1
|
Hilgers RH, Das KC. Redox Regulation of K + Channel: Role of Thioredoxin. Antioxid Redox Signal 2024; 41:818-844. [PMID: 39099341 PMCID: PMC11631806 DOI: 10.1089/ars.2023.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 08/06/2024]
Abstract
Significance: Potassium channels regulate the influx and efflux of K+ ions in various cell types that generate and propagate action potential associated with excitation, contraction, and relaxation of various cell types. Although redox active cysteines are critically important for channel activity, the redox regulation of K+ channels by thioredoxin (Trx) has not been systematically reviewed. Recent Advances: Redox regulation of K+ channel is now increasingly recognized as drug targets in the pathological condition of several cardiovascular disease processes. The role of Trx in regulation of these channels and its implication in pathological conditions have not been adequately reviewed. This review specifically focuses on the redox-regulatory role of Trx on K+ channel structure and function in physiological and pathophysiological conditions. Critical Issues: Ion channels, including K+ channel, have been implicated in the functioning of cardiomyocyte excitation-contraction coupling, vascular hyperpolarization, cellular proliferation, and neuronal stimulation in physiological and pathophysiological conditions. Although oxidation-reduction of ion channels is critically important in their function, the role of Trx, redox regulatory protein in regulation of these channels, and its implication in pathological conditions need to be studied to gain further insight into channel function. Future Directions: Future studies need to map all redox regulatory pathways in channel structure and function using novel mouse models and redox proteomic and signal transduction studies, which modulate various currents and altered excitability of relevant cells implicated in a pathological condition. We are yet at infancy of studies related to redox control of various K+ channels and structured and focused studies with novel animal models. Antioxid. Redox Signal. 41, 818-844.
Collapse
Affiliation(s)
- Rob H.P. Hilgers
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kumuda C. Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
2
|
Wang YT, Moura AK, Zuo R, Zhou W, Wang Z, Roudbari K, Hu JZ, Li PL, Zhang Y, Li X. Coronary Microvascular Dysfunction is Associated with Augmented Lysosomal Signaling in Hypercholesterolemic Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.603000. [PMID: 39026774 PMCID: PMC11257577 DOI: 10.1101/2024.07.10.603000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Accumulating evidence indicates that coronary microvascular dysfunction (CMD) caused by hypercholesterolemia can lead to myocardial ischemia, with or without obstructive atherosclerotic coronary artery disease (CAD). However, the molecular pathways associated with compromised coronary microvascular function prior to the development of myocardial ischemic injury remain poorly defined. In this study, we investigated the effects of hypercholesterolemia on the function and integrity of the coronary microcirculation in mice and the underlying mechanisms. Mice were fed with a hypercholesterolemic Paigen's diet (PD) for 8 weeks. Echocardiography data showed that PD caused CMD, characterized by significant reductions in coronary blood flow and coronary flow reserve (CFR), but did not affect cardiac remodeling or dysfunction. Immunofluorescence studies revealed that PD-induced CMD was associated with activation of coronary arterioles inflammation and increased myocardial inflammatory cell infiltration. These pathological changes occurred in parallel with the upregulation of lysosomal signaling pathways in endothelial cells (ECs). Treating hypercholesterolemic mice with the cholesterol-lowering drug ezetimibe significantly ameliorated PD-induced adverse effects, including hypercholesterolemia, steatohepatitis, reduced CFR, coronary EC inflammation, and myocardial inflammatory cell infiltration. In cultured mouse cardiac endothelial cells (MCECs), 7-ketocholesterol (7K) increased mitochondrial reactive oxygen species (ROS) and inflammatory responses. Meanwhile, 7K induced the activation of TFEB and lysosomal signaling in MCECs, whereas the lysosome inhibitor bafilomycin A1 blocked 7K-induced TFEB activation and exacerbated 7K-induced inflammation and cell death. Interestingly, ezetimibe synergistically enhanced 7K-induced TFEB activation and attenuated 7K-induced mitochondrial ROS and inflammatory responses in MCECs. These results suggest that CMD can develop and precede detectable cardiac functional or structural changes in the setting of hypercholesterolemia, and that upregulation of TFEB-mediated lysosomal signaling in ECs plays a protective role against CMD.
Collapse
Affiliation(s)
- Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Wei Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| |
Collapse
|
3
|
Britto-Júnior J, Furlaneto R, Lima AT, de Oliveira MG, Severino B, Frecentese F, Fiorino F, Caliendo G, Muscará MN, De Nucci G. GKT137831 and hydrogen peroxide increase the release of 6-nitrodopamine from the human umbilical artery, rat-isolated right atrium, and rat-isolated vas deferens. Front Pharmacol 2024; 15:1348876. [PMID: 38645555 PMCID: PMC11026650 DOI: 10.3389/fphar.2024.1348876] [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: 12/03/2023] [Accepted: 02/01/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: The human umbilical artery (HUA), rat-isolated right atrium, and rat-isolated vas deferens present a basal release of 6-nitrodopamine (6-ND). The basal release of 6-ND from these tissues was significantly decreased (but not abolished) when the tissues were pre-incubated with Nω-nitro-L-arginine methyl ester (L-NAME). Methods: In this study, the effect of the pharmacological modulation of the redox environment on the basal release of 6-ND was investigated. The basal release of 6-ND was measured using Liquid chromatography with tandem mass spectrometry (LC-MS/MS). Results and Discussion: Pre-incubation (30 min) of the tissues with GKT137831 (1 μM) caused a significant increase in the basal release of 6-ND from all tissues. In the HUA, pre-incubation with diphenyleneiodonium (DPI) (100 μM) also caused significant increases in the basal release of 6-ND. Preincubation of the HUA with hydrogen peroxide (H2O2) (100 μM) increased 6-ND basal release, whereas pre-incubation with catalase (1,000 U/mL) significantly decreased it. Pre-incubation of the HUA with superoxide dismutase (SOD) (250 U/mL; 30 min) also significantly increased the basal release of 6-ND. Preincubation of the HUA with either allopurinol (100 μM) or uric acid (1 mM) had no effect on the basal release of 6-ND. Pre-treatment of the HUA with L-NAME (100 μM) prevented the increase in the basal release of 6-ND induced by GKT137831, diphenyleneiodonium, and H2O2. The results obtained indicate a major role of endogenous H2O2 and peroxidases as modulators of 6- ND biosynthesis/release and a lack of peroxynitrite contribution.
Collapse
Affiliation(s)
- José Britto-Júnior
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Rafael Furlaneto
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Department of Pharmacology, Faculty of Medicine, Sao Leopoldo Mandic, Campinas, São Paulo, Brazil
| | - Antonio Tiago Lima
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Beatrice Severino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesco Frecentese
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Ferdinando Fiorino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Giuseppe Caliendo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Marcelo Nicolás Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), Sāo Paulo, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), Sāo Paulo, Brazil
- Department of Pharmacology, Faculty of Medicine, Sao Leopoldo Mandic, Campinas, São Paulo, Brazil
| |
Collapse
|
4
|
Longden TA, Lederer WJ. Electro-metabolic signaling. J Gen Physiol 2024; 156:e202313451. [PMID: 38197953 PMCID: PMC10783436 DOI: 10.1085/jgp.202313451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Precise matching of energy substrate delivery to local metabolic needs is essential for the health and function of all tissues. Here, we outline a mechanistic framework for understanding this critical process, which we refer to as electro-metabolic signaling (EMS). All tissues exhibit changes in metabolism over varying spatiotemporal scales and have widely varying energetic needs and reserves. We propose that across tissues, common signatures of elevated metabolism or increases in energy substrate usage that exceed key local thresholds rapidly engage mechanisms that generate hyperpolarizing electrical signals in capillaries that then relax contractile elements throughout the vasculature to quickly adjust blood flow to meet changing needs. The attendant increase in energy substrate delivery serves to meet local metabolic requirements and thus avoids a mismatch in supply and demand and prevents metabolic stress. We discuss in detail key examples of EMS that our laboratories have discovered in the brain and the heart, and we outline potential further EMS mechanisms operating in tissues such as skeletal muscle, pancreas, and kidney. We suggest that the energy imbalance evoked by EMS uncoupling may be central to cellular dysfunction from which the hallmarks of aging and metabolic diseases emerge and may lead to generalized organ failure states-such as diverse flavors of heart failure and dementia. Understanding and manipulating EMS may be key to preventing or reversing these dysfunctions.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W. Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Zatz R, De Nucci G. Endothelium-Derived Dopamine and 6-Nitrodopamine in the Cardiovascular System. Physiology (Bethesda) 2024; 39:44-59. [PMID: 37874898 PMCID: PMC11283902 DOI: 10.1152/physiol.00020.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 10/26/2023] Open
Abstract
The review deals with the release of endothelium-derived dopamine and 6-nitrodopamine (6-ND) and its effects on isolated vascular tissues and isolated hearts. Basal release of both dopamine and 6-ND is present in human isolated umbilical cord vessels, human popliteal vessels, nonhuman primate vessels, and reptilia aortas. The 6-ND basal release was significantly reduced when the tissues were treated with Nω-nitro-l-arginine methyl ester and virtually abolished when the endothelium was mechanically removed. 6-Nitrodopamine is a potent vasodilator, and the mechanism of action responsible for this effect is the antagonism of dopamine D2-like receptors. As a vasodilator, 6-ND constitutes a novel mechanism by which nitric oxide modulates vascular tone. The basal release of 6-ND was substantially decreased in endothelial nitric oxide synthase knockout (eNOS-/-) mice and not altered in neuronal nitric oxide synthase knockout (nNOS-/-) mice, indicating a nonneurogenic source for 6-ND in the heart. Indeed, in rat isolated right atrium, the release of 6-ND was not affected when the atria were treated with tetrodotoxin. In the rat isolated right atrium, 6-ND is the most potent endogenous positive chronotropic agent, and in Langendorff's heart preparation, it is the most potent endogenous positive inotropic agent. The positive chronotropic and inotropic effects of 6-ND are antagonized by β1-adrenoceptor antagonists at concentrations that do not affect the effects induced by noradrenaline, adrenaline, and dopamine, indicating that blockade of the 6-ND receptor is the major modulator of heart chronotropism and inotropism. The review proposes that endothelium-derived catecholamines may constitute a major mechanism for control of vascular tone and heart functions, in contrast to the overrated role attributed to the autonomic nervous system.
Collapse
Affiliation(s)
- Roberto Zatz
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (ICB-USP), São Paulo, Brazil
- Department of Pharmacology, Faculty of Medicine, São Leopoldo Mandic, Campinas, São Paulo, Brazil
- Department of Pharmacology, Faculty of Medicine, Metropolitan University of Santos, Santos, São Paulo, Brazil
| |
Collapse
|
6
|
Awata WMC, Alves JV, Costa RM, Bruder-Nascimento A, Singh S, Barbosa GS, Tirapelli CR, Bruder-Nascimento T. Vascular injury associated with ethanol intake is driven by AT1 receptor and mitochondrial dysfunction. Biomed Pharmacother 2023; 169:115845. [PMID: 37951022 DOI: 10.1016/j.biopha.2023.115845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND Renin-angiotensin (Ang II)-aldosterone system (RAAS) is crucial for the cardiovascular risk associated with excessive ethanol consumption. Disturbs in mitochondria have been implicated in multiple cardiovascular diseases. However, if mitochondria dysfunction contributes to ethanol-induced vascular dysfunction is still unknown. We investigated whether ethanol leads to vascular dysfunction via RAAS activation, mitochondria dysfunction, and mitochondrial reactive oxygen species (mtROS). METHODS Male C57/BL6J or mt-keima mice (6-8-weeks old) were treated with ethanol (20% vol./vol.) for 12 weeks with or without Losartan (10 mg/kg/day). RESULTS Ethanol induced aortic hypercontractility in an endothelium-dependent manner. PGC1α (a marker of biogenesis), Mfn2, (an essential protein for mitochondria fusion), as well as Pink-1 and Parkin (markers of mitophagy), were reduced in aortas from ethanol-treated mice. Disturb in mitophagy flux was further confirmed in arteries from mt-keima mice. Additionally, ethanol increased mtROS and reduced SOD2 expression. Strikingly, losartan prevented vascular hypercontractility, mitochondrial dysfunction, mtROS, and restored SOD2 expression. Both MnTMPyP (SOD2 mimetic) and CCCP (a mitochondrial uncoupler) reverted ethanol-induced vascular dysfunction. Moreover, L-NAME (NOS inhibitor) and EUK 134 (superoxide dismutase/catalase mimetic) did not affect vascular response in ethanol group, suggesting that ethanol reduces aortic nitric oxide (NO) and H2O2 bioavailability. These responses were prevented by losartan. CONCLUSION AT1 receptor modulates ethanol-induced vascular hypercontractility by promoting mitochondrial dysfunction, mtROS, and reduction of NO and H2O2 bioavailability. Our findings shed a new light in our understanding of ethanol-induced vascular toxicity and open perspectives of new therapeutic approaches for patients with disorder associated with abusive ethanol drinking.
