysis of the role of nitric oxide (NO) and its precursors, particularly L-arginine, in vascular regulation and health, with an emphasis on findings from our experimental research in animal models. NO serves as a critical mediator of vascular function, contributing to vasodilation, the regulation of blood flow, and the prevention of thrombosis. As a primary precursor of NO, L-arginine is essential for maintaining endothelial integrity, modulating mitochondrial function, and reducing oxidative damage. This review synthesises the data and contextualises these findings within the physiological challenges faced by blood donors, such as repeated blood donation and associated oxidative stress. It examines the effects of L-arginine supplementation on mitochondrial respiration, lipid peroxidation, and microsomal oxidation in different conditions, including differences in age, gender, and dietary interventions. The mechanisms by which L-arginine enhances NO production, improves vascular elasticity, and alleviates endothelial dysfunction caused by reduced NO bioavailability are also investigated. By integrating experimental findings with insights from the existing literature, this review provides a perspective on the potential of L-arginine supplementation to address the specific physiological needs of blood donors. It highlights the importance of personalised nutritional approaches in enhancing donor recovery and vascular resilience. In addition, this review assesses the wider implications of L-arginine supplementation in mitigating oxidative stress and preserving vascular function. The interplay between NO bioavailability, dietary factors, and physiological adaptation in blood donors is highlighted, along with the identification of current knowledge gaps and recommendations for future research. By presenting both original experimental evidence and a critical synthesis of the literature, this article highlights the therapeutic potential of NO precursors, particularly L-arginine, in promoting vascular health in the context of blood donation.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22b, 76-200 Słupsk, Poland;
| | | |
Collapse
|
3
|
Sandner P, Follmann M, Becker-Pelster E, Hahn MG, Meier C, Freitas C, Roessig L, Stasch JP. Soluble GC stimulators and activators: Past, present and future. Br J Pharmacol 2024; 181:4130-4151. [PMID: 34600441 DOI: 10.1111/bph.15698] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
The discovery of soluble GC (sGC) stimulators and sGC activators provided valuable tools to elucidate NO-sGC signalling and opened novel pharmacological opportunities for cardiovascular indications and beyond. The first-in-class sGC stimulator riociguat was approved for pulmonary hypertension in 2013 and vericiguat very recently for heart failure. sGC stimulators enhance sGC activity independent of NO and also act synergistically with endogenous NO. The sGC activators specifically bind to, and activate, the oxidised haem-free form of sGC. Substantial research efforts improved on the first-generation sGC activators such as cinaciguat, culminating in the discovery of runcaciguat, currently in clinical Phase II trials for chronic kidney disease and diabetic retinopathy. Here, we highlight the discovery and development of sGC stimulators and sGC activators, their unique modes of action, their preclinical characteristics and the clinical studies. In the future, we expect to see more sGC agonists in new indications, reflecting their unique therapeutic potential.
Collapse
Affiliation(s)
- Peter Sandner
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
- Institute of Pharmacology, Hannover Medical School, Hanover, Germany
| | - Markus Follmann
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | | | - Michael G Hahn
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | - Christian Meier
- Pharmaceuticals Medical Affairs and Pharmacovigilance, Bayer AG, Berlin, Germany
| | - Cecilia Freitas
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | - Lothar Roessig
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | - Johannes-Peter Stasch
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
- Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
4
|
Zhang Y, Jiang M, Wang T. Reactive oxygen species (ROS)-responsive biomaterials for treating myocardial ischemia-reperfusion injury. Front Bioeng Biotechnol 2024; 12:1469393. [PMID: 39286345 PMCID: PMC11402825 DOI: 10.3389/fbioe.2024.1469393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a critical issue that arises when restoring blood flow after an ischemic event in the heart. Excessive reactive oxygen species (ROS) production during this process exacerbates cellular damage and impairs cardiac function. Recent therapeutic strategies have focused on leveraging the ROS microenvironment to design targeted drug delivery systems. ROS-responsive biomaterials have emerged as promising candidates, offering enhanced therapeutic efficacy with reduced systemic adverse effects. This review examines the mechanisms of ROS overproduction during myocardial ischemia-reperfusion and summarizes significant advancements in ROS-responsive biomaterials for MIRI treatment. We discuss various chemical strategies to impart ROS sensitivity to these materials, emphasizing ROS-induced solubility switches and degradation mechanisms. Additionally, we highlight various ROS-responsive therapeutic platforms, such as nanoparticles and hydrogels, and their unique advantages in drug delivery for MIRI. Preclinical studies demonstrating the efficacy of these materials in mitigating MIRI in animal models are reviewed, alongside their mechanisms of action and potential clinical implications. We also address the challenges and future prospects of translating these state of the art biomaterial-based therapeutics into clinical practice to improve MIRI management and cardiac outcomes. This review will provide valuable insights for researchers and clinicians working on novel therapeutic strategies for MIRI intervention.
Collapse
Affiliation(s)
- Ying Zhang
- Natural and Biomimetic Medicine Research Center, Tissue-Orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Mantang Jiang
- Natural and Biomimetic Medicine Research Center, Tissue-Orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Wang
- Natural and Biomimetic Medicine Research Center, Tissue-Orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Chen Y, Li B, Quan J, Li Z, Li Y, Tang Y. Inhibition of Ferroptosis by Mesenchymal Stem Cell-Derived Exosomes in Acute Spinal Cord Injury: Role of Nrf2/GCH1/BH4 Axis. Neurospine 2024; 21:642-655. [PMID: 38955534 PMCID: PMC11224743 DOI: 10.14245/ns.2448038.019] [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: 01/05/2024] [Revised: 03/26/2024] [Accepted: 04/06/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE The therapeutic benefits of exosomes obtained from mesenchymal stem cells (MSCs) in acute spinal cord injury (SCI) have been demonstrated in recent years, but the precise mechanisms remain unknown. In this study, the efficacy and mechanisms of MSC-derived exosomes (MSC-Exo) in acute SCI were investigated. METHODS By utilizing a BV2 ferroptosis cellular model and an SCI rat model, we investigated the effects of MSC-Exo on iron death related indicators and NF-E2 related factor 2 (Nrf2)/GTP cyclolase I (GCH1)/5,6,7,8-tetrahydrobiopterin (BH4) signaling axis, as well as their therapeutic effects on SCI rats. RESULTS The results revealed that MSC-Exo effectively inhibited the production of ferrous iron, lipid peroxidation products malonaldehyde and reactive oxygen species, and ferroptosis-promoting factor prostaglandin-endoperoxide synthase 2. Concurrently, they upregulated ferroptosis suppressors FTH-1 (ferritin heavy chain 1), SLC7A11 (solute carrier family 7 member 11), FSP1 (ferroptosis suppressor protein 1), and GPX4 (glutathione peroxidase 4), contributing to enhanced neurological recovery in SCI rats. Further analysis showed the Nrf2/GTP/BH4 signaling pathway's critical role in suppressing ferroptosis. Additionally, MSC-Exo was found to inhibit lipopolysaccharide-induced ferroptosis in BV2 cells and SCI rats by activating the Nrf2/GCH1/BH4 axis. CONCLUSION In summary, the study demonstrates that MSC-Exo mitigates microglial cell ferroptosis via the Nrf2/GCH1/BH4 axis, showing potential for preserving and restoring neurological function post-SCI.
Collapse
Affiliation(s)
- Yixin Chen
- Department of Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Bingfa Li
- Department of Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Jing Quan
- Department of Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Zhe Li
- Department of Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Yan Li
- Department of Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Yinbo Tang
- Department of Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
6
|
Xia W, Zhang M, Liu C, Wang S, Xu A, Xia Z, Pang L, Cai Y. Exploring the therapeutic potential of tetrahydrobiopterin for heart failure with preserved ejection fraction: A path forward. Life Sci 2024; 345:122594. [PMID: 38537900 DOI: 10.1016/j.lfs.2024.122594] [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: 01/17/2024] [Revised: 03/10/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
A large number of patients are affected by classical heart failure (HF) symptomatology with preserved ejection fraction (HFpEF) and multiorgan syndrome. Due to high morbidity and mortality rate, hospitalization and mortality remain serious socioeconomic problems, while the lack of effective pharmacological or device treatment means that HFpEF presents a major unmet medical need. Evidence from clinical and basic studies demonstrates that systemic inflammation, increased oxidative stress, and impaired mitochondrial function are the common pathological mechanisms in HFpEF. Tetrahydrobiopterin (BH4), beyond being an endogenous co-factor for catalyzing the conversion of some essential biomolecules, has the capacity to prevent systemic inflammation, enhance antioxidant resistance, and modulate mitochondrial energy production. Therefore, BH4 has emerged in the last decade as a promising agent to prevent or reverse the progression of disorders such as cardiovascular disease. In this review, we cover the clinical progress and limitations of using downstream targets of nitric oxide (NO) through NO donors, soluble guanylate cyclase activators, phosphodiesterase inhibitors, and sodium-glucose co-transporter 2 inhibitors in treating cardiovascular diseases, including HFpEF. We discuss the use of BH4 in association with HFpEF, providing new evidence for its potential use as a pharmacological option for treating HFpEF.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Miao Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Guangdong, China
| | - Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
7
|
Belenichev I, Popazova O, Bukhtiyarova N, Savchenko D, Oksenych V, Kamyshnyi O. Modulating Nitric Oxide: Implications for Cytotoxicity and Cytoprotection. Antioxidants (Basel) 2024; 13:504. [PMID: 38790609 PMCID: PMC11118938 DOI: 10.3390/antiox13050504] [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: 03/26/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Despite the significant progress in the fields of biology, physiology, molecular medicine, and pharmacology; the designation of the properties of nitrogen monoxide in the regulation of life-supporting functions of the organism; and numerous works devoted to this molecule, there are still many open questions in this field. It is widely accepted that nitric oxide (•NO) is a unique molecule that, despite its extremely simple structure, has a wide range of functions in the body, including the cardiovascular system, the central nervous system (CNS), reproduction, the endocrine system, respiration, digestion, etc. Here, we systematize the properties of •NO, contributing in conditions of physiological norms, as well as in various pathological processes, to the mechanisms of cytoprotection and cytodestruction. Current experimental and clinical studies are contradictory in describing the role of •NO in the pathogenesis of many diseases of the cardiovascular system and CNS. We describe the mechanisms of cytoprotective action of •NO associated with the regulation of the expression of antiapoptotic and chaperone proteins and the regulation of mitochondrial function. The most prominent mechanisms of cytodestruction-the initiation of nitrosative and oxidative stresses, the production of reactive oxygen and nitrogen species, and participation in apoptosis and mitosis. The role of •NO in the formation of endothelial and mitochondrial dysfunction is also considered. Moreover, we focus on the various ways of pharmacological modulation in the nitroxidergic system that allow for a decrease in the cytodestructive mechanisms of •NO and increase cytoprotective ones.
Collapse
Affiliation(s)
- Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Olena Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Dmytro Savchenko
- Department of Pharmacy and Industrial Drug Technology, Bogomolets National Medical University, 01601 Kyiv, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
8
|
Yan R, Zhang X, Xu W, Li J, Sun Y, Cui S, Xu R, Li W, Jiao L, Wang T. ROS-Induced Endothelial Dysfunction in the Pathogenesis of Atherosclerosis. Aging Dis 2024; 16:AD.2024.0309. [PMID: 38502586 PMCID: PMC11745424 DOI: 10.14336/ad.2024.0309] [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: 01/28/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
Various signaling pathways are regulated by reactive oxygen species (ROS), which are radical oxygen intermediates under normal physiological conditions. However, when the buffering capacity of antioxidant enzymes is exceeded by the accumulation of ROS, oxidative stress, and endothelial cell dysfunction occur, which have been recognized as key contributors to the development of atherosclerosis. In this review, an overview is provided on mechanisms underlying ROS generation in endothelial cells and the involved regulatory pathways. Further, we discuss the ROS induced endothelial cell dysfunction and its relationship with atherosclerosis. Current knowledge on ROS-induced endothelial impairment is presented, characterized by decreased NO bioavailability, intracellular dysfunction and ox-LDL accumulation. Furthermore, biomarkers such as oxidative products of lipid, protein, and nucleotide are discussed as measurements for ROS levels. Novel interventions targeting oxidative stress are listed as potential pharmacotherapies in clinical practice. In conclusion, this review presents a systematic analysis of the mechanisms underlying ROS generation and elucidates how manipulation of these mechanisms can safeguard endothelial cell function.
