1
|
Wang Z, Gao H, Ma X, Zhu D, Zhao L, Xiao W. Adrenic acid: A promising biomarker and therapeutic target (Review). Int J Mol Med 2025; 55:20. [PMID: 39575474 PMCID: PMC11611323 DOI: 10.3892/ijmm.2024.5461] [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: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 01/05/2025] Open
Abstract
Adrenic acid is a 22‑carbon unsaturated fatty acid that is widely present in the adrenal gland, liver, brain, kidney and vascular system that plays a regulatory role in various pathophysiological processes, such as inflammatory reactions, lipid metabolism, oxidative stress, vascular function, and cell death. Adrenic acid is a potential biomarker for various ailments, including metabolic, neurodegenerative and cardiovascular diseases and cancer. In addition, adrenic acid is influenced by the pharmacological properties of several natural products, such as astragaloside IV, evodiamine, quercetin, kaempferol, Berberine‑baicalin and prebiotics, so it is a promising new target for clinical treatment and drug development. However, the molecular mechanisms by which adrenic acid exerts are unclear. The present study systematically reviewed the biosynthesis and metabolism of adrenic acid, focusing on intrinsic mechanisms that influence the progression of metabolic, cardiovascular and neurological disease. These mechanisms regulate several key processes, including immuno‑inflammatory response, oxidative stress, vascular function and cell death. In addition, the present study explored the potential clinical translational value of adrenic acid as a biomarker and therapeutic target. To the best of our knowledge, the present study is first systematic summary of the mechanisms of action of adrenic acid across a range of diseases. The present study provides understanding of the wide range of metabolic activities of adrenic acid and a basis for further exploring the pathogenesis and therapeutic targets of various diseases.
Collapse
Affiliation(s)
- Ze Wang
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Haoyang Gao
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Xiaotong Ma
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Danlin Zhu
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Linlin Zhao
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
- School of Physical Education, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Weihua Xiao
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| |
Collapse
|
2
|
Suarez A, Fernandez L, Riera J. Characterizing astrocyte-mediated neurovascular coupling by combining optogenetics and biophysical modeling. J Cereb Blood Flow Metab 2025:271678X241311010. [PMID: 39791314 PMCID: PMC11719438 DOI: 10.1177/0271678x241311010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
Vasoactive signaling from astrocytes is an important contributor to the neurovascular coupling (NVC), which aims at providing energy to neurons during brain activation by increasing blood perfusion in the surrounding vasculature. Pharmacological manipulations have been previously combined with experimental techniques (e.g., transgenic mice, uncaging, and multiphoton microscopy) and stimulation paradigms to isolate in vivo individual pathways of the astrocyte-mediated NVC. Unfortunately, these pathways are highly nonlinear and non-additive. To separate these pathways in a unified framework, we combine a comprehensive biophysical model of vasoactive signaling from astrocytes with a unique optogenetic stimulation method that selectively induces astrocytic Ca2+ signaling in a large population of astrocytes. We also use a sensitivity analysis and an optimization technique to estimate key model parameters. Optogenetically-induced Ca2+ signals in astrocytes cause a cerebral blood flow (CBF) response with two major components. Component-1 was rapid and smaller (ΔCBF∼13%, 18 seconds), while component-2 was slowest and highest (ΔCBF ∼18%, 45 seconds). The proposed biophysical model was adequate in reproducing component-2, which was validated with a pharmacological manipulation. Model's predictions were not in contradiction with previous studies. Finally, we discussed scenarios accounting for the existence of component-1, which once validated might be included in our model.