Collapse
Affiliation(s)
- Wanessa M C Awata
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Juliano V Alves
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rafael M Costa
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ariane Bruder-Nascimento
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA
| | - Shubhnita Singh
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA
| | - Gabriela S Barbosa
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; UNIPEX, Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | | | - Thiago Bruder-Nascimento
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Vascular Medicine, Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Shimokawa H. Roles of endothelial and smooth muscle cell dysfunction and vasa vasorum in vasomotor disorders in ischemia with no obstructive coronary artery disease. Vascul Pharmacol 2023; 153:107234. [PMID: 37741354 DOI: 10.1016/j.vph.2023.107234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Recently, the importance has emerged of ischemia with no obstructive coronary artery disease (INOCA), for which endothelial and vascular smooth muscle cell (VSMC) dysfunctions and alterations in coronary vasa vasorum are involved. Regarding endothelial vasodilator functions, both endothelium-derived nitric oxide and endothelium-derived hyperpolarizing factor play important roles in modulating vascular tone, especially in the microcirculation. Recent studies have suggested systemic endothelial dysfunction in INOCA. Regarding VSMC dysfunction, Rho-kinase has been identified as a key molecular mechanism of VSMC hyperconstriction in INOCA. Finally, recent advances of coronary imaging have demonstrated the important role of altered adventitial vasa vasorum functions in INOCA.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- International University of Health and Welfare, 4-3 Kozunomori, Narita 286-8686, Japan.
| |
Collapse
|
8
|
Alves-Lopes R, Lacchini S, Neves KB, Harvey A, Montezano AC, Touyz RM. Vasoprotective effects of NOX4 are mediated via polymerase and transient receptor potential melastatin 2 cation channels in endothelial cells. J Hypertens 2023; 41:1389-1400. [PMID: 37272080 PMCID: PMC10399938 DOI: 10.1097/hjh.0000000000003478] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND NOX4 activation has been implicated to have vasoprotective and blood pressure (BP)-lowering effects. Molecular mechanisms underlying this are unclear, but NOX4-induced regulation of the redox-sensitive Ca 2+ channel TRPM2 and effects on endothelial nitric oxide synthase (eNOS)-nitric oxide signalling may be important. METHOD Wild-type and LinA3, renin-expressing hypertensive mice, were crossed with NOX4 knockout mice. Vascular function was measured by myography. Generation of superoxide (O 2- ) and hydrogen peroxide (H 2 O 2 ) were assessed by lucigenin and amplex red, respectively, and Ca 2+ influx by Cal-520 fluorescence in rat aortic endothelial cells (RAEC). RESULTS BP was increased in NOX4KO, LinA3 and LinA3/NOX4KO mice. This was associated with endothelial dysfunction and vascular remodelling, with exaggerated effects in NOX4KO groups. The TRPM2 activator, ADPR, improved vascular relaxation in LinA3/NOX4KO mice, an effect recapitulated by H 2 O 2 . Inhibition of PARP and TRPM2 with olaparib and 2-APB, respectively, recapitulated endothelial dysfunction in NOX4KO. In endothelial cells, Ang II increased H 2 O 2 generation and Ca 2+ influx, effects reduced by TRPM2 siRNA, TRPM2 inhibitors (8-br-cADPR, 2-APB), olaparib and GKT137831 (NOX4 inhibitor). Ang II-induced eNOS activation was blocked by NOX4 and TRPM2 siRNA, GKT137831, PEG-catalase and 8-br-cADPR. CONCLUSION Our findings indicate that NOX4-induced H 2 O 2 production activates PARP/TRPM2, Ca 2+ influx, eNOS activation and nitric oxide release in endothelial cells. NOX4 deficiency impairs Ca 2+ homeostasis leading to endothelial dysfunction, an effect exacerbated in hypertension. We define a novel pathway linking endothelial NOX4/H 2 O 2 to eNOS/nitric oxide through PARP/TRPM2/Ca 2+ . This vasoprotective pathway is perturbed when NOX4 is downregulated and may have significance in conditions associated with endothelial dysfunction, including hypertension.
Collapse
Affiliation(s)
- Rheure Alves-Lopes
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Silvia Lacchini
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karla B. Neves
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Adam Harvey
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Augusto C. Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rhian M. Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
9
|
Wolhuter K, Kong SM, Stanley CP, Kovacic JC. The Role of Oxidants in Percutaneous Coronary Intervention-Induced Endothelial Dysfunction: Can We Harness Redox Signaling to Improve Clinical Outcomes? Antioxid Redox Signal 2023; 38:1022-1040. [PMID: 36641638 PMCID: PMC10402708 DOI: 10.1089/ars.2022.0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/03/2023] [Indexed: 01/16/2023]
Abstract
Significance: Coronary artery disease (CAD) is commonly treated using percutaneous coronary interventions (PCI). However, PCI with stent placement damages the endothelium, and failure to restore endothelial function may result in PCI failure with poor patient outcomes. Recent Advances: Oxidative signaling is central to maintaining endothelial function. Potentiation of oxidant production, as observed post-PCI, results in endothelial dysfunction (ED). This review delves into our current understanding of the physiological role that endothelial-derived oxidants play within the vasculature and the effects of altered redox signaling during dysfunction. We then examine the impact of PCI and intracoronary stent placement on oxidant production in the endothelium, which can culminate in stent failure. Finally, we explore how recent advances in PCI and stent technologies aim to mitigate PCI-induced oxidative damage and improve clinical outcomes. Critical Issues: Current PCI technologies exacerbate cellular oxidant levels, driving ED. If left uncontrolled, oxidative signaling leads to increased intravascular inflammation, restenosis, and neoatherosclerosis. Future Directions: Through the development of novel biomaterials and therapeutics, we can limit PCI-induced oxidant production, allowing for the restoration of a healthy endothelium and preventing CAD recurrence.
Collapse
Affiliation(s)
- Kathryn Wolhuter
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Stephanie M.Y. Kong
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | | | - Jason C. Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Godo S, Takahashi J, Shiroto T, Yasuda S, Shimokawa H. Coronary Microvascular Spasm: Clinical Presentation and Diagnosis. Eur Cardiol 2023; 18:e07. [PMID: 37377449 PMCID: PMC10291603 DOI: 10.15420/ecr.2022.50] [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: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 06/29/2023] Open
Abstract
Professor Maseri pioneered the research and treatment of coronary vasomotion abnormalities represented by coronary vasospasm and coronary microvascular dysfunction (CMD). These mechanisms can cause myocardial ischaemia even in the absence of obstructive coronary artery disease, and have been appreciated as an important aetiology and therapeutic target with major clinical implications in patients with ischaemia with non-obstructive coronary artery disease (INOCA). Coronary microvascular spasm is one of the key mechanisms responsible for myocardial ischaemia in patients with INOCA. Comprehensive assessment of coronary vasomotor reactivity by invasive functional coronary angiography or interventional diagnostic procedure is recommended to identify the underlying mechanisms of myocardial ischaemia and to tailor the best treatment and management based on the endotype of INOCA. This review highlights the pioneering works of Professor Maseri and contemporary research on coronary vasospasm and CMD with reference to endothelial dysfunction, Rho-kinase activation and inflammation.
Collapse
Affiliation(s)
- Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of MedicineSendai, Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of MedicineSendai, Japan
| | - Takashi Shiroto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of MedicineSendai, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of MedicineSendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of MedicineSendai, Japan
- Graduate School, International University of Health and WelfareNarita, Japan
| |
Collapse
|
11
|
Zhu H, Wang H, Zhu X, Chen Q, Fang X, Xu X, Ping Y, Gao B, Tong G, Ding Y, Chen T, Huang J. The Importance of Integrated Regulation Mechanism of Coronary Microvascular Function for Maintaining the Stability of Coronary Microcirculation: An Easily Overlooked Perspective. Adv Ther 2023; 40:76-101. [PMID: 36279093 DOI: 10.1007/s12325-022-02343-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/28/2022] [Indexed: 01/25/2023]
Abstract
Coronary microvascular dysfunction (CMD) refers to a group of disorders affecting the structure and function of coronary microcirculation and is associated with an increased risk of major adverse cardiovascular events. At present, great progress has been made in the diagnosis of CMD, but there is no specific treatment for it because of the complexity of CMD pathogenesis. Vascular dysfunction is one of the important causes of CMD, but previous reviews mostly considered microvascular dysfunction as a whole abnormality so the obtained conclusions are skewed. The coronary microvascular function is co-regulated by multiple mechanisms, and the mechanisms by which microvessels of different luminal diameters are regulated vary. The main purpose of this review is to revisit the mechanisms by which coronary microvessels at different diameters regulate coronary microcirculation through integrated sequential activation and briefly discuss the pathogenesis, diagnosis, and treatment progress of CMD from this perspective.
Collapse
Affiliation(s)
- Houyong Zhu
- Department of Cardiology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Stadium Road, Hangzhou, 310007, Zhejiang, China.
| | - Hanxin Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xinyu Zhu
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Qilan Chen
- Department of Cardiology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Xiaojiang Fang
- Department of Cardiology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Xiaoqun Xu
- Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yan Ping
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Beibei Gao
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Guoxin Tong
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Yu Ding
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Tielong Chen
- Department of Cardiology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Stadium Road, Hangzhou, 310007, Zhejiang, China.
| | - Jinyu Huang
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
12
|
Rouver WDN, Delgado NTB, Gonçalves LT, Giesen JAS, Santos da Costa C, Merlo E, Damasceno Costa E, Lemos VS, Bernardes Graceli J, Santos RLD. Sex hormones and vascular reactivity: a temporal evaluation in resistance arteries of male rats. J Mol Endocrinol 2023; 70:e220147. [PMID: 36476761 DOI: 10.1530/jme-22-0147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
The role of androgens in vascular reactivity is controversial, particularly regarding their age-related actions. The objective of this study was to conduct a temporal evaluation of the vascular reactivity of resistance arteries of young male rats, as well as to understand how male sex hormones can influence the vascular function of these animals. Endothelium-mediated relaxation was characterized in third-order mesenteric arteries of 10-, 12-, 16-, and 18w (week-old) male rats. Concentration-response curves to acetylcholine (ACh, 0.1 nmol/L-10 µmol/L) were constructed in arteries previously contracted with phenylephrine (PE, 3 µmol/L), before and after the use of nitric oxide synthase or cyclooxygenase inhibitors. PE concentration-response curves (1 nmol/L-100 μmol/L) were also built. The levels of vascular nitric oxide, superoxide anion, and hydrogen peroxide were assessed and histomorphometry analysis was performed. The 18w group had impaired endothelium-dependent relaxation. All groups showed prostanoid-independent and nitric oxide-dependent vasodilatory response, although this dependence seems to be smaller in the 18w group. The 18w group had the lowest nitric oxide and hydrogen peroxide production, in addition to the highest superoxide anion levels. Besides functional impairment, 18w animals showed morphological differences in third-order mesenteric arteries compared with the other groups. Our data show that time-dependent exposure to male sex hormones appears to play an important role in the development of vascular changes that can lead to impaired vascular reactivity in mesenteric arteries, which could be related to the onset of age-related cardiovascular changes in males.
Collapse
Affiliation(s)
- Wender do Nascimento Rouver
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Leticia Tinoco Gonçalves
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Charles Santos da Costa
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Merlo
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Damasceno Costa
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Virginia Soares Lemos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jones Bernardes Graceli
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| |
Collapse
|
13
|
Hu XQ, Zhang L. Oxidative Regulation of Vascular Ca v1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11122432. [PMID: 36552639 PMCID: PMC9774363 DOI: 10.3390/antiox11122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca2+ (Cav1.2) channel in small arteries and arterioles plays an essential role in regulating Ca2+ influx, vascular resistance, and blood pressure. Hypertension and preeclampsia are characterized by high blood pressure. In addition, diabetes has a high prevalence of hypertension. The etiology of these disorders remains elusive, involving the complex interplay of environmental and genetic factors. Common to these disorders are oxidative stress and vascular dysfunction. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria are primary sources of vascular oxidative stress, whereas dysfunction of the Cav1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav1.2 and potential roles of ROS-mediated Cav1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia.