Collapse
Affiliation(s)
- Ruiyi Yan
- Eight-year Medical Doctor Program, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Jiayao Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Yixin Sun
- First Hospital, Peking University, Beijing, China.
| | - Shengyan Cui
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| |
Collapse
|
9
|
Clark GC, Lai A, Agarwal A, Liu Z, Wang XY. Biopterin metabolism and nitric oxide recoupling in cancer. Front Oncol 2024; 13:1321326. [PMID: 38469569 PMCID: PMC10925643 DOI: 10.3389/fonc.2023.1321326] [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: 10/13/2023] [Accepted: 12/26/2023] [Indexed: 03/13/2024] Open
Abstract
Tetrahydrobiopterin is a cofactor necessary for the activity of several enzymes, the most studied of which is nitric oxide synthase. The role of this cofactor-enzyme relationship in vascular biology is well established. Recently, tetrahydrobiopterin metabolism has received increasing attention in the field of cancer immunology and immunotherapy due to its involvement in the cytotoxic T cell response. Past research has demonstrated that when the availability of BH4 is low, as it is in chronic inflammatory conditions and tumors, electron transfer in the active site of nitric oxide synthase becomes uncoupled from the oxidation of arginine. This results in the production of radical species that are capable of a direct attack on tetrahydrobiopterin, further depleting its local availability. This feedforward loop may act like a molecular switch, reinforcing low tetrahydrobiopterin levels leading to altered NO signaling, restrained immune effector activity, and perpetual vascular inflammation within the tumor microenvironment. In this review, we discuss the evidence for this underappreciated mechanism in different aspects of tumor progression and therapeutic responses. Furthermore, we discuss the preclinical evidence supporting a clinical role for tetrahydrobiopterin supplementation to enhance immunotherapy and radiotherapy for solid tumors and the potential safety concerns.
Collapse
Affiliation(s)
- Gene Chatman Clark
- Department of Biochemistry, Virginia Commonwealth University, Richmond, VA, United States
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Zheng Liu
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Xiang-Yang Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
10
|
Wilson SK, Thomas J. BH4 as a Therapeutic Target for ADHD: Relevance to Neurotransmitters and Stress-Driven Symptoms. J Atten Disord 2024; 28:161-167. [PMID: 37942650 DOI: 10.1177/10870547231204012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Tetrahydrobiopterin (BH4) is a critical cofactor in a variety of metabolic pathways that have been linked to ADHD. There have been no previous studies utilizing BH4 as a supplement for ADHD. BH4 has been approved as a treatment for phenylketonuria (PKU). Individuals with PKU and ADHD appear to have low DA levels in common, suggesting that the hypodopaminergic state seen in both illnesses could be a relationship between the two. Clinical research involving supplementation of BH4 has shown low occurrence of adverse. In experiments, BH4 has also been found to have good blood-brain barrier permeability. BH4 also has the ability in scavenging ROS activity, which is an implication of stress and is seen in ADHD. BH4's significance in ADHD is reviewed in this paper because of its involvement in numerous neurodevelopmental metabolic pathways, and we anticipate that exogenous BH4 can be used to treat ADHD.
Collapse
Affiliation(s)
- Samson K Wilson
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala
| | - Jaya Thomas
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala
| |
Collapse
|
11
|
Han X, Wang H, Du F, Zeng X, Guo C. Nrf2 for a key member of redox regulation: A novel insight against myocardial ischemia and reperfusion injuries. Biomed Pharmacother 2023; 168:115855. [PMID: 37939614 DOI: 10.1016/j.biopha.2023.115855] [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: 08/09/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023] Open
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2), a nuclear transcription factor, modulates genes responsible for antioxidant responses against toxic and oxidative stress to maintain redox homeostasis and participates in varieties of cellular processes such as metabolism and inflammation during myocardial ischemia and reperfusion injuries (MIRI). The accumulation of reactive oxygen species (ROS) from damaged mitochondria, xanthine oxidase, NADPH oxidases, and inflammation contributes to depraved myocardial ischemia and reperfusion injuries. Considering that Nrf2 played crucial roles in antagonizing oxidative stress, it is reasonable to delve into the up or down-regulated molecular mechanisms of Nrf2 in the progression of MIRI to provide the possibility of new therapeutic medicine targeting Nrf2 in cardiovascular diseases. This review systematically describes the generation of ROS, the regulatory metabolisms of Nrf2 as well as several natural or synthetic compounds activating Nrf2 during MIRI, which might provide novel insights for the anti-oxidative stress and original ideas targeting Nrf2 for the prevention and treatment in cardiovascular diseases.
Collapse
Affiliation(s)
- Xuejie Han
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China
| | - Fenghe Du
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, PR China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China.
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China.
| |
Collapse
|
12
|
Mollace R, Scarano F, Bava I, Carresi C, Maiuolo J, Tavernese A, Gliozzi M, Musolino V, Muscoli S, Palma E, Muscoli C, Salvemini D, Federici M, Macrì R, Mollace V. Modulation of the nitric oxide/cGMP pathway in cardiac contraction and relaxation: Potential role in heart failure treatment. Pharmacol Res 2023; 196:106931. [PMID: 37722519 DOI: 10.1016/j.phrs.2023.106931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Evidence exists that heart failure (HF) has an overall impact of 1-2 % in the global population being often associated with comorbidities that contribute to increased disease prevalence, hospitalization, and mortality. Recent advances in pharmacological approaches have significantly improved clinical outcomes for patients with vascular injury and HF. Nevertheless, there remains an unmet need to clarify the crucial role of nitric oxide/cyclic guanosine 3',5'-monophosphate (NO/cGMP) signalling in cardiac contraction and relaxation, to better identify the key mechanisms involved in the pathophysiology of myocardial dysfunction both with reduced (HFrEF) as well as preserved ejection fraction (HFpEF). Indeed, NO signalling plays a crucial role in cardiovascular homeostasis and its dysregulation induces a significant increase in oxidative and nitrosative stress, producing anatomical and physiological cardiac alterations that can lead to heart failure. The present review aims to examine the molecular mechanisms involved in the bioavailability of NO and its modulation of downstream pathways. In particular, we focus on the main therapeutic targets and emphasize the recent evidence of preclinical and clinical studies, describing the different emerging therapeutic strategies developed to counteract NO impaired signalling and cardiovascular disease (CVD) development.
Collapse
Affiliation(s)
- Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Saverio Muscoli
- Division of Cardiology, Foundation PTV Polyclinic Tor Vergata, Rome 00133, Italy
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy.
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Renato Dulbecco Institute, Lamezia Terme, Catanzaro 88046, Italy.
| |
Collapse
|
13
|
Spinelli S, Guida L, Passalacqua M, Magnone M, Cossu V, Sambuceti G, Marini C, Sturla L, Zocchi E. Abscisic Acid and Its Receptors LANCL1 and LANCL2 Control Cardiomyocyte Mitochondrial Function, Expression of Contractile, Cytoskeletal and Ion Channel Proteins and Cell Proliferation via ERRα. Antioxidants (Basel) 2023; 12:1692. [PMID: 37759995 PMCID: PMC10526111 DOI: 10.3390/antiox12091692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The cross-kingdom stress hormone abscisic acid (ABA) and its mammalian receptors LANCL1 and LANCL2 regulate the response of cardiomyocytes to hypoxia by activating NO generation. The overexpression of LANCL1/2 increases transcription, phosphorylation and the activity of eNOS and improves cell vitality after hypoxia/reoxygenation via the AMPK/PGC-1α axis. Here, we investigated whether the ABA/LANCL system also affects the mitochondrial oxidative metabolism and structural proteins. Mitochondrial function, cell cycle and the expression of cytoskeletal, contractile and ion channel proteins were studied in H9c2 rat cardiomyoblasts overexpressing or silenced by LANCL1 and LANCL2, with or without ABA. Overexpression of LANCL1/2 significantly increased, while silencing conversely reduced the mitochondrial number, OXPHOS complex I, proton gradient, glucose and palmitate-dependent respiration, transcription of uncoupling proteins, expression of proteins involved in cytoskeletal, contractile and electrical functions. These effects, and LANCL1/2-dependent NO generation, are mediated by transcription factor ERRα, upstream of the AMPK/PGC1-α axis and transcriptionally controlled by the LANCL1/2-ABA system. The ABA-LANCL1/2 hormone-receptor system controls fundamental aspects of cardiomyocyte physiology via an ERRα/AMPK/PGC-1α signaling axis and ABA-mediated targeting of this axis could improve cardiac function and resilience to hypoxic and dysmetabolic conditions.
Collapse
Affiliation(s)
- Sonia Spinelli
- Laboratorio di Nefrologia Molecolare, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy
| | - Lucrezia Guida
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (L.G.); (M.P.); (M.M.)
| | - Mario Passalacqua
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (L.G.); (M.P.); (M.M.)
| | - Mirko Magnone
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (L.G.); (M.P.); (M.M.)
| | - Vanessa Cossu
- Section Human Anatomy, Department of Experimental Medicine (DIMES), University of Genova, 16126 Genova, Italy;
- U.O. Medicina Nucleare, IRCCS Ospedale Policlinico San Martino, 16131 Genova, Italy; (G.S.); (C.M.)
| | - Gianmario Sambuceti
- U.O. Medicina Nucleare, IRCCS Ospedale Policlinico San Martino, 16131 Genova, Italy; (G.S.); (C.M.)
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy
| | - Cecilia Marini
- U.O. Medicina Nucleare, IRCCS Ospedale Policlinico San Martino, 16131 Genova, Italy; (G.S.); (C.M.)
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), 20100 Milan, Italy
| | - Laura Sturla
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (L.G.); (M.P.); (M.M.)
| | - Elena Zocchi
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (L.G.); (M.P.); (M.M.)
| |
Collapse
|
14
|
Feng X, Mei Y, Xie P, Xing Z, Wang X, Cui L, Xu R. Serum folic acid: an effective indicator for arteriogenic erectile dysfunction. Front Endocrinol (Lausanne) 2023; 14:1080188. [PMID: 37554765 PMCID: PMC10405823 DOI: 10.3389/fendo.2023.1080188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/30/2023] [Indexed: 08/10/2023] Open
Abstract
Background The present study is the first to explore the correlation between serum folic acid (FA) level and penile arterial peak systolic velocity (PSV) as measured via penile color Doppler ultrasonography (PDU), which directly reflects endothelial function in the penile artery. Materials and methods A total of 244 consecutive erectile dysfunction (ED) patients and 72 healthy controls, recruited from the Andrology department and the Healthy Physical Examination Center of our hospital, respectively, from June 2020 to April 2022, were included in the study. Serum FA was measured in ED patients and healthy controls, and PDU examinations were conducted for all eligible ED patients. The Pearson method was used to evaluate the correlation between FA levels and PDU parameters in ED patients. A receiver operating characteristic (ROC) curve analysis was also performed to calculate the sensitivity and specificity of these parameters for prediction of arteriogenic ED. Results After the PDU test, the average serum FA level among patients diagnosed with arteriogenic ED was 8.08 ± 2.64 ng/ml, lower than the average of 10.78 ± 2.87 ng/ml among healthy controls. There were no statistically significant inter-group differences on any basic parameters, including age, body mass index, fasting blood glucose, total cholesterol, and triglyceride. For further analysis, we divided the arteriogenic ED group into three subgroups by PSV range to compare serum FA levels among these subgroups. The mean FA levels in each of these groups were 5.97 ± 1.51ng/ml, and 8.21 ± 2.37ng/ml, and 10.55 ± 2.56ng/ml, while the corresponding PSV values were 15.75 ± 2.39cm/s, 23.53 ± 2.19cm/s, and 32.72 ± 1.64cm/s. Overall, a positive correlation between PSV and FA level was found among patients with arteriogenic ED (r=0.605, P<0.001). Furthermore, when FA level was used, with a cut-off value of 10.045 ng/ml, as a criterion to distinguish patients with arteriogenic ED from healthy controls, the area under the curve (AUC) was 0.772 (95% confidential interval: [0.696, 0.848]), for a sensitivity of 0.611 and specificity of 0.824. Conclusion Serum FA level is positively correlated with PSV in ED patients, and has the ability to distinguish patients with arteriogenic ED from healthy controls. Taking these findings together, FA deficiency should be regarded as an independent risk factor for arteriogenic ED.