Collapse
Affiliation(s)
- Alejandro Suarez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Lazaro Fernandez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Jorge Riera
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| |
Collapse
|
3
|
Parchem K, Letsiou S, Petan T, Oskolkova O, Medina I, Kuda O, O'Donnell VB, Nicolaou A, Fedorova M, Bochkov V, Gladine C. Oxylipin profiling for clinical research: Current status and future perspectives. Prog Lipid Res 2024; 95:101276. [PMID: 38697517 DOI: 10.1016/j.plipres.2024.101276] [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: 12/12/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Oxylipins are potent lipid mediators with increasing interest in clinical research. They are usually measured in systemic circulation and can provide a wealth of information regarding key biological processes such as inflammation, vascular tone, or blood coagulation. Although procedures still require harmonization to generate comparable oxylipin datasets, performing comprehensive profiling of circulating oxylipins in large studies is feasible and no longer restricted by technical barriers. However, it is essential to improve and facilitate the biological interpretation of complex oxylipin profiles to truly leverage their potential in clinical research. This requires regular updating of our knowledge about the metabolism and the mode of action of oxylipins, and consideration of all factors that may influence circulating oxylipin profiles independently of the studied disease or condition. This review aims to provide the readers with updated and necessary information regarding oxylipin metabolism, their different forms in systemic circulation, the current limitations in deducing oxylipin cellular effects from in vitro bioactivity studies, the biological and technical confounding factors needed to consider for a proper interpretation of oxylipin profiles.
Collapse
Affiliation(s)
- Karol Parchem
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Gabriela Narutowicza St., 80-233 Gdańsk, Poland; Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 53210 Pardubice, Czech Republic.
| | - Sophia Letsiou
- Department of Biomedical Sciences, University of West Attica, Ag. Spiridonos St. Egaleo, 12243 Athens, Greece.
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia.
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Isabel Medina
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain.
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| | - Anna Nicolaou
- School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, 01307 Dresden, Germany.
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France.
| |
Collapse
|
4
|
Hansen CE, Kamermans A, Mol K, Berve K, Rodriguez-Mogeda C, Fung WK, van Het Hof B, Fontijn RD, van der Pol SMA, Michalick L, Kuebler WM, Kenkhuis B, van Roon-Mom W, Liedtke W, Engelhardt B, Kooij G, Witte ME, de Vries HE. Inflammation-induced TRPV4 channels exacerbate blood-brain barrier dysfunction in multiple sclerosis. J Neuroinflammation 2024; 21:72. [PMID: 38521959 PMCID: PMC10960997 DOI: 10.1186/s12974-024-03069-9] [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: 12/21/2023] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) dysfunction and immune cell migration into the central nervous system (CNS) are pathogenic drivers of multiple sclerosis (MS). Ways to reinstate BBB function and subsequently limit neuroinflammation present promising strategies to restrict disease progression. However, to date, the molecular players directing BBB impairment in MS remain poorly understood. One suggested candidate to impact BBB function is the transient receptor potential vanilloid-type 4 ion channel (TRPV4), but its specific role in MS pathogenesis remains unclear. Here, we investigated the role of TRPV4 in BBB dysfunction in MS. MAIN TEXT In human post-mortem MS brain tissue, we observed a region-specific increase in endothelial TRPV4 expression around mixed active/inactive lesions, which coincided with perivascular microglia enrichment in the same area. Using in vitro models, we identified that microglia-derived tumor necrosis factor-α (TNFα) induced brain endothelial TRPV4 expression. Also, we found that TRPV4 levels influenced brain endothelial barrier formation via expression of the brain endothelial tight junction molecule claudin-5. In contrast, during an inflammatory insult, TRPV4 promoted a pathological endothelial molecular signature, as evidenced by enhanced expression of inflammatory mediators and cell adhesion molecules. Moreover, TRPV4 activity mediated T cell extravasation across the brain endothelium. CONCLUSION Collectively, our findings suggest a novel role for endothelial TRPV4 in MS, in which enhanced expression contributes to MS pathogenesis by driving BBB dysfunction and immune cell migration.