Collapse
|
14
|
Avtaar Singh SS, Nappi F. Pathophysiology and Outcomes of Endothelium Function in Coronary Microvascular Diseases: A Systematic Review of Randomized Controlled Trials and Multicenter Study. Biomedicines 2022; 10:biomedicines10123010. [PMID: 36551766 PMCID: PMC9775403 DOI: 10.3390/biomedicines10123010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Coronary macrovascular disease is a concept that has been well-studied within the literature and has long been the subject of debates surrounding coronary artery bypass grafting (CABG) vs. Percutaneous Coronary Intervention (PCI). ISCHEMIA trial reported no statistical difference in the primary clinical endpoint between initial invasive management and initial conservative management, while in the ORBITA trial PCI did not improve angina frequency score significantly more than placebo, albeit PCI resulted in more patient-reported freedom from angina than placebo. However, these results did not prove the superiority of the PCI against OMT, therefore do not indicate the benefit of PCI vs. the OMT. Please rephrase the sentence. We reviewed the role of different factors responsible for endothelial dysfunction from recent randomized clinical trials (RCTs) and multicentre studies. METHODS A detailed search strategy was performed using a dataset that has previously been published. Data of pooled analysis include research articles (human and animal models), CABG, and PCI randomized controlled trials (RCTs). Details of the search strategy and the methods used for data pooling have been published previously and registered with Open-Source Framework. RESULTS The roles of nitric oxide (NO), endothelium-derived contracting factors (EDCFs), and vasodilator prostaglandins (e.g., prostacyclin), as well as endothelium-dependent hyperpolarization (EDH) factors, are crucial for the maintenance of vasomotor tone within the coronary vasculature. These homeostatic mechanisms are affected by sheer forces and other several factors that are currently being studied, such as vaping. The role of intracoronary testing is crucial when determining the effects of therapeutic medications with further studies on the horizon. CONCLUSION The true impact of coronary microvascular dysfunction (CMD) is perhaps underappreciated, which supports the role of medical therapy in determining outcomes. Ongoing trials are underway to further investigate the role of therapeutic agents in secondary prevention.
Collapse
Affiliation(s)
| | - Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord of Saint-Denis, 93200 Saint-Denis, France
- Correspondence: ; Tel.: +33-(14)-9334104; Fax: +33-149334119
| |
Collapse
|
15
|
da Silva FC, de Araújo BJ, Cordeiro CS, Arruda VM, Faria BQ, Guerra JFDC, Araújo TGD, Fürstenau CR. Endothelial dysfunction due to the inhibition of the synthesis of nitric oxide: Proposal and characterization of an in vitro cellular model. Front Physiol 2022; 13:978378. [PMID: 36467706 PMCID: PMC9714775 DOI: 10.3389/fphys.2022.978378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/02/2022] [Indexed: 08/13/2023] Open
Abstract
The vascular endothelium plays a pivotal role in the maintenance of vascular homeostasis, mediated by vasoactive molecules produced by endothelial cells. The balance between vasoconstrictor and vasodilator biomolecules is what guarantees this equilibrium. Therefore, an increase in the bioavailability of vasoconstrictors along with a reduction in vasodilators may indicate a condition known as endothelial dysfunction. Endothelial dysfunction is marked by an inflammatory process and reduced activity of vasoprotective enzymes, being characterized by some factors like the reduction of the bioavailability of nitric oxide (NO) and increase in the production of reactive oxygen species (ROS), pro-inflammatory and vasoconstrictor molecules. This condition is a predictive marker of several cardiovascular diseases (e.g., atherosclerosis, hypertension, and diabetes). Research is affected by the scarcity of suitable in vitro models that simulate endothelial dysfunction. The goal of this study was to induce an in vitro condition to mimic endothelial dysfunction by inhibiting NO synthesis in cells. Thymus-derived endothelial cells (tEnd.1) were treated with different concentrations of L-NAME (from 1 to 1,000 μM) for different times (12, 24, 48, 72, 96, and 120 h without and with retreatment every 24 h). Cell viability, nitrite concentration, p22phox, NOX2, NOX4, IL-6, and ACE genes expression and lipid peroxidation were evaluated. The results indicate that the treatment with 100 μM L-NAME for 72 h without retreatment reduced NO concentration and NOX4 gene expression while increasing ACE expression, thus mimicking reduced vascular protection and possibly increased vasoconstriction. On the other hand, treatment with 100 μM L-NAME for 96 h with retreatment reduced the concentration of NO and the expression of the p22phox gene while increasing the expression of the IL-6 and ACE genes, mimicking the increase in inflammation and vasoconstriction parameters. Based on these results, we thus propose that both 100 μM L-NAME for 72 h without retreatment and 100 μM L-NAME for 96 h with retreatment may be used as models for in vitro endothelial dysfunction according to the purpose of the study to be conducted.
Collapse
Affiliation(s)
- Fernanda Cardoso da Silva
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Bruna Juber de Araújo
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Carina Santos Cordeiro
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Vinícius Marques Arruda
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Bruno Quintanilha Faria
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Joyce Ferreira Da Costa Guerra
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Thaise Gonçalves De Araújo
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Cristina Ribas Fürstenau
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| |
Collapse
|
16
|
Kavurma MM, Bursill C, Stanley CP, Passam F, Cartland SP, Patel S, Loa J, Figtree GA, Golledge J, Aitken S, Robinson DA. Endothelial cell dysfunction: Implications for the pathogenesis of peripheral artery disease. Front Cardiovasc Med 2022; 9:1054576. [PMID: 36465438 PMCID: PMC9709122 DOI: 10.3389/fcvm.2022.1054576] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/24/2022] [Indexed: 08/27/2023] Open
Abstract
Peripheral artery disease (PAD) is caused by occluded or narrowed arteries that reduce blood flow to the lower limbs. The treatment focuses on lifestyle changes, management of modifiable risk factors and vascular surgery. In this review we focus on how Endothelial Cell (EC) dysfunction contributes to PAD pathophysiology and describe the largely untapped potential of correcting endothelial dysfunction. Moreover, we describe current treatments and clinical trials which improve EC dysfunction and offer insights into where future research efforts could be made. Endothelial dysfunction could represent a target for PAD therapy.
Collapse
Affiliation(s)
- Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Christina Bursill
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
| | | | - Freda Passam
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
- Central Clinical School, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jacky Loa
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
- The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD, Australia
| | - Sarah Aitken
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Concord Institute of Academic Surgery, Concord Hospital, Sydney, NSW, Australia
| | | |
Collapse
|
17
|
Peixoto P, Vieira-Alves I, Couto GK, Lemos VS, Rossoni LV, Bissoli NS, Dos Santos RL. Sex differences in the participation of endothelial mediators and signaling pathways involved in the vasodilator effect of a selective GPER agonist in resistance arteries of gonadectomized Wistar rats. Life Sci 2022; 308:120917. [PMID: 36044974 DOI: 10.1016/j.lfs.2022.120917] [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: 07/10/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022]
Abstract
AIM Endothelial mechanisms underlying the vascular effects of estrogen modulated by the G protein-coupled estrogen receptor (GPER) are not well understood, especially in gonadal sex hormone deprivation. Thus, we investigated vascular function and endothelial signaling pathways involved in the selective activation of GPER in resistance arteries of gonadectomized rats. METHODS Gonadectomy was performed in Wistar rats of both sexes. After 21 days, the animals were euthanized. Concentration-response curves were obtained by cumulative additions of G-1 in third-order mesenteric arteries. The vasodilatory effects of G-1 were evaluated before and after endothelium removal or incubation with pharmacological inhibitors. Tissue protein expression was measured by western blotting. Assays with 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM) and 2',7' dichlorodihydrofluorescein-diacetate (DCF-DA) were performed in the arteries investigated. Immunolocalization was assessed by immunofluorescence. RESULTS G-1 induced partially endothelium-dependent relaxation in both sexes. The three isoforms of the enzyme nitric oxide synthase contributed to the production and release of nitric oxide in both gonadectomized groups, but the role of inducible nitric oxide synthase is more expressive in males. The mechanistic pathway by which endothelial nitric oxide synthase is phosphorylated appears to differ between sexes, with the rapid signaling pathway phosphatidylinositol-3-kinase/protein kinase B/endothelial nitric oxide synthase (PI3k-Akt-eNOS) being identified for males and mitogen-activated protein kinase/extracellular signal-regulated kinase/endothelial nitric oxide synthase (MEK-ERK-eNOS) for females. The contribution of hydrogen peroxide as an endothelial relaxation mediator seems to be greater in females. CONCLUSION These results provide new insights into the effects of estrogen-induced responses via GPER on vascular function in gonadal sex hormone deprivation.
Collapse
Affiliation(s)
- Pollyana Peixoto
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil
| | - Ildernandes Vieira-Alves
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gisele Kruger Couto
- Department of Physiology and Biophysics, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Virgínia Soares Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Nazaré Souza Bissoli
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil.
| |
Collapse
|
18
|
Shimauchi T, Numaga-Tomita T, Kato Y, Morimoto H, Sakata K, Matsukane R, Nishimura A, Nishiyama K, Shibuta A, Horiuchi Y, Kurose H, Kim SG, Urano Y, Ohshima T, Nishida M. A TRPC3/6 Channel Inhibitor Promotes Arteriogenesis after Hind-Limb Ischemia. Cells 2022; 11:cells11132041. [PMID: 35805125 PMCID: PMC9266111 DOI: 10.3390/cells11132041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/04/2022] Open
Abstract
Retarded revascularization after progressive occlusion of large conductance arteries is a major cause of bad prognosis for peripheral artery disease (PAD). However, pharmacological treatment for PAD is still limited. We previously reported that suppression of transient receptor potential canonical (TRPC) 6 channel activity in vascular smooth muscle cells (VSMCs) facilitates VSMC differentiation without affecting proliferation and migration. In this study, we found that 1-benzilpiperadine derivative (1-BP), a selective inhibitor for TRPC3 and TRPC6 channel activities, induced VSMC differentiation. 1-BP-treated mice showed increased capillary arterialization and improvement of peripheral circulation and skeletal muscle mass after hind-limb ischemia (HLI) in mice. 1-BP had no additive effect on the facilitation of blood flow recovery after HLI in TRPC6-deficient mice, suggesting that suppression of TRPC6 underlies facilitation of the blood flow recovery by 1-BP. 1-BP also improved vascular nitric oxide bioavailability and blood flow recovery after HLI in hypercholesterolemic mice with endothelial dysfunction, suggesting the retrograde interaction from VSMCs to endothelium. These results suggest that 1-BP becomes a potential seed for PAD treatments that target vascular TRPC6 channels.
Collapse
Affiliation(s)
- Tsukasa Shimauchi
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Takuro Numaga-Tomita
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Molecular Pharmacology, Shinshu University School of Medicine and Health Sciences, Matsumoto 390-8621, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Hiroyuki Morimoto
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Kosuke Sakata
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Ryosuke Matsukane
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
| | - Kazuhiro Nishiyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Atsushi Shibuta
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Yutoku Horiuchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Hitoshi Kurose
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Sang Geon Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang-si 10326, Gyeonggi-Do, Korea;
| | - Yasuteru Urano
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan;
| | - Takashi Ohshima
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Motohiro Nishida
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
- Correspondence: ; Tel./Fax: +81-92-642-6556
| |
Collapse
|
19
|
Szczepaniak P, Siedlinski M, Hodorowicz-Zaniewska D, Nosalski R, Mikolajczyk TP, Dobosz AM, Dikalova A, Dikalov S, Streb J, Gara K, Basta P, Krolczyk J, Sulicka-Grodzicka J, Jozefczuk E, Dziewulska A, Saju B, Laksa I, Chen W, Dormer J, Tomaszewski M, Maffia P, Czesnikiewicz-Guzik M, Crea F, Dobrzyn A, Moslehi J, Grodzicki T, Harrison DG, Guzik TJ. Breast cancer chemotherapy induces vascular dysfunction and hypertension through NOX4 dependent mechanism. J Clin Invest 2022; 132:149117. [PMID: 35617030 PMCID: PMC9246378 DOI: 10.1172/jci149117] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is the major cause of morbidity and mortality in breast cancer survivors. Chemotherapy contributes to this risk. We aimed to define the mechanisms of long-term vascular dysfunction caused by neoadjuvant chemotherapy (NACT) and identify novel therapeutic targets.We studied arteries from postmenopausal women who had undergone breast cancer treatment using docetaxel, doxorubicin and cyclophosphamide (NACT), and women with no history of such treatment matched for key clinical parameters. Mechanisms were explored in wild-type and Nox4-/- mice and human microvascular endothelial cells.Endothelium-dependent vasodilatation is severely impaired in patients after NACT, while endothelium-independent responses remain normal. This was mimicked by 24-hour exposure of arteries to NACT agents ex-vivo. When applied individually, only docetaxel impaired endothelial function in human vessels. Mechanistic studies showed that NACT increased inhibitory eNOS phosphorylation of threonine 495 in a ROCK-dependent manner and augmented vascular superoxide and hydrogen peroxide production and NADPH oxidase activity. Docetaxel increased expression of NADPH oxidase NOX4 in endothelial and smooth muscle cells and NOX2 in the endothelium. NOX4 increase in human arteries may be mediated epigenetically by diminished DNA methylation of the NOX4 promoter. Docetaxel induced endothelial dysfunction and hypertension in mice. These were prevented in Nox4-/- and by pharmacological inhibition of Nox4 or Rock.Commonly used chemotherapeutic agents, and in particular, docetaxel, alter vascular function by promoting inhibitory phosphorylation of eNOS and enhancing ROS production by NADPH oxidases.