Collapse
Affiliation(s)
- Xingliang Feng
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Urology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Yangyang Mei
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Pinpeng Xie
- Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, Hefei, China
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhaoyu Xing
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Urology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Xiaogang Wang
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Urology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Li Cui
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Urology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Renfang Xu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Urology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| |
Collapse
|
15
|
Minjares M, Wu W, Wang JM. Oxidative Stress and MicroRNAs in Endothelial Cells under Metabolic Disorders. Cells 2023; 12:1341. [PMID: 37174741 PMCID: PMC10177439 DOI: 10.3390/cells12091341] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
Reactive oxygen species (ROS) are radical oxygen intermediates that serve as important second messengers in signal transduction. However, when the accumulation of these molecules exceeds the buffering capacity of antioxidant enzymes, oxidative stress and endothelial cell (EC) dysfunction occur. EC dysfunction shifts the vascular system into a pro-coagulative, proinflammatory state, thereby increasing the risk of developing cardiovascular (CV) diseases and metabolic disorders. Studies have turned to the investigation of microRNA treatment for CV risk factors, as these post-transcription regulators are known to co-regulate ROS. In this review, we will discuss ROS pathways and generation, normal endothelial cell physiology and ROS-induced dysfunction, and the current knowledge of common metabolic disorders and their connection to oxidative stress. Therapeutic strategies based on microRNAs in response to oxidative stress and microRNA's regulatory roles in controlling ROS will also be explored. It is important to gain an in-depth comprehension of the mechanisms generating ROS and how manipulating these enzymatic byproducts can protect endothelial cell function from oxidative stress and prevent the development of vascular disorders.
Collapse
Affiliation(s)
- Morgan Minjares
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
| | - Wendy Wu
- Vera P Shiffman Medical Library, Wayne State University, 320 E Canfield St., Detroit, MI 48201, USA;
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
- Center for Molecular Medicine and Genetics, Wayne State University, 320 E Canfield St., Detroit, MI 48201, USA
- Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI 48201, USA
| |
Collapse
|
16
|
Ye J, Lyu TJ, Li LY, Liu Y, Zhang H, Wang X, Xi X, Liu ZJ, Gao JQ. Ginsenoside Re attenuates myocardial ischemia/reperfusion induced ferroptosis via miR-144-3p/SLC7A11. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154681. [PMID: 36893674 DOI: 10.1016/j.phymed.2023.154681] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 01/12/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Ginsenoside Re is an active component in ginseng that confers protection against myocardial ischemia/reperfusion (I/R) injury. Ferroptosis is a type of regulated cell death found in various diseases. PURPOSE Our study aims to investigate the role of ferroptosis and the protective mechanism of Ginsenoside Re in myocardial ischemia/reperfusion. METHODS In the present study, we treated rats for five days with Ginsenoside Re, then established the myocardial ischemia/reperfusion injury rat model to detect molecular implications in myocardial ischemia/reperfusion regulation and to determine the underlying mechanism. RESULTS This study identifies the mechanism behind ginsenoside Re's effect on myocardial ischemia/reperfusion injury and its regulation of ferroptosis through miR-144-3p. Ginsenoside Re significantly reduced cardiac damage caused by ferroptosis during myocardial ischemia/reperfusion injury and glutathione decline. To determine how Ginsenoside Re regulated ferroptosis, we isolated exosomes from VEGFR2+ endothelial progenitor cells after ischemia/reperfusion injury and performed miRNA profiling to screen the miRNAs aberrantly expressed in the process of myocardial ischemia/reperfusion injury and ginsenoside Re treatment. We identified that miR-144-3p was upregulated in myocardial ischemia/reperfusion injury by luciferase report and qRT-PCR. We further confirmed that the solute carrier family 7 member 11 (SLC7A11) was the target gene of miR-144-3p by database analysis and western blot. In comparison with ferropstatin-1, a ferroptosis inhibitor, in vivo studies confirmed that ferropstatin-1 also diminished myocardial ischemia/reperfusion injury induced cardiac function damage. CONCLUSION We demonstrated that ginsenoside Re attenuates myocardial ischemia/reperfusion induced ferroptosis via miR-144-3p/SLC7A11.
Collapse
Affiliation(s)
- Jian Ye
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Tian-Jiao Lyu
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Ling-Yan Li
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Ying Liu
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Hong Zhang
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Xu Wang
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Xin Xi
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China
| | - Zong-Jun Liu
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China; Department of Cardiology, Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Shanghai 200062, People's Republic of China.
| | - Jun-Qing Gao
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, People's Republic of China.
| |
Collapse
|
17
|
Yahiya YI, Hadi NR, Abu Raghif A, AL Habooby NGS. Protective effect of IAXO-102 on renal ischemia-reperfusion injury in rats. J Med Life 2023; 16:623-630. [PMID: 37305825 PMCID: PMC10251395 DOI: 10.25122/jml-2022-0280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/06/2023] [Indexed: 06/13/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) is a common cause of kidney damage, characterized by oxidative stress and inflammation. In this study, we investigated the potential protective effects of IAXO-102, a chemical compound, on experimentally induced IRI in male rats. The bilateral renal IRI model was used, with 24 adult male rats randomly divided into four groups (N=6): sham group (laparotomy without IRI induction), control group (laparotomy plus bilateral IRI for 30 minutes followed by 2 hours of reperfusion), vehicle group (same as control but pre-injected with the vehicle), and treatment group (similar to control but pre-injected with IAXO-102). We measured several biomarkers involved in IRI pathophysiology using enzyme-linked immunosorbent assay (ELISA), including High mobility group box1 (HMGB1), nuclear factor kappa b-p65 (NF-κB p65), interleukin beta-1 (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), 8-isoprostane, Bcl-2 associated X protein (BAX), heat shock protein 27 (HSP27), and Bcl-2. Statistical analysis was performed using one-way ANOVA and Tukey post hoc tests. Our results showed that IAXO-102 significantly improved kidney function, reduced histological alterations, and decreased the inflammatory response (IL-1, IL-6, and TNF) caused by IRI. IAXO-102 also decreased apoptosis by reducing pro-apoptotic Bax and increasing anti-apoptotic Bcl-2 without impacting HSP27. In conclusion, our findings suggest that IAXO-102 had a significant protective effect against IRI damage in the kidneys.
Collapse
Affiliation(s)
- Yahiya Ibrahim Yahiya
- Department of Pharmacology, Faculty of Pharmacy, University of Alkafeel, Najaf, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Ahmed Abu Raghif
- Deptartment of Pharmacology, College of Medicine, Al Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
18
|
Alam A, Smith SC, Gobalakrishnan S, McGinn M, Yakovlev VA, Rabender CS. Uncoupled nitric oxide synthase activity promotes colorectal cancer progression. Front Oncol 2023; 13:1165326. [PMID: 36998441 PMCID: PMC10046306 DOI: 10.3389/fonc.2023.1165326] [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: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Increased levels of reactive oxygen/nitrogen species are one hallmark of chronic inflammation contributing to the activation of pro-inflammatory/proliferative pathways. In the cancers analyzed, the tetrahydrobiopterin:dihydrobiopterin ratio is lower than that of the corresponding normal tissue, leading to an uncoupled nitric oxide synthase activity and increased generation of reactive oxygen/nitrogen species. Previously, we demonstrated that prophylactic treatment with sepiapterin, a salvage pathway precursor of tetrahydrobiopterin, prevents dextran sodium sulfate-induced colitis in mice and associated azoxymethane-induced colorectal cancer. Herein, we report that increasing the tetrahydrobiopterin:dihydrobiopterin ratio and recoupling nitric oxide synthase with sepiapterin in the colon cancer cell lines, HCT116 and HT29, inhibit their proliferation and enhance cell death, in part, by Akt/GSK-3β-mediated downregulation of β-catenin. Therapeutic oral gavage with sepiapterin of mice bearing azoxymethane/dextran sodium sulfate-induced colorectal cancer decreased metabolic uptake of [18F]-fluorodeoxyglucose and enhanced apoptosis nine-fold in these tumors. Immunohistochemical analysis of both mouse and human tissues indicated downregulated expression of key enzymes in tetrahydrobiopterin biosynthesis in the colorectal cancer tumors. Human stage 1 colon tumors exhibited a significant decrease in the expression of quinoid dihydropteridine reductase, a key enzyme involved in recycling tetrahydrobiopterin suggesting a potential mechanism for the reduced tetrahydrobiopterin:dihydrobiopterin ratio in these tumors. In summary, sepiapterin treatment of colorectal cancer cells increases the tetrahydrobiopterin:dihydrobiopterin ratio, recouples nitric oxide synthase, and reduces tumor growth. We conclude that nitric oxide synthase coupling may provide a useful therapeutic target for treating patients with colorectal cancer.
Collapse
Affiliation(s)
- Asim Alam
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Steven C. Smith
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Mina McGinn
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Vasily A. Yakovlev
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Christopher S. Rabender
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
19
|
Madokoro Y, Kamikokuryo C, Niiyama S, Ito T, Hara S, Ichinose H, Kakihana Y. Early ascorbic acid administration prevents vascular endothelial cell damage in septic mice. Front Pharmacol 2022; 13:929448. [PMID: 36278212 PMCID: PMC9582851 DOI: 10.3389/fphar.2022.929448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Oxidation of BH4, a cofactor of nitric oxide synthase (NOS), produces reactive oxygen species (ROS) through uncoupling of NOS and affects vascular endothelial dysfunction. Ascorbic acid (AsA) inhibits the oxidation of BH4 and reduces ROS. However, the kinetic changes of BH4 in sepsis and its effect on the kinetic changes in AsA administration therapy, as well as the appropriate timing of AsA administration for AsA therapy to be effective, are unclear. Mice with sepsis, induced by cecal ligation and puncture (CLP), were examined for the effect of AsA administration (200 mg/kg) on vascular endothelial cell dysfunction at two administration timings: early group (AsA administered immediately after CLP) and late group (AsA administered 12 h after CLP). Survival rates were compared between the early and late administration groups, and vascular endothelial cell damage, indicated by the dihydrobiopterin/tetrahydrobiopterin ratio, serum syndecan-1, and endothelial nitric oxide synthase, as well as liver damage, were examined. The early group showed significantly improved survival compared to the non-treatment group (p < 0.05), while the late group showed no improved survival compared to the non-treatment group. Compared to the non-treated group, the early AsA group showed less oxidation of BH4 in sepsis. Syndecan1, a marker of vascular endothelial cell damage, was less elevated and organ damage was reduced in the early AsA-treated group. In septic mice, early AsA administration immediately after CLP may protect vascular endothelial cells by inhibiting BH4 oxidation, thereby reducing organ dysfunction and improving survival.