Collapse
Grants
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 91719305 Dutch Research Council, NWO, Vidi grant
- 91719305 Dutch Research Council, NWO, Vidi grant
- 91719305 Dutch Research Council, NWO, Vidi grant
- 18-1023MS Stichting MS Research
- 20-1106MS Stichting MS Research
- 20-1106MS Stichting MS Research
- 18-1023MS Stichting MS Research
- 20-1106MS Stichting MS Research
- 81X3100216 Deutsches Zentrum für Herz-Kreislaufforschung
- SFB-TR84 : subprojects A02 & C09, SFB-1449 subproject B01, SFB 1470 subproject A04, KU1218/9-1, KU1218/11-1, and KU1218/12-1 Deutsche Forschungsgemeinschaft
- PROVID (01KI20160A) and SYMPATH (01ZX1906A) Bundesministerium für Bildung und Forschung
- HA2016-02-02 Hersenstichting
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Kevin Mol
- Department of Biomedical Engineering and Physics, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Kristina Berve
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Carla Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Wing Ka Fung
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Ruud D Fontijn
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Laura Michalick
- Institute of Physiology, Corporate member of the Freie Universität Berlin and Humboldt Universität to Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Corporate member of the Freie Universität Berlin and Humboldt Universität to Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| | - Boyd Kenkhuis
- Department of Human Genetics, Leiden University Medical Center Leiden, Leiden, The Netherlands
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Willeke van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center Leiden, Leiden, The Netherlands
| | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, NY, USA
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, USA
| | | | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maarten E Witte
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Asulin M, Gorodetzer N, Fridman R, Shelly Ben-Shushan R, Cohen Z, Beyer AM, Chuyun D, Gutterman DD, Szuchman-Sapir A. 5,6-diHETE lactone (EPA-L) mediates hypertensive microvascular dilation by activating the endothelial GPR-PLC-IP 3 signaling pathway. Biochem Biophys Res Commun 2024; 700:149585. [PMID: 38290177 DOI: 10.1016/j.bbrc.2024.149585] [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: 12/20/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Endothelial microvascular dysfunction affects multi-organ pathologic processes that contribute to increased vascular tone and is at the base of impaired metabolic and cardiovascular diseases. The vascular dilation impaired by nitric oxide (NO) deficiency in such dysfunctional endothelium is often balanced by endothelial-derived hyperpolarizing factors (EDHFs), which play a critical role in managing vascular tone. Our latest research has uncovered a new group of lactone oxylipins produced in the polyunsaturated fatty acids (PUFAs) CYP450 epoxygenase pathway, significantly affecting vascular dilation. The lactone oxylipin, derived from arachidonic acid (5,6-diHET lactone, AA-L), has been previously shown to facilitate vasodilation dependent on the endothelium in isolated human microvessels. The administration of the lactone oxylipin derived from eicosapentaenoic acid (5,6-diHETE lactone, EPA-L) to hypertensive rats demonstrated a significant decrease in blood pressure and improvement in the relaxation of microvessels. However, the molecular signaling processes that underlie these observations were not fully understood. The current study delineates the molecular pathways through which EPA-L promotes endothelium-dependent vascular dilation. In microvessels from hypertensive individuals, it was found that EPA-L mediates endothelium-dependent vasodilation while the signaling pathway was not dependent on NO. In vitro studies on human endothelial cells showed that the hyperpolarization mediated by EPA-L relies on G-protein-coupled receptor (GPR)-phospholipase C (PLC)-IP3 signaling that further activates calcium-dependent potassium flux. The pathway was confirmed using a range of inhibitors and cells overexpressing GPR40, where a specific antagonist reduced the calcium levels and outward currents induced by EPA-L. The downstream AKT and endothelial NO synthase (eNOS) phosphorylations were non-significant. These findings show that the GPR-PLC-IP3 pathway is a key mediator in the EPA-L-triggered vasodilation of arterioles. Therefore, EPA-L is identified as a significant lactone-based PUFA metabolite that contributes to endothelial and vascular health.