Collapse
Affiliation(s)
- Piotr Szczepaniak
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Mateusz Siedlinski
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | | | - Ryszard Nosalski
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Tomasz P Mikolajczyk
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Aneta M Dobosz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anna Dikalova
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - Joanna Streb
- Department of Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Katarzyna Gara
- Department of Surgery, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Pawel Basta
- Department of Gynecology and Gynecological Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Jaroslaw Krolczyk
- Department of Internal Medicine and Gerontology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | | | - Ewelina Jozefczuk
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Anna Dziewulska
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Blessy Saju
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Iwona Laksa
- Department of Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Wei Chen
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - John Dormer
- Department of Cellular Pathology, University Hospitals of Leicester, Leicester, United Kingdom
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Marta Czesnikiewicz-Guzik
- Department of Periodontology and Oral Sciences Research Group, University of Glasgow, Glasgow, United Kingdom
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, University of the Sacred Heart, Rome, Italy
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Javid Moslehi
- University of California San Fransisco, San Francisco, United States of America
| | - Tomasz Grodzicki
- Department of Internal Medicine and Gerontology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
20
|
Comerma-Steffensen S, Prat-Duran J, Mogensen S, Fais R, Pinilla E, Simonsen U. Erectile Dysfunction and Altered Contribution of KCa1.1 and KCa2.3 Channels in the Penile Tissue of Type-2 Diabetic db/db Mice. J Sex Med 2022; 19:697-710. [PMID: 37057569 DOI: 10.1016/j.jsxm.2022.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/15/2022] [Accepted: 02/19/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Activation of endothelial small conductance calcium-activated K+ channels (KCa2.3) and intermediate conductance calcium-activated K+ channels (KCa3.1) leads to vascular relaxation. We found endothelial KCa2.3 down-regulation in the corpus cavernosum diminishes erectile function. AIM We hypothesized that in type-2 diabetic mice, the function of KCa2.3 and KCa1.1 channels is impaired in erectile tissue. METHODS Erectile function was measured, and corpus cavernosum strips were mounted for functional studies and processed for qPCR and immunoblotting. OUTCOMES Effects of type 2 diabetes on erectile function, expression and function of calcium-activated potassium channels. RESULTS In anesthetized diabetic db/db mice, erectile function was markedly decreased compared to non-diabetic heterozygous db/+ mice, and the impairment was even more pronounced compared to normal C57BL/6 mice. qPCR revealed KCa2.3 and KCa1.1α channel expressions were upregulated in corpus cavernosum from db/db mice. Immunoblotting showed down-regulation of KCa2.3 channels in the corpus cavernosum from db/db mice. Acetylcholine relaxations were impaired while relaxations induced by the nitric oxide, donor SNP were unaltered in corpus cavernosum from db/db compared to C57BL/6 and db/+ mice. Apamin, a blocker of KCa2 channels, inhibited acetylcholine relaxation in corpus cavernosum from all experimental groups. In the presence of apamin, acetylcholine relaxation was markedly decreased in corpus cavernosum from db/db vs C57BL/6 and db/+ mice. An opener of KCa2 and KCa3.1 channels, NS309, potentiated acetylcholine relaxations in corpus cavernosum from db/+ and db/db mice. Iberiotoxin, a blocker of KCa1.1 channels, inhibited acetylcholine relaxation in corpus cavernosum from db/+ mice, while there was no effect in tissue from db/db mice. CLINICAL TRANSLATION Erectile function in diabetic db/db mice was severely affected compared to heterozygous and control mice, findings suggesting the non-diabetic db/+ and diabetic db/db mice for translational purpose can be used for drug testing on, respectively, moderate and severe erectile dysfunction. The altered expressions and impaired acetylcholine relaxation in the presence of apamin compared to C57BL/6 mice may suggest decreased KCa1.1 channel function may underpin impaired endothelium-dependent relaxation and erectile dysfunction in diabetic db/db mice. STRENGTHS & LIMITATIONS The present study provides a mouse model for type 2 diabetes to test moderate and severe erectile dysfunction drugs. Decreased KCa1.1 channel function contributes to erectile dysfunction, and it is a limitation that it is not supported by electrophysiological measurements. CONCLUSION Our results suggest that the contribution of iberiotoxin-sensitive KCa1.1 channels to relaxation is reduced in the corpus cavernosum, while apamin-sensitive KCa2.3 channels appear upregulated. The impaired KCa1.1 channel function may contribute to the impaired erectile function in diabetic db/db mice.
Collapse
Affiliation(s)
- Simon Comerma-Steffensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Biomedical Sciences/Animal Physiology, Veterinary Faculty, Central University of Venezuela, Maracay, Aragua, Venezuela
| | | | - Susie Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Rafael Fais
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Pharmacology Department, Ribeirao Preto Medical School, Sao Paulo University, Brasil
| | | | - Ulf Simonsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Kendrick DJ, Mishra RC, John CM, Zhu HL, Braun AP. Effects of Pharmacological Inhibitors of NADPH Oxidase on Myogenic Contractility and Evoked Vasoactive Responses in Rat Resistance Arteries. Front Physiol 2022; 12:752366. [PMID: 35140625 PMCID: PMC8818784 DOI: 10.3389/fphys.2021.752366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Reactive oxygen species (ROS), such as superoxide anions and hydrogen peroxide, are reported to contribute to the dynamic regulation of contractility in various arterial preparations, however, the situation in pressurized, myogenically active resistance arteries is much less clear. In the present study, we have utilized established pharmacological inhibitors of NADPH oxidase activity to examine the potential contribution of ROS to intrinsic myogenic contractility in adult Sprague–Dawley rat resistance arteries and responses to vasoactive agents acting via the endothelium (i.e., acetylcholine, SKA-31) or smooth muscle (i.e., sodium nitroprusside, phenylephrine). In cannulated and pressurized cremaster skeletal muscle and middle cerebral arteries, the NOX inhibitors 2-acetylphenothiazine (2-APT) and VAS2870, selective for NOX1 and NOX2, respectively, evoked concentration-dependent inhibition of basal myogenic tone in a reversible and irreversible manner, respectively, whereas the non-selective inhibitor apocynin augmented myogenic contractility. The vasodilatory actions of 2-APT and VAS2870 occurred primarily via the vascular endothelium and smooth muscle, respectively. Functional responses to established endothelium-dependent and –independent vasoactive agents were largely unaltered in the presence of either 2-APT or apocynin. In cremaster arteries from Type 2 Diabetic (T2D) Goto-Kakizaki rats with endothelial dysfunction, treatment with either 2-APT or apocynin did not modify stimulus-evoked vasoactive responses, but did affect basal myogenic tone. These same NOX inhibitors produced robust inhibition of total NADPH oxidase activity in aortic tissue homogenates from control and T2D rats, and NOX isozymes 1, 2 and 4, along with superoxide dismutase 1, were detected by qPCR in cremaster arteries and aorta from both species. Based on the diverse effects that we observed for established, chemically distinct NOX inhibitors, the functional contribution of vascular NADPH oxidase activity to stimulus-evoked vasoactive signaling in myogenically active, small resistance arteries remains unclear.
Collapse
|
22
|
Mechanism of the switch from NO to H 2O 2 in endothelium-dependent vasodilation in diabetes. Basic Res Cardiol 2022; 117:2. [PMID: 35024970 PMCID: PMC8886611 DOI: 10.1007/s00395-022-00910-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023]
Abstract
Coronary microvascular dysfunction is prevalent among people with diabetes and is correlated with cardiac mortality. Compromised endothelial-dependent dilation (EDD) is an early event in the progression of diabetes, but its mechanisms remain incompletely understood. Nitric oxide (NO) is the major endothelium-dependent vasodilatory metabolite in the healthy coronary circulation, but this switches to hydrogen peroxide (H2O2) in coronary artery disease (CAD) patients. Because diabetes is a significant risk factor for CAD, we hypothesized that a similar NO-to-H2O2 switch would occur in diabetes. Vasodilation was measured ex vivo in isolated coronary arteries from wild type (WT) and microRNA-21 (miR-21) null mice on a chow or high-fat/high-sugar diet, and B6.BKS(D)-Leprdb/J (db/db) mice using myography. Myocardial blood flow (MBF), blood pressure, and heart rate were measured in vivo using contrast echocardiography and a solid-state pressure sensor catheter. RNA from coronary arteries, endothelial cells, and cardiac tissues was analyzed via quantitative real-time PCR for gene expression, and cardiac protein expression was assessed via western blot analyses. Superoxide was detected via electron paramagnetic resonance. (1) Ex vivo coronary EDD and in vivo MBF were impaired in diabetic mice. (2) Nω-Nitro-L-arginine methyl ester, an NO synthase inhibitor (L-NAME), inhibited ex vivo coronary EDD and in vivo MBF in WT. In contrast, polyethylene glycol-catalase, an H2O2 scavenger (Peg-Cat), inhibited diabetic mouse EDD ex vivo and MBF in vivo. (3) miR-21 was upregulated in diabetic mouse endothelial cells, and the deficiency of miR-21 prevented the NO-to-H2O2 switch and ameliorated diabetic mouse vasodilation impairments. (4) Diabetic mice displayed increased serum NO and H2O2, upregulated mRNA expression of Sod1, Sod2, iNos, and Cav1, and downregulated Pgc-1α in coronary arteries, but the deficiency of miR-21 reversed these changes. (5) miR-21-deficient mice exhibited increased cardiac PGC-1α, PPARα and eNOS protein and reduced endothelial superoxide. (6) Inhibition of PGC-1α changed the mRNA expression of genes regulated by miR-21, and overexpression of PGC-1α decreased the expression of miR-21 in high (25.5 mM) glucose treated coronary endothelial cells. Diabetic mice exhibit a NO-to-H2O2 switch in the mediator of coronary EDD, which contributes to microvascular dysfunction and is mediated by miR-21. This study represents the first mouse model recapitulating the NO-to-H2O2 switch seen in CAD patients in diabetes.
Collapse
|
23
|
Luk C, Haywood NJ, Bridge KI, Kearney MT. Paracrine Role of the Endothelium in Metabolic Homeostasis in Health and Nutrient Excess. Front Cardiovasc Med 2022; 9:882923. [PMID: 35557517 PMCID: PMC9086712 DOI: 10.3389/fcvm.2022.882923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 02/02/2023] Open
Abstract
The vascular endothelium traditionally viewed as a simple physical barrier between the circulation and tissue is now well-established as a key organ mediating whole organism homeostasis by release of a portfolio of anti-inflammatory and pro-inflammatory vasoactive molecules. Healthy endothelium releases anti-inflammatory signaling molecules such as nitric oxide and prostacyclin; in contrast, diseased endothelium secretes pro-inflammatory signals such as reactive oxygen species, endothelin-1 and tumor necrosis factor-alpha (TNFα). Endothelial dysfunction, which has now been identified as a hallmark of different components of the cardiometabolic syndrome including obesity, type 2 diabetes and hypertension, initiates and drives the progression of tissue damage in these disorders. Recently it has become apparent that, in addition to vasoactive molecules, the vascular endothelium has the potential to secrete a diverse range of small molecules and proteins mediating metabolic processes in adipose tissue (AT), liver, skeletal muscle and the pancreas. AT plays a pivotal role in orchestrating whole-body energy homeostasis and AT dysfunction, characterized by local and systemic inflammation, is central to the metabolic complications of obesity. Thus, understanding and targeting the crosstalk between the endothelium and AT may generate novel therapeutic opportunities for the cardiometabolic syndrome. Here, we provide an overview of the role of the endothelial secretome in controlling the function of AT. The endothelial-derived metabolic regulatory factors are grouped and discussed based on their physical properties and their downstream signaling effects. In addition, we focus on the therapeutic potential of these regulatory factors in treating cardiometabolic syndrome, and discuss areas of future study of potential translatable and clinical significance. The vascular endothelium is emerging as an important paracrine/endocrine organ that secretes regulatory factors in response to nutritional and environmental cues. Endothelial dysfunction may result in imbalanced secretion of these regulatory factors and contribute to the progression of AT and whole body metabolic dysfunction. As the vascular endothelium is the first responder to local nutritional changes and adipocyte-derived signals, future work elucidating the changes in the endothelial secretome is crucial to improve our understanding of the pathophysiology of cardiometabolic disease, and in aiding our development of new therapeutic strategies to treat and prevent cardiometabolic syndrome.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
24
|
Godo S, Takahashi J, Yasuda S, Shimokawa H. Endothelium in Coronary Macrovascular and Microvascular Diseases. J Cardiovasc Pharmacol 2021; 78:S19-S29. [PMID: 34840261 PMCID: PMC8647695 DOI: 10.1097/fjc.0000000000001089] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/05/2021] [Indexed: 01/09/2023]
Abstract
ABSTRACT The endothelium plays a pivotal role in the regulation of vascular tone by synthesizing and liberating endothelium-derived relaxing factors inclusive of vasodilator prostaglandins (eg, prostacyclin), nitric oxide (NO), and endothelium-dependent hyperpolarization factors in a distinct blood vessel size-dependent manner. Large conduit arteries are predominantly regulated by NO and small resistance arteries by endothelium-dependent hyperpolarization factors. Accumulating evidence over the past few decades has demonstrated that endothelial dysfunction and coronary vasomotion abnormalities play crucial roles in the pathogenesis of various cardiovascular diseases. Structural and functional alterations of the coronary microvasculature have been coined as coronary microvascular dysfunction (CMD), which is highly prevalent and associated with adverse clinical outcomes in many clinical settings. The major mechanisms of coronary vasomotion abnormalities include enhanced coronary vasoconstrictive reactivity at epicardial and microvascular levels, impaired endothelium-dependent and endothelium-independent coronary vasodilator capacities, and elevated coronary microvascular resistance caused by structural factors. Recent experimental and clinical research has highlighted CMD as the systemic small artery disease beyond the heart, emerging modulators of vascular functions, novel insights into the pathogenesis of cardiovascular diseases associated with CMD, and potential therapeutic interventions to CMD with major clinical implications. In this article, we will summarize the current knowledge on the endothelial modulation of vascular tone and the pathogenesis of coronary macrovascular and microvascular diseases from bench to bedside, with a special emphasis placed on the mechanisms and clinical implications of CMD.