Collapse
Affiliation(s)
- Yutaro Madokoro
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Chinatsu Kamikokuryo
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shuhei Niiyama
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takashi Ito
- Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Hara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Ichinose
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yasuyuki Kakihana
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- *Correspondence: Yasuyuki Kakihana,
| |
Collapse
|
20
|
Mustapha S, Azemi AK, Wan Ahmad WAN, Rasool AHG, Mustafa MR, Mokhtar SS. Inhibition of Endoplasmic Reticulum Stress Improves Acetylcholine-Mediated Relaxation in the Aorta of Type-2 Diabetic Rats. Molecules 2022; 27:5107. [PMID: 36014347 PMCID: PMC9413505 DOI: 10.3390/molecules27165107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
Endoplasmic reticulum (ER) stress contributes to insulin resistance and macro- and microvascular complications associated with diabetes. This study aimed to evaluate the effect of ER stress inhibition on endothelial function in the aorta of type-2 diabetic rats. Type-2 diabetes was developed in male Sprague-Dawley rats using a high-fat diet and low-dose streptozotocin. Rat aortic tissues were harvested to study endothelial-dependent relaxation. The mechanisms for acetylcholine-mediated relaxation were investigated using pharmacological blockers, Western blotting, oxidative stress, and inflammatory markers. Acetylcholine-mediated relaxation was diminished in the aorta of diabetic rats compared to control rats; supplementation with TUDCA improved relaxation. In the aortas of control and diabetic rats receiving TUDCA, the relaxation was mediated via eNOS/PI3K/Akt, NAD(P)H, and the KATP channel. In diabetic rats, acetylcholine-mediated relaxation involved eNOS/PI3K/Akt and NAD(P)H, but not the KATP channel. The expression of ER stress markers was upregulated in the aorta of diabetic rats and reduced with TUDCA supplementation. The expression of eNOS and Akt were lower in diabetic rats but were upregulated after supplementation with TUDCA. The levels of MDA, IL-6, and SOD activity were higher in the aorta of the diabetic rats compared to control rats. This study demonstrated that endothelial function was impaired in diabetes, however, supplementation with TUDCA improved the function via eNOS/Akt/PI3K, NAD(P)H, and the KATP channel. The improvement of endothelial function was associated with increased expressions of eNOS and Akt. Thus, ER stress plays a crucial role in the impairment of endothelial-dependent relaxation. Mitigating ER stress could be a potential strategy for improving endothelial dysfunction in type-2 diabetes.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Selangor, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| |
Collapse
|
21
|
Petraina A, Nogales C, Krahn T, Mucke H, Lüscher TF, Fischmeister R, Kass DA, Burnett JC, Hobbs AJ, Schmidt HHHW. Cyclic GMP modulating drugs in cardiovascular diseases: mechanism-based network pharmacology. Cardiovasc Res 2022; 118:2085-2102. [PMID: 34270705 PMCID: PMC9302891 DOI: 10.1093/cvr/cvab240] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Mechanism-based therapy centred on the molecular understanding of disease-causing pathways in a given patient is still the exception rather than the rule in medicine, even in cardiology. However, recent successful drug developments centred around the second messenger cyclic guanosine-3'-5'-monophosphate (cGMP), which is regulating a number of cardiovascular disease modulating pathways, are about to provide novel targets for such a personalized cardiovascular therapy. Whether cGMP breakdown is inhibited or cGMP synthesis is stimulated via guanylyl cyclases or their upstream regulators in different cardiovascular disease phenotypes, the outcomes seem to be so far uniformly protective. Thus, a network of cGMP-modulating drugs has evolved that act in a mechanism-based, possibly causal manner in a number of cardiac conditions. What remains a challenge is the detection of cGMPopathy endotypes amongst cardiovascular disease phenotypes. Here, we review the growing clinical relevance of cGMP and provide a glimpse into the future on how drugs interfering with this pathway may change how we treat and diagnose cardiovascular diseases altogether.
Collapse
Affiliation(s)
- Alexandra Petraina
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Thomas Krahn
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Hermann Mucke
- H.M. Pharma Consultancy, Enenkelstrasse 28/32, A-1160, Vienna, Austria
| | - Thomas F Lüscher
- Royal Brompton & Harefield Hospitals, Heart Division and National Heart and Lung Institute, Guy Scadding Building, Imperial College, Dovehouse Street London SW3 6LY, United Kingdom
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistreet 12, CH-8952 Schlieren, Switzerland
| | - Rodolphe Fischmeister
- INSERM UMR-S 1180, Faculty of Pharmacy, Université Paris-Saclay, F-92296 Châtenay-Malabry, France
| | - David A Kass
- Division of Cardiology, Department of Medicine, Ross Research Building, Rm 858, Johns Hopkins Medical Institutions, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - John C Burnett
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, London, UK
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
22
|
Das BB. Therapeutic Approaches in Heart Failure with Preserved Ejection Fraction (HFpEF) in Children: Present and Future. Paediatr Drugs 2022; 24:235-246. [PMID: 35501560 DOI: 10.1007/s40272-022-00508-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 12/29/2022]
Abstract
For a long time, pediatric heart failure (HF) with preserved systolic function (HFpEF) has been noted in patients with cardiomyopathies and congenital heart disease. HFpEF is infrequently reported in children and instead of using the HFpEF terminology the HF symptoms are attributed to diastolic dysfunction. Identifying HFpEF in children is challenging because of heterogeneous etiologies and unknown pathophysiological mechanisms. Advances in echocardiography and cardiac magnetic resonance imaging techniques have further increased our understanding of HFpEF in children. However, the literature does not describe the incidence, etiology, clinical features, and treatment of HFpEF in children. At present, treatment of HFpEF in children is extrapolated from clinical trials in adults. There are significant differences between pediatric and adult HF with reduced ejection fraction, supported by a lack of adequate response to adult HF therapies. Evidence-based clinical trials in children are still not available because of the difficulty of conducting trials with a limited number of pediatric patients with HF. The treatment of HFpEF in children is based upon the clinician's experience, and the majority of children receive off-level medications. There are significant differences between pediatric and adult HFpEF pharmacotherapies in many areas, including side-effect profiles, underlying pathophysiologies, the β-receptor physiology, and pharmacokinetics and pharmacodynamics. This review describes the present and future treatments for children with HFpEF compared with adults. This review also highlights the need to urgently test new therapies in children with HFpEF to demonstrate the safety and efficacy of drugs and devices with proven benefits in adults.
Collapse
Affiliation(s)
- Bibhuti B Das
- Department of Pediatrics, Division of Cardiology, University of Mississippi Medical Center, 2500 N State St., Jackson, MS, 39216, USA.
| |
Collapse
|
23
|
Liu Z, Dong N, Hui H, Wang Y, Liu F, Xu L, Liu M, Rao Z, Yuan Z, Shang Y, Feng J, Cai Z, Li F. Endothelial cell-derived tetrahydrobiopterin prevents aortic valve calcification. Eur Heart J 2022; 43:1652-1664. [PMID: 35139535 DOI: 10.1093/eurheartj/ehac037] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Tetrahydrobiopterin (BH4) is a critical determinant of the biological function of endothelial nitric oxide synthase. The present study was to investigate the role of valvular endothelial cell (VEC)-derived BH4 in aortic valve calcification. METHODS AND RESULTS Plasma and aortic valve BH4 concentrations and the BH4:BH2 ratio were significantly lower in calcific aortic valve disease patients than in controls. There was a significant decrease of the two key enzymes of BH4 biosynthesis, guanosine 5'-triphosphate cyclohydrolase I (GCH1) and dihydrofolate reductase (DHFR), in calcified aortic valves compared with the normal ones. Endothelial cell-specific deficiency of Gch1 in Apoe-/- (Apoe-/-Gch1fl/flTie2Cre) mice showed a marked increase in transvalvular peak jet velocity, calcium deposition, runt-related transcription factor 2 (Runx2), dihydroethidium (DHE), and 3-nitrotyrosine (3-NT) levels in aortic valve leaflets compared with Apoe-/-Gch1fl/fl mice after a 24-week western diet (WD) challenge. Oxidized LDL (ox-LDL) induced osteoblastic differentiation of valvular interstitial cells (VICs) co-cultured with either si-GCH1- or si-DHFR-transfected VECs, while the effects could be abolished by BH4 supplementation. Deficiency of BH4 in VECs caused peroxynitrite formation increase and 3-NT protein increase under ox-LDL stimulation in VICs. SIN-1, the peroxynitrite generator, significantly up-regulated alkaline phosphatase (ALP) and Runx2 expression in VICs via tyrosine nitration of dynamin-related protein 1 (DRP1) at Y628. Finally, folic acid (FA) significantly attenuated aortic valve calcification in WD-fed Apoe-/- mice through increasing DHFR and salvaging BH4 biosynthesis. CONCLUSION The reduction in endothelial-dependent BH4 levels promoted peroxynitrite formation, which subsequently resulted in DRP1 tyrosine nitration and osteoblastic differentiation of VICs, thereby leading to aortic valve calcification. Supplementation of FA in diet attenuated hypercholesterolaemia-induced aortic valve calcification by salvaging BH4 bioavailability.
Collapse
Affiliation(s)
- Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Haipeng Hui
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 28# Fuxing Road, Beijing 100853, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Fayun Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Ming Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Zhenqi Rao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| | - Zhen Yuan
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Yuqiang Shang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, China
| | - Jun Feng
- Department of Emergency and Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Zhejun Cai
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China
| |
Collapse
|
24
|
Villaume WA. Marginal BH4 Deficiencies, iNOS, and Self-perpetuating Oxidative Stress in Post-acute Sequelae of Covid-19. Med Hypotheses 2022; 163:110842. [PMID: 35431403 PMCID: PMC9006446 DOI: 10.1016/j.mehy.2022.110842] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 11/30/2022]
Abstract
The treatment of post-acute sequelae of Covid-19 (PASC) has been informed primarily by symptomatic parallels with other chronic inflammatory syndromes. This manuscript takes a more systemic approach by examining how a marginal deficiency of tetrahydrobiopterin (BH4) resulting from mutations of the GCH1 (GTP cyclohydrolase 1) gene may result in the uncoupling of inducible Nitric Oxide Synthase (iNOS) early in the initial response of the innate immune system to SARS-CoV-2. The resulting production of superoxide instead of nitric oxide leads to a self-perpetuating cycle of oxidative stress with the potential to impair numerous metabolic processes and damage multiple organ systems. This marginal deficiency of BH4 may be exhibited by 30% or more of the patient population that have heterozygous or homozygous mutations of GCH1. As the cycle of oxidative stress continues, there is less BH4 available for other metabolic needs such as 1) resisting increased ferroptosis with its damage to organs, and 2) regulating the deactivation of the hyperinflammatory state. Finally, possible steps are proposed for clinical treatment of the hypothesized oxidative stress involved with PASC.