Collapse
Affiliation(s)
- Meitar Asulin
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Nadav Gorodetzer
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Rotem Fridman
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
| | | | - Zohar Cohen
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Andreas M Beyer
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - David D Gutterman
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andrea Szuchman-Sapir
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
6
|
Ertuglu LA, Mutchler AP, Jamison S, Laffer CL, Saleem M, Blackwell DJ, Kryshtal DO, Sahinoz M, Sheng Q, Wanjalla CN, Pakala S, Justin Y, Gutierrez OM, Kleyman TR, Knollmann BC, Ikizler TA, Kirabo A. Eicosanoid-Regulated Myeloid ENaC and Isolevuglandin Formation in Human Salt-Sensitive Hypertension. Hypertension 2024; 81:516-529. [PMID: 37675576 PMCID: PMC10918035 DOI: 10.1161/hypertensionaha.123.21285] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The mechanisms by which salt increases blood pressure in people with salt sensitivity remain unclear. Our previous studies found that high sodium enters antigen-presenting cells (APCs) via the epithelial sodium channel and leads to the production of isolevuglandins and hypertension. In the current mechanistic clinical study, we hypothesized that epithelial sodium channel-dependent isolevuglandin-adduct formation in APCs is regulated by epoxyeicosatrienoic acids (EETs) and leads to salt-sensitive hypertension in humans. METHODS Salt sensitivity was assessed in 19 hypertensive subjects using an inpatient salt loading and depletion protocol. Isolevuglandin-adduct accumulation in APCs was analyzed using flow cytometry. Gene expression in APCs was analyzed using cellular indexing of transcriptomes and epitopes by sequencing analysis of blood mononuclear cells. Plasma and urine EETs were measured using liquid chromatography-mass spectrometry. RESULTS Baseline isolevuglandin+ APCs correlated with higher salt-sensitivity index. Isolevuglandin+ APCs significantly decreased from salt loading to depletion with an increasing salt-sensitivity index. We observed that human APCs express the epithelial sodium channel δ subunit, SGK1 (salt-sensing kinase serum/glucocorticoid kinase 1), and cytochrome P450 2S1. We found a direct correlation between baseline urinary 14,15 EET and salt-sensitivity index, whereas changes in urinary 14,15 EET negatively correlated with isolevuglandin+ monocytes from salt loading to depletion. Coincubation with 14,15 EET inhibited high-salt-induced increase in isolevuglandin+ APC. CONCLUSIONS Isolevuglandin formation in APCs responds to acute changes in salt intake in salt-sensitive but not salt-resistant people with hypertension, and this may be regulated by renal 14,15 EET. Baseline levels of isolevuglandin+ APCs or urinary 14,15 EET may provide diagnostic tools for salt sensitivity without a protocol of salt loading.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley Pitzer Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - S Jamison
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Cheryl L. Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Daniel J. Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Dmytro O. Kryshtal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Melis Sahinoz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Suman Pakala
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Yu Justin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, Pharmacology, and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Björn C. Knollmann
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - T. Alp Ikizler
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Vanderbilt Center for Immunobiology (VCI)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4)
- Vanderbilt Institute for Global Health (VIGH)
| |
Collapse
|
7
|
Alarcon-Martinez L, Shiga Y, Villafranca-Baughman D, Cueva Vargas JL, Vidal Paredes IA, Quintero H, Fortune B, Danesh-Meyer H, Di Polo A. Neurovascular dysfunction in glaucoma. Prog Retin Eye Res 2023; 97:101217. [PMID: 37778617 DOI: 10.1016/j.preteyeres.2023.101217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal provision of oxygen and nutrients to sustain their activity. The timely regulation of blood flow is, therefore, essential to supply firing neurons in active areas with the oxygen and glucose they need for energy. Many glaucoma patients suffer from vascular deficits including reduced blood flow, impaired autoregulation, neurovascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the ability of its members to regulate neurovascular interactions and how their function might be altered during glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes, which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysiological implications, while highlighting opportunities to develop strategies for vascular protection and regeneration to improve functional outcomes in glaucoma.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada; Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Isaac A Vidal Paredes
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Heberto Quintero
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Helen Danesh-Meyer
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada.