Collapse
Affiliation(s)
- Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; and
| | - Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; and
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; and
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; and
- Graduate School, International University of Health and Welfare, Narita, Japan
| |
Collapse
|
25
|
Queiroz RF, Stanley CP, Wolhuter K, Kong SMY, Rajivan R, McKinnon N, Nguyen GTH, Roveri A, Guttzeit S, Eaton P, Donald WA, Ursini F, Winterbourn CC, Ayer A, Stocker R. Hydrogen peroxide signaling via its transformation to a stereospecific alkyl hydroperoxide that escapes reductive inactivation. Nat Commun 2021; 12:6626. [PMID: 34785665 PMCID: PMC8595612 DOI: 10.1038/s41467-021-26991-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
During systemic inflammation, indoleamine 2,3-dioxygenase 1 (IDO1) becomes expressed in endothelial cells where it uses hydrogen peroxide (H2O2) to oxidize L-tryptophan to the tricyclic hydroperoxide, cis-WOOH, that then relaxes arteries via oxidation of protein kinase G 1α. Here we show that arterial glutathione peroxidases and peroxiredoxins that rapidly eliminate H2O2, have little impact on relaxation of IDO1-expressing arteries, and that purified IDO1 forms cis-WOOH in the presence of peroxiredoxin 2. cis-WOOH oxidizes protein thiols in a selective and stereospecific manner. Compared with its epimer trans-WOOH and H2O2, cis-WOOH reacts slower with the major arterial forms of glutathione peroxidases and peroxiredoxins while it reacts more readily with its target, protein kinase G 1α. Our results indicate a paradigm of redox signaling by H2O2 via its enzymatic conversion to an amino acid-derived hydroperoxide that 'escapes' effective reductive inactivation to engage in selective oxidative activation of key target proteins.
Collapse
Affiliation(s)
- Raphael F Queiroz
- Department of Natural Sciences, Southwest Bahia State University, Vitoria da Conquista, Bahia, Brazil
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Christopher P Stanley
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | - Kathryn Wolhuter
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | | | - Ragul Rajivan
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Naomi McKinnon
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Giang T H Nguyen
- School of Chemistry, University of New South Wales, Sydney, New South Wales, Australia
| | - Antonella Roveri
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Philip Eaton
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - William A Donald
- School of Chemistry, University of New South Wales, Sydney, New South Wales, Australia
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Christine C Winterbourn
- Centre for Free Radical Research, Department of Pathology, University of Otago Christchurch, Christchurch, New Zealand
| | - Anita Ayer
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
- Heart Research Institute, The University of Sydney, Sydney, Australia.
- St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
- Heart Research Institute, The University of Sydney, Sydney, Australia.
- St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia.
| |
Collapse
|
26
|
Otsuka Y, Bai X, Tanaka Y, Ihara E, Chinen T, Ogino H, Ogawa Y. Involvement of interstitial cells of Cajal in nicotinic acetylcholine receptor-induced relaxation of the porcine lower esophageal sphincter. Eur J Pharmacol 2021; 910:174491. [PMID: 34506779 DOI: 10.1016/j.ejphar.2021.174491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/15/2022]
Abstract
The interstitial cells of Cajal (ICCs) play an important role in coordinated gastrointestinal motility. The present study aimed to elucidate whether or how ICCs are involved in the lower esophageal sphincter (LES) relaxation induced by stimulation of the nicotinic acetylcholine receptor. The application of 1,1-dimethyl-4-phenyl-piperazinium (DMPP; a nicotinic acetylcholine receptor agonist) induced a transient relaxation in the circular smooth muscle of the porcine LES. DMPP-induced relaxation was abolished by not only 1 μM tetrodotoxin but also the inhibition of ICC activity by pretreatment with 100 μM carbenoxolone (a gap junction inhibitor), pretreatment with 100 μM CaCCinh-A01 (an anoctamin-1 blocker acting as a calcium-activated chloride channel inhibitor), and pretreatment with Cl--free solution. However, pretreatment with 100 μM Nω-nitro-L-arginine methyl ester had little effect on DMPP-induced relaxation. Furthermore, DMPP-induced relaxation was inhibited by pretreatment with 1 mM suramin, a purinergic P2 receptor antagonist, but not by 1 μM VIP (6-28), a vasoactive intestinal peptide (VIP) receptor antagonist. Stimulation of the purinergic P2 receptor with adenosine triphosphate (ATP) induced relaxation, which was abolished by the inhibition of ICC activity by pretreatment with CaCCinh-A01. In conclusion, membrane hyperpolarization of the ICCs via the activation of anoctamin-1 plays a central role in DMPP-induced relaxation. ATP may be a neurotransmitter for inhibitory enteric neurons, which stimulate the ICCs. The ICCs act as the interface of neurotransmission of nicotinic acetylcholine receptor in order to induce LES relaxation.
Collapse
Affiliation(s)
- Yoshihiro Otsuka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Xiaopeng Bai
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshimasa Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eikichi Ihara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takatoshi Chinen
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruei Ogino
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
27
|
Roberts AM, Moulana NZ, Jagadapillai R, Cai L, Gozal E. Intravital assessment of precapillary pulmonary arterioles of type 1 diabetic mice shows oxidative damage and increased tone in response to NOS inhibition. J Appl Physiol (1985) 2021; 131:1552-1564. [PMID: 34590907 PMCID: PMC11961051 DOI: 10.1152/japplphysiol.00395.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/07/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022] Open
Abstract
Microvascular dilation, important for peripheral tissue glucose distribution, also modulates alveolar perfusion and is inhibited by loss of bioavailable nitric oxide (NO) in diabetes mellitus (DM). We hypothesized that DM-induced oxidative stress decreases bioavailable NO and pulmonary precapillary arteriolar diameter, causing endothelial injury. We examined subpleural pulmonary arterioles after acute NO synthase (NOS) inhibition with NG-nitro-l-arginine methyl ester (l-NAME) in streptozotocin (STZ)- and saline (CTRL)-treated C57BL/6J mice. Microvascular changes were assessed by intravital microscopy in the right lung of anesthetized mice with open chest and ventilated lungs. Arteriolar tone in pulmonary arterioles (27.2-48.7 µm diameter) increased in CTRL mice (18.0 ± 11% constriction, P = 0.034, n = 5) but decreased in STZ mice (13.6 ± 7.5% dilation, P = 0.009, n = 5) after l-NAME. Lung tissue dihydroethidium (DHE) fluorescence (superoxide), inducible NOS expression, and protein nitrosylation (3-nitrotyrosine) increased in STZ mice and correlated with increased glucose levels (103.8 ± 8.8 mg/dL). Fluorescently labeled fibrinogen administration and fibrinogen immunostaining showed fibrinogen adhesion, indicating endothelial injury in STZ mice. In CTRL mice, vasoconstriction to l-NAME was likely due to the loss of bioavailable NO. Vasodilation in STZ mice may be due to decreased formation of a vasoconstrictor or emergence of a vasodilator. These findings provide novel evidence that DM targets the pulmonary microcirculation and that decreased NO bioavailability and increased precapillary arteriolar tone could potentially lead to ventilation-perfusion abnormalities, exacerbating systemic DM complications.NEW & NOTEWORTHY Diabetes pulmonary and microvascular consequences are well recognized but have not been characterized. We assessed lung microvascular changes in a live anesthetized mouse model of type 1 diabetes, using a novel intravital microscopy technique. Our results show new evidence that a diabetes-induced decrease in lung nitric oxide bioavailability underlies oxidative damage, enhanced platelet activation, and endothelial injury causing pulmonary microvascular dysfunction and altered vasoreactivity. These findings could provide novel strategies to prevent or reverse diabetes systemic consequences.
Collapse
Affiliation(s)
- Andrew M Roberts
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| | - Nayeem Z Moulana
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Rekha Jagadapillai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| | - Evelyne Gozal
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
28
|
Matthies M, Rosenstand K, Nissen I, Muitjens S, Riber LP, De Mey JGR, Bloksgaard M. Nitric oxide (NO) synthase but not NO, HNO or H 2 O 2 mediates endothelium-dependent relaxation of resistance arteries from patients with cardiovascular disease. Br J Pharmacol 2021; 179:1049-1064. [PMID: 34664280 DOI: 10.1111/bph.15712] [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: 02/11/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Superoxide anions can reduce the bioavailability and actions of endothelium-derived NO. In human resistance-sized arteries, endothelium-dependent vasodilatation can be mediated by H2 O2 instead of NO. Here, we tested the hypothesis that in resistance arteries from patients with cardiovascular disease, endothelium-dependent vasodilatation is mediated by a reactive oxygen species and not impaired by oxidative stress. EXPERIMENTAL APPROACH Small arteries were isolated from biopsies of the parietal pericardium of patients undergoing elective cardiothoracic surgery and were studied using immunohistochemical and organ chamber techniques. KEY RESULTS NO synthases 1, 2 and 3, superoxide dismutase 1 and catalase proteins were observed in the microvascular wall. Relaxing responses to bradykinin were endothelium dependent. During submaximal depolarization-induced contraction, bradykinin-mediated relaxations were inhibited by inhibitors of NO synthases (NOS) and soluble guanylyl cyclase (sGC) but not by scavengers of NO or HNO, inhibitors of cyclooxygenases, neuronal NO synthase, superoxide dismutase or catalase, or by exogenous catalase. During contraction stimulated by endothelin-1, these relaxations were not reduced by any of these interventions except DETCA, which caused a small reduction. CONCLUSION AND IMPLICATIONS In resistance arteries from patients with cardiovascular disease, endothelium-dependent relaxations seem not to be mediated by NO, HNO or H2 O2 , although NOS and sGC can be involved. These vasodilator responses continue during excessive oxidative stress.
Collapse
Affiliation(s)
- Maximilian Matthies
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Inger Nissen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stan Muitjens
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lars P Riber
- Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
| | - Jo G R De Mey
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Pharmacology and Personalized Medicine, Maastricht University, Maastricht, The Netherlands
| | - Maria Bloksgaard
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
29
|
Takahashi J, Suda A, Nishimiya K, Godo S, Yasuda S, Shimokawa H. Pathophysiology and Diagnosis of Coronary Functional Abnormalities. Eur Cardiol 2021; 16:e30. [PMID: 34603510 PMCID: PMC8478147 DOI: 10.15420/ecr.2021.23] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/07/2021] [Indexed: 01/17/2023] Open
Abstract
Approximately one-half of patients undergoing diagnostic coronary angiography for angina have no significant coronary atherosclerotic stenosis. This clinical condition has recently been described as ischaemia with non-obstructive coronary arteries (INOCA). Coronary functional abnormalities are central to the pathogenesis of INOCA, including epicardial coronary spasm and coronary microvascular dysfunction composed of a variable combination of increased vasoconstrictive reactivity and/or reduced vasodilator function. During the last decade - in INOCA patients in particular - evidence for the prognostic impact of coronary functional abnormalities has accumulated and various non-invasive and invasive diagnostic techniques have enabled the evaluation of coronary vasomotor function in a comprehensive manner. In this review, the authors briefly summarise the recent advances in the understanding of pathophysiology and diagnosis of epicardial coronary artery spasm and coronary microvascular dysfunction.