Collapse
|
25
|
El-Mahdy MA, Ewees MG, Eid MS, Mahgoup EM, Khaleel SA, Zweier JL. Electronic Cigarette Exposure Causes Vascular Endothelial Dysfunction Due to NADPH Oxidase Activation and eNOS Uncoupling. Am J Physiol Heart Circ Physiol 2022; 322:H549-H567. [PMID: 35089811 PMCID: PMC8917923 DOI: 10.1152/ajpheart.00460.2021] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We recently reported a mouse model of chronic electronic cigarette (e-cig) exposure-induced cardiovascular pathology, where long-term exposure to e-cig vape (ECV) induces cardiac abnormalities, impairment of endothelial function, and systemic hypertension. Here, we delineate the underlying mechanisms of ECV-induced vascular endothelial dysfunction (VED), a central trigger of cardiovascular disease. C57/BL6 male mice were exposed to ECV generated from e-cig liquid containing 0, 6, or 24 mg/ml nicotine for 16 and 60 weeks. Time-dependent elevation in blood pressure and systemic vascular resistance were observed, along with an impairment of acetylcholine-induced aortic relaxation in ECV-exposed mice, compared to air-exposed control. Decreased intravascular nitric oxide (NO) levels and increased superoxide generation with elevated 3-nitrotyrosine levels in the aorta of ECV-exposed mice were observed, indicating that ECV-induced superoxide reacts with NO to generate cytotoxic peroxynitrite. Exposure increased NADPH oxidase expression, supporting its role in ECV-induced superoxide generation. Downregulation of endothelial nitric oxide synthase (eNOS) expression and Akt-dependent eNOS phosphorylation occurred in the aorta of ECV-exposed mice, indicating that exposure inhibited de novo NO synthesis. Following ECV exposure, the critical NOS cofactor tetrahydrobiopterin was decreased, with a concomitant loss of its salvage enzyme, dihydrofolate reductase. NADPH oxidase and NOS inhibitors abrogated ECV-induced superoxide generation in the aorta of ECV exposed mice. Together, our data demonstrate that ECV exposure activates NADPH oxidase and uncouples eNOS, causing a vicious cycle of superoxide generation and vascular oxidant stress that triggers VED and hypertension with predisposition to other cardiovascular disease.
Collapse
Affiliation(s)
- Mohamed A El-Mahdy
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Mohamed G Ewees
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Mahmoud S Eid
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Elsayed M Mahgoup
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Sahar A Khaleel
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Jay L Zweier
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
26
|
Maslov LN, Popov SV, Mukhomedzyanov AV, Naryzhnaya NV, Voronkov NS, Ryabov VV, Boshchenko AA, Khaliulin I, Prasad NR, Fu F, Pei JM, Logvinov SV, Oeltgen PR. Reperfusion Cardiac Injury: Receptors and the Signaling Mechanisms. Curr Cardiol Rev 2022; 18:63-79. [PMID: 35422224 PMCID: PMC9896422 DOI: 10.2174/1573403x18666220413121730] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/01/2022] [Accepted: 01/10/2022] [Indexed: 11/22/2022] Open
Abstract
It has been documented that Ca2+ overload and increased production of reactive oxygen species play a significant role in reperfusion injury (RI) of cardiomyocytes. Ischemia/reperfusion induces cell death as a result of necrosis, necroptosis, apoptosis, and possibly autophagy, pyroptosis and ferroptosis. It has also been demonstrated that the NLRP3 inflammasome is involved in RI of the heart. An increase in adrenergic system activity during the restoration of coronary perfusion negatively affected cardiac resistance to RI. Toll-like receptors are involved in RI of the heart. Angiotensin II and endothelin-1 aggravated ischemic/reperfusion injury of the heart. Activation of neutrophils, monocytes, CD4+ T-cells and platelets contributes to cardiac ischemia/reperfusion injury. Our review outlines the role of these factors in reperfusion cardiac injury.
Collapse
Affiliation(s)
- Leonid N. Maslov
- Address correspondence to this author at the Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Kyevskskaya 111A, 634012 Tomsk, Russia; Tel. +7 3822 262174; E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kobayashi K, Liu C, Jonas RA, Ishibashi N. The Current Status of Neuroprotection in Congenital Heart Disease. CHILDREN 2021; 8:children8121116. [PMID: 34943311 PMCID: PMC8700367 DOI: 10.3390/children8121116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022]
Abstract
Neurological deficits are a serious and common sequelae of congenital heart disease (CHD). While their underlying mechanisms have not been fully characterized, their manifestations are well-known and understood to persist through adulthood. Development of therapies to address or prevent these deficits are critical to attenuate future morbidity and improve quality of life. In this review, we aim to summarize the current status of neuroprotective therapy in CHD. Through an exploration of present research in the pre-operative, intra-operative, and post-operative phases of patient management, we will describe existing clinical and bench efforts as well as current endeavors underway within this research area.
Collapse
Affiliation(s)
- Kei Kobayashi
- Center for Neuroscience Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC 20010, USA; (K.K.); (C.L.); (R.A.J.)
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Christopher Liu
- Center for Neuroscience Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC 20010, USA; (K.K.); (C.L.); (R.A.J.)
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Richard A. Jonas
- Center for Neuroscience Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC 20010, USA; (K.K.); (C.L.); (R.A.J.)
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC 20010, USA
- School of Medicine and Health Science, George Washington University, Washington, DC 20052, USA
| | - Nobuyuki Ishibashi
- Center for Neuroscience Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC 20010, USA; (K.K.); (C.L.); (R.A.J.)
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC 20010, USA
- School of Medicine and Health Science, George Washington University, Washington, DC 20052, USA
- Correspondence:
| |
Collapse
|
28
|
Ricci A, Di Pierro E, Marcacci M, Ventura P. Mechanisms of Neuronal Damage in Acute Hepatic Porphyrias. Diagnostics (Basel) 2021; 11:diagnostics11122205. [PMID: 34943446 PMCID: PMC8700611 DOI: 10.3390/diagnostics11122205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/12/2023] Open
Abstract
Porphyrias are a group of congenital and acquired diseases caused by an enzymatic impairment in the biosynthesis of heme. Depending on the specific enzyme involved, different types of porphyrias (i.e., chronic vs. acute, cutaneous vs. neurovisceral, hepatic vs. erythropoietic) are described, with different clinical presentations. Acute hepatic porphyrias (AHPs) are characterized by life-threatening acute neuro-visceral crises (acute porphyric attacks, APAs), featuring a wide range of neuropathic (central, peripheral, autonomic) manifestations. APAs are usually unleashed by external "porphyrinogenic" triggers, which are thought to cause an increased metabolic demand for heme. During APAs, the heme precursors δ-aminolevulinic acid (ALA) and porphobilinogen (PBG) accumulate in the bloodstream and urine. Even though several hypotheses have been developed to explain the protean clinical picture of APAs, the exact mechanism of neuronal damage in AHPs is still a matter of debate. In recent decades, a role has been proposed for oxidative damage caused by ALA, mitochondrial and synaptic ALA toxicity, dysfunction induced by relative heme deficiency on cytochromes and other hemeproteins (i.e., nitric oxide synthases), pyridoxal phosphate functional deficiency, derangements in the metabolic pathways of tryptophan, and other factors. Since the pathway leading to the biosynthesis of heme is inscribed into a complex network of interactions, which also includes some fundamental processes of basal metabolism, a disruption in any of the steps of this pathway is likely to have multiple pathogenic effects. Here, we aim to provide a comprehensive review of the current evidence regarding the mechanisms of neuronal damage in AHPs.
Collapse
Affiliation(s)
- Andrea Ricci
- Internal Medicine Unit, Department of Medical and Surgical Science for Children and Adults, University of Modena e Reggio Emilia, 41124 Modena, Italy; (A.R.); (M.M.)
| | - Elena Di Pierro
- Dipartimento di Medicina Interna, Fondazione IRCSS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Matteo Marcacci
- Internal Medicine Unit, Department of Medical and Surgical Science for Children and Adults, University of Modena e Reggio Emilia, 41124 Modena, Italy; (A.R.); (M.M.)
| | - Paolo Ventura
- Internal Medicine Unit, Department of Medical and Surgical Science for Children and Adults, University of Modena e Reggio Emilia, 41124 Modena, Italy; (A.R.); (M.M.)
- Correspondence: ; Tel.: +39-059-4225-542
| |
Collapse
|
29
|
Fujihara Y, Kodo Y, Miyoshi SI, Watanabe R, Toyoda H, Mankura M, Kabuto H, Takayama F. Spirulina platensis and its ingredient biopterin glucoside improved insulin sensitivity in non-alcoholic steatohepatitis model. J Clin Biochem Nutr 2021; 69:151-157. [PMID: 34616107 PMCID: PMC8482380 DOI: 10.3164/jcbn.20-201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/25/2020] [Indexed: 11/25/2022] Open
Abstract
Non-alcoholic steatohepatitis is the chronic liver disease leading to cirrhosis and cancer and its prevalence is increasing. Some agents are under clinical trials for non-alcoholic steatohepatitis treatment. We previously reported Spirulina (Arthrospira) platensis effectively prevented non-alcoholic steatohepatitis progression in our model rats. The contribution of phycocyanin, an ingredient of Spirulina (Arthrospira) platensis, was limited. We, therefore, have looked for more active components of Spirulina (Arthrospira) platensis. In this study, we pursued the effect of biopterin glucoside, another bioactive ingredient of Spirulina (Arthrospira) platensis. We found Spirulina (Arthrospira) platensis and biopterin glucoside oral administrations effectively alleviated oxidative stress, inflammation and insulin signal failure, and prevented fibroblast growth factor 21 gene overexpression in non-alcoholic steatohepatitis rat livers. We concluded biopterin glucoside is a major component of Spirulina (Arthrospira) platensis action.
Collapse
Affiliation(s)
- Yuri Fujihara
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Yasumasa Kodo
- Spirulina BioLab. Co., Ltd., 1-13-6 Nishinakajima, Yodogawa-ku, Osaka 532-0011, Japan
| | - Shin-Ichi Miyoshi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Ritsuko Watanabe
- Okayama Kyoritsu General Hospital, 8-10 Akasakahonmachi, Naka-ku, Okayama 703-8288, Japan
| | - Hiroshi Toyoda
- Okayama Kyoritsu General Hospital, 8-10 Akasakahonmachi, Naka-ku, Okayama 703-8288, Japan
| | - Mitsumasa Mankura
- Kurashiki Sakuyo University, 3515 Tamashima Nagao, Kurashiki, Okayama 710-0292, Japan
| | - Hideaki Kabuto
- Kagawa Prefectural College of Health Sciences, 281-1 Murechohara, Takamatsu, Kagawa 761-0123, Japan
| | - Fusako Takayama
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
30
|
Toral M, de la Fuente-Alonso A, Campanero MR, Redondo JM. The NO signalling pathway in aortic aneurysm and dissection. Br J Pharmacol 2021; 179:1287-1303. [PMID: 34599830 DOI: 10.1111/bph.15694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Recent studies have shown that NO is a central mediator in diseases associated with thoracic aortic aneurysm, such as Marfan syndrome. The progressive dilation of the aorta in thoracic aortic aneurysm ultimately leads to aortic dissection. Unfortunately, current medical treatments have neither halt aortic enlargement nor prevented rupture, leaving surgical repair as the only effective treatment. There is therefore a pressing need for effective therapies to delay or even avoid the need for surgical repair in thoracic aortic aneurysm patients. Here, we summarize the mechanisms through which NO signalling dysregulation causes thoracic aortic aneurysm, particularly in Marfan syndrome. We discuss recent advances based on the identification of new Marfan syndrome mediators related to pathway overactivation that represent potential disease biomarkers. Likewise, we propose iNOS, sGC and PRKG1, whose pharmacological inhibition reverses aortopathy in Marfan syndrome mice, as targets for therapeutic intervention in thoracic aortic aneurysm and are candidates for clinical trials.
Collapse
Affiliation(s)
- Marta Toral
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andrea de la Fuente-Alonso
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel R Campanero
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
31
|
Ow CPC, Trask-Marino A, Betrie AH, Evans RG, May CN, Lankadeva YR. Targeting Oxidative Stress in Septic Acute Kidney Injury: From Theory to Practice. J Clin Med 2021; 10:jcm10173798. [PMID: 34501245 PMCID: PMC8432047 DOI: 10.3390/jcm10173798] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Sepsis is the leading cause of acute kidney injury (AKI) and leads to increased morbidity and mortality in intensive care units. Current treatments for septic AKI are largely supportive and are not targeted towards its pathophysiology. Sepsis is commonly characterized by systemic inflammation and increased production of reactive oxygen species (ROS), particularly superoxide. Concomitantly released nitric oxide (NO) then reacts with superoxide, leading to the formation of reactive nitrogen species (RNS), predominantly peroxynitrite. Sepsis-induced ROS and RNS can reduce the bioavailability of NO, mediating renal microcirculatory abnormalities, localized tissue hypoxia and mitochondrial dysfunction, thereby initiating a propagating cycle of cellular injury culminating in AKI. In this review, we discuss the various sources of ROS during sepsis and their pathophysiological interactions with the immune system, microcirculation and mitochondria that can lead to the development of AKI. We also discuss the therapeutic utility of N-acetylcysteine and potential reasons for its efficacy in animal models of sepsis, and its inefficacy in ameliorating oxidative stress-induced organ dysfunction in human sepsis. Finally, we review the pre-clinical studies examining the antioxidant and pleiotropic actions of vitamin C that may be of benefit for mitigating septic AKI, including future implications for clinical sepsis.
Collapse
Affiliation(s)
- Connie P. C. Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka 564-8565, Japan
| | - Anton Trask-Marino
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
| | - Ashenafi H. Betrie
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roger G. Evans
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Clive N. May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Yugeesh R. Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
- Correspondence: ; Tel.: +61-3-8344-0417; Fax: +61-3-9035-3107
| |
Collapse
|
32
|
Novel Insights into the Molecular Mechanisms of Ischemia/Reperfusion Injury in Kidney Transplantation. TRANSPLANTOLOGY 2021. [DOI: 10.3390/transplantology2020018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ischemia reperfusion injury (IRI) is one of the most important mechanisms involved in delayed or reduced graft function after kidney transplantation. It is a complex pathophysiological process, followed by a pro-inflammatory response that enhances the immunogenicity of the graft and the risk of acute rejection. Many biologic processes are involved in its development, such as transcriptional reprogramming, the activation of apoptosis and cell death, endothelial dysfunction and the activation of the innate and adaptive immune response. Recent evidence has highlighted the importance of complement activation in IRI cascade, which expresses a pleiotropic action on tubular cells, on vascular cells (pericytes and endothelial cells) and on immune system cells. The effects of IRI in the long term lead to interstitial fibrosis and tubular atrophy, which contribute to chronic graft dysfunction and subsequently graft failure. Furthermore, several metabolic alterations occur upon IRI. Metabolomic analyses of IRI detected a “metabolic profile” of this process, in order to identify novel biomarkers that may potentially be useful for both early diagnosis and monitoring the therapeutic response. The aim of this review is to update the most relevant molecular mechanisms underlying IRI, and also to discuss potential therapeutic targets in future clinical practice.
Collapse
|
33
|
Role of Oxidative Stress in Reperfusion following Myocardial Ischemia and Its Treatments. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6614009. [PMID: 34055195 PMCID: PMC8149218 DOI: 10.1155/2021/6614009] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/21/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Myocardial ischemia is a disease with high morbidity and mortality, for which reperfusion is currently the standard intervention. However, the reperfusion may lead to further myocardial damage, known as myocardial ischemia/reperfusion injury (MI/RI). Oxidative stress is one of the most important pathological mechanisms in reperfusion injury, which causes apoptosis, autophagy, inflammation, and some other damage in cardiomyocytes through multiple pathways, thus causing irreversible cardiomyocyte damage and cardiac dysfunction. This article reviews the pathological mechanisms of oxidative stress involved in reperfusion injury and the interventions for different pathways and targets, so as to form systematic treatments for oxidative stress-induced myocardial reperfusion injury and make up for the lack of monotherapy.
Collapse
|
34
|
Aortic disease in Marfan syndrome is caused by overactivation of sGC-PRKG signaling by NO. Nat Commun 2021; 12:2628. [PMID: 33976159 PMCID: PMC8113458 DOI: 10.1038/s41467-021-22933-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
Thoracic aortic aneurysm, as occurs in Marfan syndrome, is generally asymptomatic until dissection or rupture, requiring surgical intervention as the only available treatment. Here, we show that nitric oxide (NO) signaling dysregulates actin cytoskeleton dynamics in Marfan Syndrome smooth muscle cells and that NO-donors induce Marfan-like aortopathy in wild-type mice, indicating that a marked increase in NO suffices to induce aortopathy. Levels of nitrated proteins are higher in plasma from Marfan patients and mice and in aortic tissue from Marfan mice than in control samples, indicating elevated circulating and tissue NO. Soluble guanylate cyclase and cGMP-dependent protein kinase are both activated in Marfan patients and mice and in wild-type mice treated with NO-donors, as shown by increased plasma cGMP and pVASP-S239 staining in aortic tissue. Marfan aortopathy in mice is reverted by pharmacological inhibition of soluble guanylate cyclase and cGMP-dependent protein kinase and lentiviral-mediated Prkg1 silencing. These findings identify potential biomarkers for monitoring Marfan Syndrome in patients and urge evaluation of cGMP-dependent protein kinase and soluble guanylate cyclase as therapeutic targets. Aortic aneurysm and dissection, the major problem linked to Marfan syndrome (MFS), lacks effective pharmacological treatment. Here, the authors show that the NO pathway is overactivated in MFS and that inhibition of guanylate cyclase and cGMP-dependent protein kinase reverts MFS aortopathy in mice.
Collapse
|
35
|
Mustapha S, Mohammed M, Azemi AK, Yunusa I, Shehu A, Mustapha L, Wada Y, Ahmad MH, Ahmad WANW, Rasool AHG, Mokhtar SS. Potential Roles of Endoplasmic Reticulum Stress and Cellular Proteins Implicated in Diabesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8830880. [PMID: 33995826 PMCID: PMC8099518 DOI: 10.1155/2021/8830880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
The role of the endoplasmic reticulum (ER) has evolved from protein synthesis, processing, and other secretory pathways to forming a foundation for lipid biosynthesis and other metabolic functions. Maintaining ER homeostasis is essential for normal cellular function and survival. An imbalance in the ER implied stressful conditions such as metabolic distress, which activates a protective process called unfolded protein response (UPR). This response is activated through some canonical branches of ER stress, i.e., the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6). Therefore, chronic hyperglycemia, hyperinsulinemia, increased proinflammatory cytokines, and free fatty acids (FFAs) found in diabesity (a pathophysiological link between obesity and diabetes) could lead to ER stress. However, limited data exist regarding ER stress and its association with diabesity, particularly the implicated proteins and molecular mechanisms. Thus, this review highlights the role of ER stress in relation to some proteins involved in diabesity pathogenesis and provides insight into possible pathways that could serve as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Pulau Pinang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, College of Pharmacy, Columbia, SC, USA
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna, Nigeria
| | - Yusuf Wada
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mubarak Hussaini Ahmad
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
- School of Pharmacy Technician, Aminu Dabo College of Health Sciences and Technology, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
36
|
McCarty MF. Nutraceutical, Dietary, and Lifestyle Options for Prevention and Treatment of Ventricular Hypertrophy and Heart Failure. Int J Mol Sci 2021; 22:ijms22073321. [PMID: 33805039 PMCID: PMC8037104 DOI: 10.3390/ijms22073321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Although well documented drug therapies are available for the management of ventricular hypertrophy (VH) and heart failure (HF), most patients nonetheless experience a downhill course, and further therapeutic measures are needed. Nutraceutical, dietary, and lifestyle measures may have particular merit in this regard, as they are currently available, relatively safe and inexpensive, and can lend themselves to primary prevention as well. A consideration of the pathogenic mechanisms underlying the VH/HF syndrome suggests that measures which control oxidative and endoplasmic reticulum (ER) stress, that support effective nitric oxide and hydrogen sulfide bioactivity, that prevent a reduction in cardiomyocyte pH, and that boost the production of protective hormones, such as fibroblast growth factor 21 (FGF21), while suppressing fibroblast growth factor 23 (FGF23) and marinobufagenin, may have utility for preventing and controlling this syndrome. Agents considered in this essay include phycocyanobilin, N-acetylcysteine, lipoic acid, ferulic acid, zinc, selenium, ubiquinol, astaxanthin, melatonin, tauroursodeoxycholic acid, berberine, citrulline, high-dose folate, cocoa flavanols, hawthorn extract, dietary nitrate, high-dose biotin, soy isoflavones, taurine, carnitine, magnesium orotate, EPA-rich fish oil, glycine, and copper. The potential advantages of whole-food plant-based diets, moderation in salt intake, avoidance of phosphate additives, and regular exercise training and sauna sessions are also discussed. There should be considerable scope for the development of functional foods and supplements which make it more convenient and affordable for patients to consume complementary combinations of the agents discussed here. Research Strategy: Key word searching of PubMed was employed to locate the research papers whose findings are cited in this essay.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA
| |
Collapse
|
37
|
Wu Y, Ding Y, Ramprasath T, Zou MH. Oxidative Stress, GTPCH1, and Endothelial Nitric Oxide Synthase Uncoupling in Hypertension. Antioxid Redox Signal 2021; 34:750-764. [PMID: 32363908 PMCID: PMC7910417 DOI: 10.1089/ars.2020.8112] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Significance: Hypertension has major health consequences, which is associated with endothelial dysfunction. Endothelial nitric oxide synthase (eNOS)-produced nitric oxide (NO) signaling in the vasculature plays an important role in maintaining vascular homeostasis. Considering the importance of NO system, this review aims to provide a brief overview of the biochemistry of members of NO signaling, including GTPCH1 [guanosine 5'-triphosphate (GTP) cyclohydrolase 1], tetrahydrobiopterin (BH4), and eNOS. Recent Advances: Being NO signaling activators and regulators of eNOS signaling, BH4 treatment is getting widespread attention either as potential therapeutic agents or as preventive agents. Recent clinical trials also support that BH4 treatment could be considered a promising therapeutic in hypertension. Critical Issues: Under conditions of BH4 depletion, eNOS-generated superoxides trigger pathological events. Abnormalities in NO availability and BH4 deficiency lead to disturbed redox regulation causing pathological events. This disturbed signaling influences the development of systemic hypertension as well as pulmonary hypertension. Future Directions: Considering the importance of BH4 and NO to improve the translational significance, it is essential to continue research on this field to manipulate BH4 to increase the efficacy for treating hypertension. Thus, this review also examines the current state of knowledge on the effects of eNOS activators on preclinical models and humans to utilize this information for potential therapy.
Collapse
Affiliation(s)
- Yin Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Tharmarajan Ramprasath
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
38
|
Xu J, Wang C, Zhang X, Ouyang J, Zhang J. Serum folic acid levels and erectile dysfunction: A meta-analysis and systematic review. Andrologia 2021; 53:e14003. [PMID: 33550658 DOI: 10.1111/and.14003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 11/27/2022] Open
Abstract
The aim of this study was to assess the relationship between serum folic acid (FA) levels and erectile dysfunction (ED) through a meta-analysis. A research was conducted in MEDLINE via PubMed, Cochrane Library, EMBASE and Web of Science up to 22 November 2020 to identify studies related to FA and ED. Two authors independently screened the literature, evaluated methodological quality and extracted the data. We used RevMan5.3 and STATA 14.0 for meta-analysis. A total of six studies including 1,842 participants were included, and the results showed that the FA levels in the non-ED group were significantly higher than those in the ED group (MD = 3.37, 95% CI 1.49-5.52, p = 0.004). Subgroup analysis indicated that with the increase in ED severity, the difference in FA levels between groups was more obvious (MD: 1.99 vs. 4.63 vs. 5.63). The differences in FA levels between groups seem more significant in the younger group (MD = 4.87, 95% CI 2.58-6.89, p < 0.001) than in the older group (MD = 3.15, 95% CI 2.21-4.08, p < 0.001). In conclusion, FA deficiency is closely related to ED, and the degree of FA deficiency may reflect the severity of ED. In addition, the association seems to be more pronounced in the younger group.