Collapse
Affiliation(s)
- Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Akira Suda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Kensuke Nishimiya
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| |
Collapse
|
30
|
Viswambharan H, Yuldasheva NY, Imrie H, Bridge K, Haywood NJ, Skromna A, Hemmings KE, Clark ER, Gatenby VK, Cordell P, Simmons KJ, Makava N, Abudushalamu Y, Endesh N, Brown J, Walker AMN, Futers ST, Porter KE, Cubbon RM, Naseem K, Shah AM, Beech DJ, Wheatcroft SB, Kearney MT, Sukumar P. Novel Paracrine Action of Endothelium Enhances Glucose Uptake in Muscle and Fat. Circ Res 2021; 129:720-734. [PMID: 34420367 PMCID: PMC8448413 DOI: 10.1161/circresaha.121.319517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Nadira Y Yuldasheva
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Helen Imrie
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katherine Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natalie J Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Anna Skromna
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Hemmings
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Emily R Clark
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - V Kate Gatenby
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Paul Cordell
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natallia Makava
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Yilizila Abudushalamu
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Naima Endesh
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Jane Brown
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Andrew M N Walker
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Simon T Futers
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Porter
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Khalid Naseem
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, King's College London (A.M.S.)
| | - David J Beech
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Piruthivi Sukumar
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| |
Collapse
|
31
|
Hamad A, Ozkan MH, Uma S. Trimethylamine-N-oxide (TMAO) Selectively Disrupts Endothelium-Dependent Hyperpolarization-Type Relaxations in a Time-Dependent Manner in Rat Superior Mesenteric Artery. Biol Pharm Bull 2021; 44:1220-1229. [PMID: 34471050 DOI: 10.1248/bpb.b20-00767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The vascular action of trimethylamine-N-oxide (TMAO)-the gut microbiota-derived metabolite-in contributing cardiovascular disease is a controversial topic. A recent study has shown that acute exposure of TMAO at moderate concentrations inhibits endothelium-dependent hyperpolarization (EDH)-type relaxations selectively in rat isolated femoral arteries, but not in mesenteric arteries. Here we determined the efficacy of higher TMAO concentrations with longer exposure times on vascular reactivity in rat isolated superior mesenteric arteries. Acetylcholine-induced EDH-type relaxations were examined before and after incubation with TMAO (0.1-10 mM) at increasing exposure times (1-24 h). One- and 4-h-incubations with TMAO at 0.1-3 mM did not cause any change in EDH-type relaxations. However, when the incubation time was increased to 24 h, responses to acetylcholine were reduced in arteries incubated with 1-3 mM TMAO. In addition, at higher TMAO concentration (10 mM) the decrease in EDH relaxations could be detected both in 4-h- and 24-h-incubations. The EDH-relaxations were preserved in rings incubated with 10 mM TMAO for 24 h in the presence of SKA-31 (10 µM), the small (SKCa)- and intermediate (IKCa)-conductance calcium-activated potassium channel activator. Contractile responses to phenylephrine increased in arteries exposed to 10 mM TMAO for 24 h. Interestingly, nitric oxide (NO)-mediated relaxations remained unchanged in arteries treated for 24 h at any TMAO concentration. Our study revealed that TMAO selectively disrupted EDH-type relaxations time-dependently without interfering with NO-induced vasodilation in rat isolated mesenteric arteries. Disruption of these relaxations may help explain the causal role of elevated TMAO levels in certain vascular diseases.
Collapse
Affiliation(s)
- Abdelrahman Hamad
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University
| | | | - Serdar Uma
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University
| |
Collapse
|
32
|
Liu B, Zhou Y. Endothelium-dependent contraction: The non-classical action of endothelial prostacyclin, its underlying mechanisms, and implications. FASEB J 2021; 35:e21877. [PMID: 34449098 DOI: 10.1096/fj.202101077r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 02/05/2023]
Abstract
Although commonly thought to produce prostacyclin (prostaglandin I2 ; PGI2 ) that evokes vasodilatation and protects vessels from the development of diseases, the endothelial cyclooxygenase (COX)-mediated metabolism has also been found to release substance(s) called endothelium-derived contracting factor(s) (EDCF) that causes endothelium-dependent contraction and implicates in endothelial dysfunction of disease conditions. Various mechanisms have been proposed for the process; however, the major endothelial COX metabolite PGI2 , which has been classically considered to activate the I prostanoid receptor (IP) that mediates vasodilatation and opposes the effects of thromboxane (Tx) A2 produced by COX in platelets, emerges as a major EDCF in health and disease conditions. Our recent studies from genetically altered mice further suggest that vasomotor reactions to PGI2 are collectively modulated by IP, the vasoconstrictor Tx-prostanoid receptor (TP; the prototype receptor of TxA2 ) and E prostanoid receptor-3 (EP3; a vasoconstrictor receptor of PGE2 ) although with differences in potency and efficacy; a contraction to PGI2 reflects activities of TP and/or EP3 outweighing that of the concurrently activated IP. Here, we discuss the history of endothelium-dependent contraction, evidences that support the above hypothesis, proposed mechanisms for the varied reactions to endothelial PGI2 synthesis as well as the relation of its dilator activity to the effect of another NO-independent vasodilator mechanism, the endothelium-derived hyperpolarizing factor. Also, we address the possible pathological and therapeutic implications as well as questions remaining to be resolved or limitations of our above findings obtained from genetically altered mouse models.
Collapse
Affiliation(s)
- Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| |
Collapse
|
33
|
Troy AM, Cheng HM. Human microvascular reactivity: a review of vasomodulating stimuli and non-invasive imaging assessment. Physiol Meas 2021; 42. [PMID: 34325417 DOI: 10.1088/1361-6579/ac18fd] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/29/2021] [Indexed: 11/11/2022]
Abstract
The microvasculature serves an imperative function in regulating perfusion and nutrient exchange throughout the body, adaptively altering blood flow to preserve hemodynamic and metabolic homeostasis. Its normal functioning is vital to tissue health, whereas its dysfunction is present in many chronic conditions, including diabetes, heart disease, and cognitive decline. As microvascular dysfunction often appears early in disease progression, its detection can offer early diagnostic information. To detect microvascular dysfunction, one uses imaging to probe the microvasculature's ability to react to a stimulus, also known as microvascular reactivity (MVR). An assessment of MVR requires an integrated understanding of vascular physiology, techniques for stimulating reactivity, and available imaging methods to capture the dynamic response. Practical considerations, including compatibility between the selected stimulus and imaging approach, likewise require attention. In this review, we provide a comprehensive foundation necessary for informed imaging of MVR, with a particular focus on the challenging endeavor of assessing microvascular function in deep tissues.
Collapse
Affiliation(s)
- Aaron M Troy
- Institute of Biomedical Engineering, University of Toronto, Toronto, CANADA
| | | |
Collapse
|
34
|
Roos CM, Zhang B, Hagler MA, Arghami A, Miller JD. MnSOD protects against vascular calcification independent of changes in vascular function in hypercholesterolemic mice. Atherosclerosis 2021; 331:31-37. [PMID: 34147244 DOI: 10.1016/j.atherosclerosis.2021.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/05/2021] [Accepted: 06/04/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS The overall goal of this study was to determine the effects of MnSOD-deficiency on vascular structure and function in hypercholesterolemic mice. Previous work suggested that increases in mitochondrial-derived reactive oxygen species (ROS) can exacerbate vascular dysfunction and atherosclerosis. It remains unknown, however, how MnSOD-deficiency and local compensatory mechanisms impact atherosclerotic plaque composition. METHODS AND RESULTS We used a hypercholesterolemic mouse model (ldlr-/-/ApoB100/100; LA), either wild-type for MnSOD (LA-MnSOD+/+) or MnSOD-haploinsufficient (LA-MnSOD+/-), that was fed a western diet for either 3 or 6 months. Consistent with previous reports, reductions of MnSOD did not significantly worsen hypercholesterolemia-induced endothelial dysfunction in the aorta. Critically, dramatic impairment of vascular function with Nox2 inhibition or catalase pretreatment suggested the presence of a significant NO-independent vasodilatory mechanism in LA-MnSOD+/- mice (e.g. H2O2). Despite remarkably well-preserved overall vascular relaxation, loss of mitochondrial antioxidant capacity in LA-MnSOD+/- mice significantly increased osteogenic signalling and vascular calcification compared to the LA-MnSOD+/+ littermates. CONCLUSIONS Collectively, these data are the first to suggest that loss of mitochondrial antioxidant capacity in hypercholesterolemic mice results in dramatic upregulation of NADPH oxidase-derived H2O2. While this appears to be adaptive in the context of preserving overall endothelium-dependent relaxation and vascular function, these increases in ROS appear to be remarkably maladaptive and deleterious in the context of vascular calcification.
Collapse
Affiliation(s)
| | - Bin Zhang
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Arman Arghami
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jordan D Miller
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA; Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Iacobini C, Vitale M, Pesce C, Pugliese G, Menini S. Diabetic Complications and Oxidative Stress: A 20-Year Voyage Back in Time and Back to the Future. Antioxidants (Basel) 2021; 10:727. [PMID: 34063078 PMCID: PMC8147954 DOI: 10.3390/antiox10050727] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Twenty years have passed since Brownlee and colleagues proposed a single unifying mechanism for diabetic complications, introducing a turning point in this field of research. For the first time, reactive oxygen species (ROS) were identified as the causal link between hyperglycemia and four seemingly independent pathways that are involved in the pathogenesis of diabetes-associated vascular disease. Before and after this milestone in diabetes research, hundreds of articles describe a role for ROS, but the failure of clinical trials to demonstrate antioxidant benefits and some recent experimental studies showing that ROS are dispensable for the pathogenesis of diabetic complications call for time to reflect. This twenty-year journey focuses on the most relevant literature regarding the main sources of ROS generation in diabetes and their role in the pathogenesis of cell dysfunction and diabetic complications. To identify future research directions, this review discusses the evidence in favor and against oxidative stress as an initial event in the cellular biochemical abnormalities induced by hyperglycemia. It also explores possible alternative mechanisms, including carbonyl stress and the Warburg effect, linking glucose and lipid excess, mitochondrial dysfunction, and the activation of alternative pathways of glucose metabolism leading to vascular cell injury and inflammation.
Collapse
Affiliation(s)
- Carla Iacobini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Carlo Pesce
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and Maternal Infantile Sciences (DINOGMI), Department of Excellence of MIUR, University of Genoa Medical School, 16132 Genoa, Italy;
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| |
Collapse
|
36
|
Godo S, Takahashi J, Yasuda S, Shimokawa H. Role of Inflammation in Coronary Epicardial and Microvascular Dysfunction. Eur Cardiol 2021; 16:e13. [PMID: 33897839 PMCID: PMC8054350 DOI: 10.15420/ecr.2020.47] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/16/2021] [Indexed: 01/09/2023] Open
Abstract
There is accumulating evidence highlighting a close relationship between inflammation and coronary microvascular dysfunction (CMD) in various experimental and clinical settings, with major clinical implications. Chronic low-grade vascular inflammation plays important roles in the underlying mechanisms behind CMD, especially in patients with coronary artery disease, obesity, heart failure with preserved ejection fraction and chronic inflammatory rheumatoid diseases. The central mechanisms of coronary vasomotion abnormalities comprise enhanced coronary vasoconstrictor reactivity, reduced endothelium-dependent and -independent coronary vasodilator capacity and increased coronary microvascular resistance, where inflammatory mediators and responses are substantially involved. How to modulate CMD to improve clinical outcomes of patients with the disorder and whether CMD management by targeting inflammatory responses can benefit patients remain challenging questions in need of further research. This review provides a concise overview of the current knowledge of the involvement of inflammation in the pathophysiology and molecular mechanisms of CMD from bench to bedside.
Collapse
Affiliation(s)
- Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| |
Collapse
|
37
|
Dagher O, Mury P, Thorin-Trescases N, Noly PE, Thorin E, Carrier M. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:658400. [PMID: 33860002 PMCID: PMC8042157 DOI: 10.3389/fcvm.2021.658400] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelium occupies a catalog of functions that contribute to the homeostasis of the cardiovascular system. It is a physically active barrier between circulating blood and tissue, a regulator of the vascular tone, a biochemical processor and a modulator of coagulation, inflammation, and immunity. Given these essential roles, it comes to no surprise that endothelial dysfunction is prodromal to chronic age-related diseases of the heart and arteries, globally termed cardiovascular diseases (CVD). An example would be ischemic heart disease (IHD), which is the main cause of death from CVD. We have made phenomenal advances in treating CVD, but the aging endothelium, as it senesces, always seems to out-run the benefits of medical and surgical therapies. Remarkably, many epidemiological studies have detected a correlation between a flavonoid-rich diet and a lower incidence of mortality from CVD. Quercetin, a member of the flavonoid class, is a natural compound ubiquitously found in various food sources such as fruits, vegetables, seeds, nuts, and wine. It has been reported to have a wide range of health promoting effects and has gained significant attention over the years. A growing body of evidence suggests quercetin could lower the risk of IHD by mitigating endothelial dysfunction and its risk factors, such as hypertension, atherosclerosis, accumulation of senescent endothelial cells, and endothelial-mesenchymal transition (EndoMT). In this review, we will explore these pathophysiological cascades and their interrelation with endothelial dysfunction. We will then present the scientific evidence to quercetin's anti-atherosclerotic, anti-hypertensive, senolytic, and anti-EndoMT effects. Finally, we will discuss the prospect for its clinical use in alleviating myocardial ischemic injuries in IHD.