Collapse
Affiliation(s)
- Jiangnan Xu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuefeng Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Ouyang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianglei Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
39
|
L-Arginine Reduces Nitro-Oxidative Stress in Cultured Cells with Mitochondrial Deficiency. Nutrients 2021; 13:nu13020534. [PMID: 33562042 PMCID: PMC7914615 DOI: 10.3390/nu13020534] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/26/2021] [Accepted: 02/03/2021] [Indexed: 12/16/2022] Open
Abstract
L-Arginine (L-ARG) supplementation has been suggested as a therapeutic option in several diseases, including Mitochondrial Encephalomyopathy, Lactic Acidosis, and Stroke-like syndrome (MELAS), arguably the most common mitochondrial disease. It is suggested that L-ARG, a nitric oxide (NO) precursor, can restore NO levels in blood vessels, improving cerebral blood flow. However, NO also participates in mitochondrial processes, such as mitochondrial biogenesis, the regulation of the respiratory chain, and oxidative stress. This study investigated the effects of L-ARG on mitochondrial function, nitric oxide synthesis, and nitro-oxidative stress in cell lines harboring the MELAS mitochondrial DNA (mtDNA) mutation (m.3243A>G). We evaluated mitochondrial enzyme activity, mitochondrial mass, NO concentration, and nitro-oxidative stress. Our results showed that m.3243A>G cells had increased NO levels and protein nitration at basal conditions. Treatment with L-ARG did not affect the mitochondrial function and mass but reduced the intracellular NO concentration and nitrated proteins in m.3243A>G cells. The same treatment led to opposite effects in control cells. In conclusion, we showed that the main effect of L-ARG was on protein nitration. Lowering protein nitration is probably involved in the mechanism related to L-ARG supplementation benefits in MELAS patients.
Collapse
|
40
|
Abstract
Neuroanatomic and functional studies show the paraventricular (PVN) of the hypothalamus to have a central role in the autonomic control that supports cardiovascular regulation. Direct and indirect projections from the PVN preautonomic neurons to the sympathetic preganglionic neurons in the spinal cord modulate sympathetic activity. The preautonomic neurons of the PVN adjust their level of activation in response to afferent signals arising from peripheral viscerosensory receptors relayed through the nucleus tractus solitarius. The prevailing sympathetic tone is a balance between excitatory and inhibitory influences that arises from the preautonomic PVN neurons. Under physiologic conditions, tonic sympathetic inhibition driven by a nitric oxide-γ-aminobutyric acid-mediated mechanism is dominant, but in pathologic situation such as heart failure there is a switch from inhibition to sympathoexcitation driven by glutamate and angiotensin II. Angiotensin II, reactive oxygen species, and hypoxia as a result of myocardial infarction/ischemia alter the tightly regulated posttranslational protein-protein interaction of CAPON (carboxy-terminal postsynaptic density protein ligand of neuronal nitric oxide synthase (NOS1)) and PIN (protein inhibitor of NOS1) signaling mechanism. Within the preautonomic neurons of the PVN, the disruption of CAPON and PIN signaling leads to a downregulation of NOS1 expression and reduced NO bioavailability. These data support the notion that CAPON-PIN dysregulation of NO bioavailability is a major contributor to the pathogenesis of sympathoexcitation in heart failure.
Collapse
Affiliation(s)
- Susan Pyner
- Department of Biosciences, Durham University, Durham, United Kingdom.
| |
Collapse
|
41
|
Boycott HE, Nguyen MN, Vrellaku B, Gehmlich K, Robinson P. Nitric Oxide and Mechano-Electrical Transduction in Cardiomyocytes. Front Physiol 2020; 11:606740. [PMID: 33384614 PMCID: PMC7770138 DOI: 10.3389/fphys.2020.606740] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
The ability§ of the heart to adapt to changes in the mechanical environment is critical for normal cardiac physiology. The role of nitric oxide is increasingly recognized as a mediator of mechanical signaling. Produced in the heart by nitric oxide synthases, nitric oxide affects almost all mechano-transduction pathways within the cardiomyocyte, with roles mediating mechano-sensing, mechano-electric feedback (via modulation of ion channel activity), and calcium handling. As more precise experimental techniques for applying mechanical stresses to cells are developed, the role of these forces in cardiomyocyte function can be further understood. Furthermore, specific inhibitors of different nitric oxide synthase isoforms are now available to elucidate the role of these enzymes in mediating mechano-electrical signaling. Understanding of the links between nitric oxide production and mechano-electrical signaling is incomplete, particularly whether mechanically sensitive ion channels are regulated by nitric oxide, and how this affects the cardiac action potential. This is of particular relevance to conditions such as atrial fibrillation and heart failure, in which nitric oxide production is reduced. Dysfunction of the nitric oxide/mechano-electrical signaling pathways are likely to be a feature of cardiac pathology (e.g., atrial fibrillation, cardiomyopathy, and heart failure) and a better understanding of the importance of nitric oxide signaling and its links to mechanical regulation of heart function may advance our understanding of these conditions.
Collapse
Affiliation(s)
- Hannah E. Boycott
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - My-Nhan Nguyen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Asensio Lopez MDC, Lax A, Hernandez Vicente A, Saura Guillen E, Hernandez-Martinez A, Fernandez Del Palacio MJ, Bayes-Genis A, Pascual Figal DA. Empagliflozin improves post-infarction cardiac remodeling through GTP enzyme cyclohydrolase 1 and irrespective of diabetes status. Sci Rep 2020; 10:13553. [PMID: 32782412 PMCID: PMC7419540 DOI: 10.1038/s41598-020-70454-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/30/2020] [Indexed: 12/30/2022] Open
Abstract
Sodium-glucose co-transporter-2 inhibitors (SGLT2i) have shown to prevent heart failure progression, although the mechanisms remain poorly understood. Here we evaluated the effect of empagliflozin (EMPA, SGLT2i) in cardiac remodeling after myocardial infarction, the interplay with diabetes status and the role of cardiac GTP enzyme cyclohydrolase 1 (cGCH1). A rat model of diabetes (50 mg/kg streptozotocin, i.p.) was subjected to myocardial infarction and left ventricular systolic dysfunction, by ligation of the left anterior descending coronary artery. EMPA therapy significantly improved cardiac remodeling parameters and ameliorated processes of fibrosis and hypertrophy, in both non-diabetic and diabetic rats. This cardioprotective effect related with a significant increase in myocardial expression levels of cGCH1, which led to activation of nNOS and eNOS, and inhibition of iNOS, and subsequently resulted in increasing of NO levels and decreasing O2.- and nitrotyrosine levels. These effects were replicated in a cardiomyocyte biomechanical stretching diabetic model, where silencing cGCH1 blocked the preventive effect of EMPA. The beneficial effects were observed irrespective of diabetes status, although the magnitude was greater in presence of diabetes. Empagliflozin improves myocardial remodeling after myocardial infarction through overexpression of cGCH1, and irrespective of diabetes status.
Collapse
Affiliation(s)
- Maria Del Carmen Asensio Lopez
- Biomedical Research Institute Virgen de La Arrixaca (IMIB-Arrixaca), University of Murcia, Ctra. Madrid-Cartagena S/N, 30120, Murcia, Spain
| | - Antonio Lax
- Biomedical Research Institute Virgen de La Arrixaca (IMIB-Arrixaca), University of Murcia, Ctra. Madrid-Cartagena S/N, 30120, Murcia, Spain.
| | - Alvaro Hernandez Vicente
- Cardiology Department, IMIB-Arrixaca, University of Murcia, Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Elena Saura Guillen
- Endocrinology Department, Hospital Virgen de La Arrixaca, University of Murcia, Murcia, Spain
| | | | | | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari German Trías i Puyol, CIBERCV, BadalonaMadrid, Spain
| | - Domingo A Pascual Figal
- Cardiology Department, IMIB-Arrixaca, University of Murcia, Hospital Virgen de la ArrixacaLAIB room 2.52, Avda. Buenavista s/n, 30120, Murcia, Spain. .,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain. .,CIBERCV, Madrid, Spain.
| |
Collapse
|
43
|
Molecular mechanisms by which iNOS uncoupling can induce cardiovascular dysfunction during sepsis: Role of posttranslational modifications (PTMs). Life Sci 2020; 255:117821. [PMID: 32445759 DOI: 10.1016/j.lfs.2020.117821] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/01/2023]
Abstract
Human sepsis is the result of a multifaceted pathological process causing marked dysregulation of cardiovascular responses. A more sophisticated understanding of the pathogenesis of sepsis is certainly prerequisite. Evidence from studies provide further insight into the role of inducible nitric oxide synthase (iNOS) isoform. Results on inhibition of iNOS in sepsis models remain inconclusive. Concern has been devoted to improving our knowledge and understanding of the role of iNOS. The aim of this review is to define the role of iNOS in redox homeostasis disturbance, the detailed mechanisms linking iNOS and posttranslational modifications (PTMs) to cardiovascular dysfunctions, and their future implications in sepsis settings. Many questions related to the iNOS and PTMs still remain open, and much more work is needed on this.
Collapse
|
44
|
Lo SB, Blaszak RT, Parajuli N. Targeting Mitochondria during Cold Storage to Maintain Proteasome Function and Improve Renal Outcome after Transplantation. Int J Mol Sci 2020; 21:E3506. [PMID: 32429129 PMCID: PMC7279041 DOI: 10.3390/ijms21103506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022] Open
Abstract
Kidney transplantation is the preferred treatment for end-stage kidney disease (ESKD). Compared to maintenance dialysis, kidney transplantation results in improved patient survival and quality of life. Kidneys from living donors perform best; however, many patients with ESKD depend on kidneys from deceased donors. After procurement, donor kidneys are placed in a cold-storage solution until a suitable recipient is located. Sadly, prolonged cold storage times are associated with inferior transplant outcomes; therefore, in most situations when considering donor kidneys, long cold-storage times are avoided. The identification of novel mechanisms of cold-storage-related renal damage will lead to the development of new therapeutic strategies for preserving donor kidneys; to date, these mechanisms remain poorly understood. In this review, we discuss the importance of mitochondrial and proteasome function, protein homeostasis, and renal recovery during stress from cold storage plus transplantation. Additionally, we discuss novel targets for therapeutic intervention to improve renal outcomes.
Collapse
Affiliation(s)
- Sorena B. Lo
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Richard T. Blaszak
- Division of Nephrology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Nirmala Parajuli
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
45
|
Gorska-Ponikowska M, Ploska A, Jacewicz D, Szkatula M, Barone G, Lo Bosco G, Lo Celso F, Dabrowska AM, Kuban-Jankowska A, Gorzynik-Debicka M, Knap N, Chmurzynski L, Dobrucki LW, Kalinowski L, Wozniak M. Modification of DNA structure by reactive nitrogen species as a result of 2-methoxyestradiol-induced neuronal nitric oxide synthase uncoupling in metastatic osteosarcoma cells. Redox Biol 2020; 32:101522. [PMID: 32305006 PMCID: PMC7162974 DOI: 10.1016/j.redox.2020.101522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022] Open
Abstract
2-methoxyestradiol (2-ME) is a physiological anticancer compound, metabolite of 17β-estradiol. Previously, our group evidenced that from mechanistic point of view one of anticancer mechanisms of action of 2-ME is specific induction and nuclear hijacking of neuronal nitric oxide synthase (nNOS), resulting in local generation of nitro-oxidative stress and finally, cancer cell death. The current study aims to establish the substantial mechanism of generation of reactive nitrogen species by 2-ME. We further achieved to identify the specific reactive nitrogen species involved in DNA-damaging mechanism of 2-ME. The study was performed using metastatic osteosarcoma 143B cells. We detected the release of biologically active (free) nitric oxide (•NO) with concurrent measurements of peroxynitrite (ONOO−) in real time in a single cell of 143B cell line by using •NO/ONOO− sensitive microsensors after stimulation with calcium ionophore. Detection of nitrogen dioxide (•NO2) and determination of chemical rate constants were carried out by a stopped-flow technique. The affinity of reactive nitrogen species toward the guanine base of DNA was evaluated by density functional theory calculations. Expression and localization of nuclear factor NF-kB was determined using imaging cytometry, while cell viability assay was evaluated by MTT assay. Herein, we presented that 2-ME triggers pro-apoptotic signalling cascade by increasing cellular reactive nitrogen species overproduction – a result of enzymatic uncoupling of increased nNOS protein levels. In particular, we proved that ONOO− and •NO2 directly formed from peroxynitrous acid (ONOOH) and/or by auto-oxidation of •NO, are inducers of DNA damage in anticancer mechanism of 2-ME. Specifically, the affinity of reactive nitrogen species toward the guanine base of DNA, evaluated by density functional theory calculations, decreased in the order: ONOOH > ONOO− > •NO2 > •NO. Therefore, we propose to consider the specific inducers of nNOS as an effective tool in the field of chemotherapy.
Collapse
Affiliation(s)
- Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy; Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany.
| | - Agata Ploska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland
| | - Dagmara Jacewicz
- Department of General and Inorganic Chemistry, University of Gdansk, Gdansk, Poland
| | - Michal Szkatula
- Department of Medical Chemistry, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland
| | - Giampaolo Barone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo,Palermo, Italy
| | - Giosuè Lo Bosco
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy; Department of Mathematics and Computer Science, University of Palermo, Palermo, Italy
| | - Fabrizio Lo Celso
- Department of Physics and Chemistry "Emilio Segrè", University of Palermo, Palermo, Italy
| | | | - Alicja Kuban-Jankowska
- Department of Medical Chemistry, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland
| | - Monika Gorzynik-Debicka
- Department of Medical Chemistry, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland
| | - Lech Chmurzynski
- Department of General and Inorganic Chemistry, University of Gdansk, Gdansk, Poland
| | - Lawrence Wawrzyniec Dobrucki
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland
| | - Michal Wozniak
- Department of Medical Chemistry, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland
| |
Collapse
|
46
|
Chen Z, Haus JM, Chen L, Wu SC, Urao N, Koh TJ, Minshall RD. CCL28-induced CCR10/eNOS interaction in angiogenesis and skin wound healing. FASEB J 2020; 34:5838-5850. [PMID: 32124475 DOI: 10.1096/fj.201902060r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/06/2020] [Accepted: 02/20/2020] [Indexed: 12/25/2022]
Abstract
Chemokines and their receptors play important roles in vascular homeostasis, development, and angiogenesis. Little is known regarding the molecular signaling mechanisms activated by CCL28 chemokine via its primary receptor CCR10 in endothelial cells (ECs). Here, we test the hypothesis that CCL28/CCR10 signaling plays an important role in regulating skin wound angiogenesis through endothelial nitric oxide synthase (eNOS)-dependent Src, PI3K, and MAPK signaling. We observed nitric oxide (NO) production in human primary ECs stimulated with exogenous CCL28, which also induced direct binding of CCR10 and eNOS resulting in inhibition of eNOS activity. Knockdown of CCR10 with siRNA lead to reduced eNOS expression and tube formation suggesting the involvement of CCR10 in EC angiogenesis. Based on this interaction, we engineered a myristoylated 7 amino acid CCR10-binding domain (Myr-CBD7) peptide and showed that this can block eNOS interaction with CCR10, but not with calmodulin, resulting in upregulation of eNOS activity. Importantly, topical administration of Myr-CBD7 peptide on mouse dermal wounds not only blocked CCR10-eNOS interaction, but also enhanced expression of eNOS, CD31, and IL-4 with reduction of CCL28 and IL-6 levels associated with improved wound healing. These results point to a potential therapeutic strategy to upregulate NO bioavailability, enhance angiogenesis, and improve wound healing by disrupting CCL28-activated CCR10-eNOS interaction.
Collapse
Affiliation(s)
- Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jacob M Haus
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Lin Chen
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Stephanie C Wu
- Center for Lower Extremity Ambulatory Research (CLEAR), Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
47
|
Lee I, Kim S, Nagar H, Choi SJ, Jeon BH, Piao S, Kim CS. CR6-interacting factor 1 deficiency reduces endothelial nitric oxide synthase activity by inhibiting biosynthesis of tetrahydrobiopterin. Sci Rep 2020; 10:842. [PMID: 31964986 PMCID: PMC6972730 DOI: 10.1038/s41598-020-57673-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/31/2019] [Indexed: 01/07/2023] Open
Abstract
Downregulation of CR6 interacting factor 1 (CRIF1) has been reported to induce mitochondrial dysfunction, resulting in reduced activity of endothelial nitric oxide synthase (eNOS) and NO production in endothelial cells. Tetrahydrobiopterin (BH4) is an important cofactor in regulating the balance between NO (eNOS coupling) and superoxide production (eNOS uncoupling). However, whether the decreased eNOS and NO production in CRIF1-deficient cells is associated with relative BH4 deficiency-induced eNOS uncoupling remains completely unknown. Our results showed that CRIF1 deficiency increased eNOS uncoupling and depleted levels of total biopterin and BH4 by reducing the enzymes of BH4 biosynthesis (GCH-1, PTS, SPR, and DHFR) in vivo and vitro, respectively. Supplementation of CRIF1-deficient cells with BH4 significantly increased the recovery of Akt and eNOS phosphorylation and NO synthesis. In addition, scavenging ROS with MitoTEMPO treatment replenished BH4 levels by elevating levels of GCH-1, PTS, and SPR, but with no effect on the level of DHFR. Downregulation of DHFR synthesis regulators p16 or p21 in CRIF1-deficient cells partially recovered the DHFR expression. In summary, CRIF1 deficiency inhibited BH4 biosynthesis and exacerbated eNOS uncoupling. This resulted in reduced NO production and increased oxidative stress, which contributes to endothelial dysfunction and is involved in the pathogenesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Ikjun Lee
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Seonhee Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Harsha Nagar
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Su-Jeong Choi
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Shuyu Piao
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea.
| | - Cuk-Seong Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea.
| |
Collapse
|
48
|
Nieuwenhuijs-Moeke GJ, Pischke SE, Berger SP, Sanders JSF, Pol RA, Struys MMRF, Ploeg RJ, Leuvenink HGD. Ischemia and Reperfusion Injury in Kidney Transplantation: Relevant Mechanisms in Injury and Repair. J Clin Med 2020; 9:jcm9010253. [PMID: 31963521 PMCID: PMC7019324 DOI: 10.3390/jcm9010253] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemia and reperfusion injury (IRI) is a complex pathophysiological phenomenon, inevitable in kidney transplantation and one of the most important mechanisms for non- or delayed function immediately after transplantation. Long term, it is associated with acute rejection and chronic graft dysfunction due to interstitial fibrosis and tubular atrophy. Recently, more insight has been gained in the underlying molecular pathways and signalling cascades involved, which opens the door to new therapeutic opportunities aiming to reduce IRI and improve graft survival. This review systemically discusses the specific molecular pathways involved in the pathophysiology of IRI and highlights new therapeutic strategies targeting these pathways.
Collapse
Affiliation(s)
- Gertrude J. Nieuwenhuijs-Moeke
- Department of Anesthesiology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: ; Tel.: +31-631623075
| | - Søren E. Pischke
- Clinic for Emergencies and Critical Care, Department of Anesthesiology, Department of Immunology, Oslo University Hospital, 4950 Nydalen, 0424 Oslo, Norway;
| | - Stefan P. Berger
- Department of Nephrology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (S.P.B.); (J.S.F.S.)
| | - Jan Stephan F. Sanders
- Department of Nephrology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (S.P.B.); (J.S.F.S.)
| | - Robert A. Pol
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (R.A.P.); (R.J.P.); (H.G.D.L.)
| | - Michel M. R. F. Struys
- Department of Anesthesiology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Department of Basic and Applied Medical Sciences, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Rutger J. Ploeg
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (R.A.P.); (R.J.P.); (H.G.D.L.)
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford OX3 9DU, UK
| | - Henri G. D. Leuvenink
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (R.A.P.); (R.J.P.); (H.G.D.L.)
| |
Collapse
|
49
|
Mulla W, Hajaj B, Elyagon S, Mor M, Gillis R, Murninkas M, Klapper-Goldstein H, Plaschkes I, Chalifa-Caspi V, Etzion S, Etzion Y. Rapid Atrial Pacing Promotes Atrial Fibrillation Substrate in Unanesthetized Instrumented Rats. Front Physiol 2019; 10:1218. [PMID: 31616316 PMCID: PMC6763969 DOI: 10.3389/fphys.2019.01218] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/06/2019] [Indexed: 12/19/2022] Open
Abstract
Aim The self-perpetuating nature of atrial fibrillation (AF) has been a subject of intense research in large mammalian models exposed to rapid atrial pacing (RAP). Recently, rodents are increasingly used to gain insight into the pathophysiology of AF. However, little is known regarding the effects of RAP on the atria of rats and mice. Using an implantable device for electrophysiological studies in rodents, we examined on a daily basis, the effects of continuous RAP on the developed AF substrate of unanesthetized rats and mice. Methods and Results Aggressive burst pacing did not induce AF at baseline in the large majority of rodents, but repeatedly induced AF episodes in rats exposed to RAP for more than 2 days. A microarray study of left atrial tissue from rats exposed to RAP for 2 days vs. control pacing identified 304 differentially expressed genes. Enrichment analysis and comparison with a dataset of atrial tissue from AF patients revealed indications of increased carbohydrate metabolism and changes in pathways that are thought to play critical roles in human AF, including TGF-beta and IL-6 signaling. Among 19 commonly affected genes in comparison with human AF, downregulation of FOXP1 and upregulation of the KCNK2 gene encoding the Kir2.1 potassium channel were conspicuous findings, suggesting NFAT activation. Further results included reduced expression of MIR-26 and MIR-101, which is in line with NFAT activation. Conclusion Our results demonstrate electrophysiological evidence for AF promoting effects of RAP in rats and several molecular similarities between the effects of RAP in large and small mammalian models.
Collapse
Affiliation(s)
- Wesam Mulla
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Barak Hajaj
- Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sigal Elyagon
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michal Mor
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Roni Gillis
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hadar Klapper-Goldstein
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Inbar Plaschkes
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vered Chalifa-Caspi
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sharon Etzion
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
50
|
Aziz MY, Chik Z. Bioanalysis of tetrahydrobiopterin with liquid chromatographic-mass spectrometric and its application for pharmacokinetics in apolipoprotein E knockout mice. J LIQ CHROMATOGR R T 2019. [DOI: 10.1080/10826076.2019.1625376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Mohd Yusmaidie Aziz
- Integrative Medicine Cluster, Institut Perubatan dan Pergigian Termaju, Universiti Sains Malaysia, Bertam, Penang, Malaysia
| | - Zamri Chik
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|