Collapse
Affiliation(s)
- Olina Dagher
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Pauline Mury
- Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | | | - Pierre Emmanuel Noly
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Eric Thorin
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Michel Carrier
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
38
|
Neves KB, Morris HE, Alves-Lopes R, Muir KW, Moreton F, Delles C, Montezano AC, Touyz RM. Peripheral arteriopathy caused by Notch3 gain-of-function mutation involves ER and oxidative stress and blunting of NO/sGC/cGMP pathway. Clin Sci (Lond) 2021; 135:753-773. [PMID: 33681964 DOI: 10.1042/cs20201412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
Notch3 mutations cause Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), which predisposes to stroke and dementia. CADASIL is characterised by vascular dysfunction and granular osmiophilic material (GOM) accumulation in cerebral small vessels. Systemic vessels may also be impacted by Notch3 mutations. However vascular characteristics and pathophysiological processes remain elusive. We investigated mechanisms underlying the peripheral vasculopathy mediated by CADASIL-causing Notch3 gain-of-function mutation. We studied: (i) small arteries and vascular smooth muscle cells (VSMCs) from TgNotch3R169C mice (CADASIL model), (ii) VSMCs from peripheral arteries from CADASIL patients, and (iii) post-mortem brains from CADASIL individuals. TgNotch3R169C vessels exhibited GOM deposits, increased vasoreactivity and impaired vasorelaxation. Hypercontractile responses were normalised by fasudil (Rho kinase inhibitor) and 4-phenylbutyrate (4-PBA; endoplasmic-reticulum (ER) stress inhibitor). Ca2+ transients and Ca2+ channel expression were increased in CADASIL VSMCs, with increased expression of Rho guanine nucleotide-exchange factors (GEFs) and ER stress proteins. Vasorelaxation mechanisms were impaired in CADASIL, evidenced by decreased endothelial nitric oxide synthase (eNOS) phosphorylation and reduced cyclic guanosine 3',5'-monophosphate (cGMP) levels, with associated increased soluble guanylate cyclase (sGC) oxidation, decreased sGC activity and reduced levels of the vasodilator hydrogen peroxide (H2O2). In VSMCs from CADASIL patients, sGC oxidation was increased and cGMP levels decreased, effects normalised by fasudil and 4-PBA. Cerebral vessels in CADASIL patients exhibited significant oxidative damage. In conclusion, peripheral vascular dysfunction in CADASIL is associated with altered Ca2+ homoeostasis, oxidative stress and blunted eNOS/sGC/cGMP signaling, processes involving Rho kinase and ER stress. We identify novel pathways underlying the peripheral arteriopathy induced by Notch3 gain-of-function mutation, phenomena that may also be important in cerebral vessels.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Hannah E Morris
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Keith W Muir
- Institute of Neuroscience and Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, U.K
| | - Fiona Moreton
- Institute of Neuroscience and Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, U.K
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| |
Collapse
|
39
|
Stanley CP, Stocker R. Regulation of vascular tone and blood pressure by singlet molecular oxygen in inflammation. Curr Opin Nephrol Hypertens 2021; 30:145-150. [PMID: 33427761 DOI: 10.1097/mnh.0000000000000679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The principle aim of this review is to prompt vascular researchers interested in vascular inflammation and oxidative stress to consider singlet molecular oxygen (1O2) as a potentially relevant contributor. A secondary goal is to propose novel treatment strategies to address haemodynamic complications associated with septic shock. RECENT FINDINGS Increased inflammation and oxidative stress are hallmarks of a range of vascular diseases. We recently showed that in systemic inflammation and oxidative stress associated with models of inflammation including sepsis, the tryptophan catabolizing enzyme indoleamine 2,3-dioxygenase-1 (Ido1) contributes to hypotension and decreased blood pressure through production of singlet molecular oxygen (1O2). Once formed, 1O2 converts tryptophan bound to Ido1 to a vasoactive hydroperoxide which decreases arterial tone and blood pressure via oxidation of a specific cysteine residue of protein kinase G1α. SUMMARY These works show, for the first time, that 1O2 contributes to arterial redox signalling and that Ido1 contributes to the regulation of blood pressure through production of a novel tryptophan-derived hydroperoxide, thus presenting a new signalling pathway as novel target in the treatment of blood pressure disorders such as sepsis.
Collapse
Affiliation(s)
- Christopher P Stanley
- Heart Research Institute, Newtown
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Roland Stocker
- Heart Research Institute, Newtown
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Paulo M, Costa DEFR, Bonaventura D, Lunardi CN, Bendhack LM. Nitric Oxide Donors as Potential Drugs for the Treatment of Vascular Diseases Due to Endothelium Dysfunction. Curr Pharm Des 2021; 26:3748-3759. [PMID: 32427079 DOI: 10.2174/1381612826666200519114442] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/07/2020] [Indexed: 11/22/2022]
Abstract
Endothelial dysfunction and consequent vasoconstriction are a common condition in patients with hypertension and other cardiovascular diseases. Endothelial cells produce and release vasodilator substances that play a pivotal role in normal vascular tone. The mechanisms underlying endothelial dysfunction are multifactorial. However, enhanced reactive oxygen species (ROS) production and consequent vasoconstriction instead of endothelium-derived relaxant generation and consequent vasodilatation contribute to this dysfunction considerably. The main targets of the drugs that are currently used to treat vascular diseases concerning enzyme activities and protein functions that are impaired by endothelial nitric oxide synthase (eNOS) uncoupling and ROS production. Nitric oxide (NO) bioavailability can decrease due to deficient NO production by eNOS and/or NO release to vascular smooth muscle cells, which impairs endothelial function. Considering the NO cellular mechanisms, tackling the issue of eNOS uncoupling could avoid endothelial dysfunction: provision of the enzyme cofactor tetrahydrobiopterin (BH4) should elicit NO release from NO donors, to activate soluble guanylyl cyclase. This should increase cyclic guanosine-monophosphate (cGMP) generation and inhibit phosphodiesterases (especially PDE5) that selectively degrade cGMP. Consequently, protein kinase-G should be activated, and K+ channels should be phosphorylated and activated, which is crucial for cell membrane hyperpolarization and vasodilation and/or inhibition of ROS production. The present review summarizes the current concepts about the vascular cellular mechanisms that underlie endothelial dysfunction and which could be the target of drugs for the treatment of patients with cardiovascular disease.
Collapse
Affiliation(s)
- Michele Paulo
- Department Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirao Preto- University of Sao Paulo Av. Do Cafe SN, Brazil
| | - Daniela E F R Costa
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Daniella Bonaventura
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Claure N Lunardi
- Laboratory of Photochemistry and Nanobiotechnology, University of Brasilia, Brasilia, Brazil
| | - Lusiane M Bendhack
- Department Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirao Preto- University of Sao Paulo Av. Do Cafe SN, Brazil
| |
Collapse
|
41
|
Costa TJ, Barros PR, Arce C, Santos JD, da Silva-Neto J, Egea G, Dantas AP, Tostes RC, Jiménez-Altayó F. The homeostatic role of hydrogen peroxide, superoxide anion and nitric oxide in the vasculature. Free Radic Biol Med 2021; 162:615-635. [PMID: 33248264 DOI: 10.1016/j.freeradbiomed.2020.11.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/08/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
Reactive oxygen and nitrogen species are produced in a wide range of physiological reactions that, at low concentrations, play essential roles in living organisms. There is a delicate equilibrium between formation and degradation of these mediators in a healthy vascular system, which contributes to maintaining these species under non-pathological levels to preserve normal vascular functions. Antioxidants scavenge reactive oxygen and nitrogen species to prevent or reduce damage caused by excessive oxidation. However, an excessive reductive environment induced by exogenous antioxidants may disrupt redox balance and lead to vascular pathology. This review summarizes the main aspects of free radical biochemistry (formation, sources and elimination) and the crucial actions of some of the most biologically relevant and well-characterized reactive oxygen and nitrogen species (hydrogen peroxide, superoxide anion and nitric oxide) in the physiological regulation of vascular function, structure and angiogenesis. Furthermore, current preclinical and clinical evidence is discussed on how excessive removal of these crucial responses by exogenous antioxidants (vitamins and related compounds, polyphenols) may perturb vascular homeostasis. The aim of this review is to provide information of the crucial physiological roles of oxidation in the endothelium, vascular smooth muscle cells and perivascular adipose tissue for developing safer and more effective vascular interventions with antioxidants.
Collapse
Affiliation(s)
- Tiago J Costa
- Pharmacology Department, Ribeirao Preto Medical School, University of São Paulo, Brazil.
| | | | - Cristina Arce
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain; Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)-University of Barcelona, Barcelona, Spain; Institut de Nanociencies i Nanotecnologia (IN2UB), University of Barcelona, Barcelona, Spain
| | | | - Júlio da Silva-Neto
- Pharmacology Department, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Gustavo Egea
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain; Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)-University of Barcelona, Barcelona, Spain; Institut de Nanociencies i Nanotecnologia (IN2UB), University of Barcelona, Barcelona, Spain
| | - Ana Paula Dantas
- Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rita C Tostes
- Pharmacology Department, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutics and Toxicology, Neuroscience Institute, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
42
|
Godo S, Shimokawa H. Gender Differences in Endothelial Function and Coronary Vasomotion Abnormalities. GENDER AND THE GENOME 2020. [DOI: 10.1177/2470289720957012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction: Structural and functional abnormalities of coronary microvasculature, referred to as coronary microvascular dysfunction (CMD), have been implicated in a wide range of cardiovascular diseases and have gained growing attention in patients with chest pain with no obstructive coronary artery disease, especially in females. The central mechanisms of coronary vasomotion abnormalities encompass enhanced coronary vasoconstrictive reactivity (ie, coronary spasm), reduced endothelium-dependent and -independent coronary vasodilator capacities, and increased coronary microvascular resistance. The 2 major endothelium-derived relaxing factors, nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH) factors, modulate vascular tone in a distinct vessel size–dependent manner; NO mainly mediates vasodilatation of relatively large, conduit vessels, while EDH factors in small resistance vessels. Endothelium-dependent hyperpolarization–mediated vasodilatation is more prominent in female resistance arteries, where estrogens exert beneficial effects on endothelium-dependent vasodilatation via multiple mechanisms. In the clinical settings, therapeutic approaches targeting NO are disappointing for the treatment of various cardiovascular diseases, where endothelial dysfunction and CMD are substantially involved. Significance: In this review, we will discuss the current knowledge on the pathophysiology and molecular mechanisms of endothelial function and coronary vasomotion abnormalities from bench to bedside, with a special reference to gender differences. Results: Recent experimental and clinical studies have demonstrated distinct gender differences in endothelial function and coronary vasomotion abnormalities with major clinical implications. Moreover, recent landmark clinical trials regarding the management of stable coronary artery disease have questioned the benefit of percutaneous coronary intervention, supporting the importance of the coronary microvascular physiology. Conclusion: Further characterization and a better understanding of the gender differences in basic vascular biology as well as those in cardiovascular diseases are indispensable to improve health care and patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
43
|
Important Roles of Endothelium-Dependent Hyperpolarization in Coronary Microcirculation and Cardiac Diastolic Function in Mice. J Cardiovasc Pharmacol 2020; 75:31-40. [PMID: 31895878 DOI: 10.1097/fjc.0000000000000763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Endothelium-dependent hyperpolarization (EDH) factor is one of endothelium-derived relaxing factors and plays important roles especially in microvessels. We have previously demonstrated that endothelium-derived hydrogen peroxide (H2O2) is an EDH factor produced by all types of nitric oxide synthases (NOSs), including endothelial NOS (eNOS), neuronal NOS (nNOS), and inducible NOS. Recent studies have suggested the association between coronary microvascular dysfunction and cardiac diastolic dysfunction. However, the role of EDH in this issue remains to be fully elucidated. We thus examined whether EDH plays an important role in coronary microcirculation and if so, whether endothelial dysfunction, especially impaired EDH, is involved in the pathogenesis of cardiac diastolic dysfunction in mice. Using a Langendorff-perfused heart experiment, we examined the increase in coronary flow in response to bradykinin in the presence of indomethacin and N-nitro-L-arginine (EDH condition) in wild-type, eNOS-knockout (KO), and nNOS/eNOS-double-KO mice. Compared with wild-type mice, EDH-mediated relaxations were increased in eNOS-KO mice but were significantly reduced in n/eNOS-KO mice. Catalase, a specific H2O2 scavenger, markedly inhibited EDH-mediated relaxations in all 3 genotypes, indicating compensatory roles of nNOS-derived H2O2 as an EDH factor in coronary microcirculation. Although both eNOS-KO and n/eNOS-KO mice exhibited similar extents of cardiac morphological changes, only n/eNOS-KO mice exhibited cardiac diastolic dysfunction. The expression of oxidized protein kinase G I-α (PKGIα) in the heart was significantly increased in eNOS-KO mice compared with n/eNOS-KO mice. These results indicate that EDH/H2O2 plays important roles in maintaining coronary microcirculation and cardiac diastolic function through oxidative PKGIα activation.
Collapse
|
44
|
Urner S, Ho F, Jha JC, Ziegler D, Jandeleit-Dahm K. NADPH Oxidase Inhibition: Preclinical and Clinical Studies in Diabetic Complications. Antioxid Redox Signal 2020; 33:415-434. [PMID: 32008354 DOI: 10.1089/ars.2020.8047] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Oxidative stress plays a critical role in the development and progression of serious micro- and macrovascular complications of diabetes. Nicotinamide adenine dinucleotide phosphate oxidase (NOX)-derived reactive oxygen species (ROS) significantly contribute to oxidative stress-associated inflammatory pathways that lead to tissue damage of different organs, including the kidneys, retina, brain, nerves, and the cardiovascular system. Recent Advances: Preclinical studies, including genetic-modified mouse models or cell culture models, have revealed the role of specific NOX isoforms in different diabetic complications, and suggested them as a promising target for the treatment of these diseases. Critical Issues: In this review, we provide an overview of the role of ROS and oxidative stress in macrovascular complications, such as stroke, myocardial infarction, coronary artery disease, and peripheral vascular disease that are all mainly driven by atherosclerosis, as well as microvascular complications, such as diabetic retinopathy, nephropathy, and neuropathy. We summarize conducted genetic deletion studies of different Nox isoforms as well as pharmacological intervention studies using NOX inhibitors in the context of preclinical as well as clinical research on diabetic complications. Future Directions: We outline the isoforms that are most promising for future clinical trials in the context of micro- and macrovascular complications of diabetes.
Collapse
Affiliation(s)
- Sofia Urner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Florence Ho
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Jay C Jha
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Karin Jandeleit-Dahm
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
45
|
Six I, Flissi N, Lenglet G, Louvet L, Kamel S, Gallet M, Massy ZA, Liabeuf S. Uremic Toxins and Vascular Dysfunction. Toxins (Basel) 2020; 12:toxins12060404. [PMID: 32570781 PMCID: PMC7354618 DOI: 10.3390/toxins12060404] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular dysfunction is an essential element found in many cardiovascular pathologies and in pathologies that have a cardiovascular impact such as chronic kidney disease (CKD). Alteration of vasomotricity is due to an imbalance between the production of relaxing and contracting factors. In addition to becoming a determining factor in pathophysiological alterations, vascular dysfunction constitutes the first step in the development of atherosclerosis plaques or vascular calcifications. In patients with CKD, alteration of vasomotricity tends to emerge as being a new, less conventional, risk factor. CKD is characterized by the accumulation of uremic toxins (UTs) such as phosphate, para-cresyl sulfate, indoxyl sulfate, and FGF23 and, consequently, the deleterious role of UTs on vascular dysfunction has been explored. This accumulation of UTs is associated with systemic alterations including inflammation, oxidative stress, and the decrease of nitric oxide production. The present review proposes to summarize our current knowledge of the mechanisms by which UTs induce vascular dysfunction.
Collapse
Affiliation(s)
- Isabelle Six
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
- Correspondence: ; Tel./Fax: +03-22-82-54-25
| | - Nadia Flissi
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Gaëlle Lenglet
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Loïc Louvet
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Said Kamel
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
- Amiens-Picardie University Hospital, Human Biology Center, 80054 Amiens, France
| | - Marlène Gallet
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Ziad A. Massy
- Service de Néphrologie et Dialyse, Assistance Publique—Hôpitaux de Paris (APHP), Hôpital Universitaire Ambroise Paré, 92100 Boulogne Billancourt, France;
- INSERM U1018, Equipe 5, CESP (Centre de Recherche en Épidémiologie et Santé des Populations), Université Paris Saclay et Université Versailles Saint Quentin en Yvelines, 94800 Villejuif, France
| | - Sophie Liabeuf
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
- Pharmacology Department, Amiens University Hospital, 80025 Amiens, France
| |
Collapse
|
46
|
Cunha TRD, Giesen JAS, Rouver WN, Costa ED, Grando MD, Lemos VS, Bendhack LM, Santos RLD. Effects of progesterone treatment on endothelium-dependent coronary relaxation in ovariectomized rats. Life Sci 2020; 247:117391. [PMID: 32017871 DOI: 10.1016/j.lfs.2020.117391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 12/24/2022]
Abstract
AIM Although progesterone (P4) has a beneficial effect on the cardiovascular system, P4 actions on the coronary bed have not yet been fully elucidated. This study evaluated the effect of progesterone treatment on endothelium-dependent coronary vascular reactivity in Wistar rats. MAIN METHODS Eight-week-old adult rats were divided into Sham, Ovariectomized (OVX), Ovariectomized and progesterone treated (OVX P4). The OVX P4 group received daily doses of progesterone (2 mg/kg/day). Vascular reactivity was assessed by a modified Langendorff technique. The intensity of eNOS, Akt, and gp91phox protein expression was quantified by Western blotting. Superoxide anion (O2●-) and hydrogen peroxide (H2O2) production was measured by dihydroethidium and 2',7'-dichlorofluorescein, respectively. KEY FINDINGS Treatment with P4 was able to prevent the reduction in baseline coronary perfusion pressure induced by ovariectomy. We observed that endothelium-dependent coronary vasodilation was reduced in the OVX group and potentiated in the OVX P4 group. Following the inhibition of the nitric oxide (NO) pathway, the bradykinin-induced relaxing response was potentiated in the OVX P4 group. With regard to the combined inhibition of NO and prostanoids pathways, the OVX P4 group showed a greater relaxing response, similar to what was found upon individual inhibition of NO. After the combined inhibition of NO, prostanoids and epoxyeicosatrienoic acids' pathways, the vasodilatory response induced by BK was abolished in all groups. SIGNIFICANCE Treatment with P4 prevented oxidative stress induced by ovariectomy. These results suggest that progesterone has a beneficial action on the coronary vascular bed.
Collapse
Affiliation(s)
- Tagana Rosa da Cunha
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Wender Nascimento Rouver
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Damasceno Costa
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcella Daruge Grando
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Virgínia Soares Lemos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lusiane Maria Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| |
Collapse
|
47
|
Triggle CR, Ding H, Marei I, Anderson TJ, Hollenberg MD. Why the endothelium? The endothelium as a target to reduce diabetes-associated vascular disease. Can J Physiol Pharmacol 2020; 98:415-430. [PMID: 32150686 DOI: 10.1139/cjpp-2019-0677] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over the past 66 years, our knowledge of the role of the endothelium in the regulation of cardiovascular function and dysfunction has advanced from the assumption that it is a single layer of cells that serves as a barrier between the blood stream and vascular smooth muscle to an understanding of its role as an essential endocrine-like organ. In terms of historical contributions, we pay particular credit to (1) the Canadian scientist Dr. Rudolf Altschul who, based on pathological changes in the appearance of the endothelium, advanced the argument in 1954 that "one is only as old as one's endothelium" and (2) the American scientist Dr. Robert Furchgott, a 1998 Nobel Prize winner in Physiology or Medicine, who identified the importance of the endothelium in the regulation of blood flow. This review provides a brief history of how our knowledge of endothelial function has advanced and now recognize that the endothelium produces a plethora of signaling molecules possessing paracrine, autocrine, and, arguably, systemic hormone functions. In addition, the endothelium is a therapeutic target for the anti-diabetic drugs metformin, glucagon-like peptide I (GLP-1) receptor agonists, and inhibitors of the sodium-glucose cotransporter 2 (SGLT2) that offset the vascular disease associated with diabetes.
Collapse
Affiliation(s)
- Chris R Triggle
- Departments of Pharmacology and Medical Education, Weill Cornell Medical College, Doha, Qatar
| | - Hong Ding
- Departments of Pharmacology and Medical Education, Weill Cornell Medical College, Doha, Qatar
| | - Isra Marei
- Departments of Pharmacology and Medical Education, Weill Cornell Medical College, Doha, Qatar
| | - Todd J Anderson
- Department of Cardiac Sciences and Libin Cardiovascular Institute, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| | - Morley D Hollenberg
- Inflammation Research Network, Snyder Institute for Chronic Disease, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada.,Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada.,Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
48
|
Obradovic M, Essack M, Zafirovic S, Sudar‐Milovanovic E, Bajic VP, Van Neste C, Trpkovic A, Stanimirovic J, Bajic VB, Isenovic ER. Redox control of vascular biology. Biofactors 2020; 46:246-262. [PMID: 31483915 PMCID: PMC7187163 DOI: 10.1002/biof.1559] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
Redox control is lost when the antioxidant defense system cannot remove abnormally high concentrations of signaling molecules, such as reactive oxygen species (ROS). Chronically elevated levels of ROS cause oxidative stress that may eventually lead to cancer and cardiovascular and neurodegenerative diseases. In this review, we focus on redox effects in the vascular system. We pay close attention to the subcompartments of the vascular system (endothelium, smooth muscle cell layer) and give an overview of how redox changes influence those different compartments. We also review the core aspects of redox biology, cardiovascular physiology, and pathophysiology. Moreover, the topic-specific knowledgebase DES-RedoxVasc was used to develop two case studies, one focused on endothelial cells and the other on the vascular smooth muscle cells, as a starting point to possibly extend our knowledge of redox control in vascular biology.
Collapse
Affiliation(s)
- Milan Obradovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Magbubah Essack
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Sonja Zafirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Emina Sudar‐Milovanovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladan P. Bajic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Christophe Van Neste
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Julijana Stanimirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| |
Collapse
|
49
|
Shimokawa H. Reactive oxygen species in cardiovascular health and disease: special references to nitric oxide, hydrogen peroxide, and Rho-kinase. J Clin Biochem Nutr 2020; 66:83-91. [PMID: 32231403 DOI: 10.3164/jcbn.19-119] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 01/09/2023] Open
Abstract
The interaction between endothelial cells and vascular smooth muscle cells (VSMC) plays an important role in regulating cardiovascular homeostasis. Endothelial cells synthesize and release endothelium-derived relaxing factors (EDRFs), including vasodilator prostaglandins, nitric oxide (NO), and endothelium-dependent hyperpolarization (EDH) factors. Importantly, the contribution of EDRFs to endothelium-dependent vasodilatation markedly varies in a vessel size-dependent manner; NO mainly mediates vasodilatation of relatively large vessels, while EDH factors in small resistance vessels. We have previously identified that endothelium-derived hydrogen peroxide (H2O2) is an EDH factor especially in microcirculation. Several lines of evidence indicate the importance of the physiological balance between NO and H2O2/EDH factor. Rho-kinase was identified as the effectors of the small GTP-binding protein, RhoA. Both endothelial NO production and NO-mediated signaling in VSMC are targets and effectors of the RhoA/Rho-kinase pathway. In endothelial cells, the RhoA/Rho-kinase pathway negatively regulates NO production. On the contrary, the pathway enhances VSMC contraction with resultant occurrence of coronary artery spasm and promotes the development of oxidative stress and vascular remodeling. In this review, I will briefly summarize the current knowledge on the regulatory roles of endothelium-derived relaxing factors, with special references to NO and H2O2/EDH factor, in relation to Rho-kinase, in cardiovascular health and disease.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
50
|
Shimokawa H, Godo S. Nitric oxide and endothelium-dependent hyperpolarization mediated by hydrogen peroxide in health and disease. Basic Clin Pharmacol Toxicol 2020; 127:92-101. [PMID: 31846200 DOI: 10.1111/bcpt.13377] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/12/2019] [Indexed: 01/09/2023]
Abstract
The endothelium plays crucial roles in modulating vascular tone by synthesizing and releasing endothelium-derived relaxing factors (EDRFs), including vasodilator prostaglandins, nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH) factors. Thus, endothelial dysfunction is the hallmark of atherosclerotic cardiovascular diseases. Importantly, the contribution of EDRFs to endothelium-dependent vasodilatation varies in a distinct vessel size-dependent manner; NO mainly mediates vasodilatation of relatively large, conduit vessels (eg epicardial coronary arteries), while EDH factors in small resistance vessels (eg coronary microvessels). Endothelium-derived hydrogen peroxide (H2 O2 ) is a physiological signalling molecule serving as one of the major EDH factors especially in microcirculations and has gained increasing attention in view of its emerging relevance for cardiovascular diseases. In the clinical settings, therapeutic approaches targeting NO (eg NO donors) or non-specific elimination of reactive oxygen species (eg antioxidant supplements) are disappointingly ineffective for the treatment of various cardiovascular diseases, in which endothelial dysfunction and coronary microvascular dysfunction are substantially involved. These lines of evidence indicate the potential importance of the physiological balance between NO and H2 O2 /EDH factor. Further characterization and better understanding of endothelium-dependent vasodilatations are important to develop novel therapeutic strategies in cardiovascular medicine. In this MiniReview, we will briefly summarize the current knowledge on the emerging regulatory roles of endothelium-dependent vasodilatations in the cardiovascular system, with a special reference to the two major EDRFs, NO and H2 O2 /EDH factor, in health and disease.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|