| |
Collapse
|
8
|
Zhou Z, Zhang M, Zhao C, Gao X, Wen Z, Wu J, Chen C, Fleming I, Hu J, Wang DW. Epoxyeicosatrienoic Acids Prevent Cardiac Dysfunction in Viral Myocarditis via Interferon Type I Signaling. Circ Res 2023; 133:772-788. [PMID: 37681352 DOI: 10.1161/circresaha.123.322619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Myocarditis is a challenging inflammatory disease of the heart, and better understanding of its pathogenesis is needed to develop specific drug therapies. Epoxyeicosatrienoic acids (EETs), active molecules synthesized by CYP (cytochrome P450) enzymes from arachidonic acids and hydrolyzed to less active dihydroxyeicosatrienoic acids by sEH (soluble epoxide hydrolase), have been attributed anti-inflammatory activity. Here, we investigated whether EETs have immunomodulatory activity and exert protective effects on coxsackie B3 virus-induced myocarditis. Viral infection altered eicosanoid epoxide and diol levels in both patients with myocarditis and in the murine heart and correlated with the increased expression and activity of sEH after coxsackie B3 virus infection. Administration of a sEH inhibitor prevented coxsackie B3 virus-induced cardiac dysfunction and inflammatory infiltration. Importantly, EET/sEH inhibitor treatment attenuated viral infection or improved viral resistance by activating type I IFN (interferon) signaling. At the molecular level, EETs enhanced the interaction between GSK3β (glycogen synthase kinase-3 beta) and TBK1 (TANK-binding kinase 1) to promote IFN-β production. Our findings revealed that EETs and sEH inhibitors prevent the progress of coxsackie B3 virus-induced myocarditis, particularly by promoting viral resistance by increasing IFN production.
Collapse
Affiliation(s)
- Zhou Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Min Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Chengcheng Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Xu Gao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Junfang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| | - Ingrid Fleming
- Sino-German Laboratory of CardioPulmonary Science (I.F., J.H., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany (I.F., J.H.)
- German Center of Cardiovascular Research, Partner Site RheinMain, Frankfurt am Main, Germany (I.F., J.H.)
| | - Jiong Hu
- Department of Histology and Embryology, School of Basic Medicine (J.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Sino-German Laboratory of CardioPulmonary Science (I.F., J.H., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany (I.F., J.H.)
- German Center of Cardiovascular Research, Partner Site RheinMain, Frankfurt am Main, Germany (I.F., J.H.)
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Sino-German Laboratory of CardioPulmonary Science (I.F., J.H., D.W.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (Z.Z., M.Z., C.Z., X.G., Z.W., J.W., C.C., D.W.W.)
| |
Collapse
|
9
|
Zhou Y, Rothe M, Schunck WH, Ruess L, Menzel R. Serotonin-induced stereospecific formation and bioactivity of the eicosanoid 17,18-epoxyeicosatetraenoic acid in the regulation of pharyngeal pumping of C. elegans. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159304. [PMID: 36914111 DOI: 10.1016/j.bbalip.2023.159304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/15/2023]
Abstract
17,18-Epoxyeicosatetraenoic acid (17,18-EEQ), the most abundant eicosanoid generated by cytochrome P450 (CYP) enzymes in C. elegans, is a potential signaling molecule in the regulation of pharyngeal pumping activity of this nematode. As a chiral molecule, 17,18-EEQ can exist in two stereoisomers, the 17(R),18(S)- and 17(S),18(R)-EEQ enantiomers. Here we tested the hypothesis that 17,18-EEQ may function as a second messenger of the feeding-promoting neurotransmitter serotonin and stimulates pharyngeal pumping and food uptake in a stereospecific manner. Serotonin treatment of wildtype worms induced a more than twofold increase of free 17,18-EEQ levels. As revealed by chiral lipidomics analysis, this increase was almost exclusively due to an enhanced release of the (R,S)-enantiomer of 17,18-EEQ. In contrast to the wildtype strain, serotonin failed to induce 17,18-EEQ formation as well as to accelerate pharyngeal pumping in mutant strains defective in the serotonin SER-7 receptor. However, the pharyngeal activity of the ser-7 mutant remained fully responsive to exogenous 17,18-EEQ administration. Short term incubations of well-fed and starved wildtype nematodes showed that both racemic 17,18-EEQ and 17(R),18(S)-EEQ were able to increase pharyngeal pumping frequency and the uptake of fluorescence-labeled microspheres, while 17(S),18(R)-EEQ and also 17,18-dihydroxyeicosatetraenoic acid (17,18-DHEQ, the hydrolysis product of 17,18-EEQ) were ineffective. Taken together, these results show that serotonin induces 17,18-EEQ formation in C. elegans via the SER-7 receptor and that both the formation of this epoxyeicosanoid and its subsequent stimulatory effect on pharyngeal activity proceed with high stereospecificity confined to the (R,S)-enantiomer.
Collapse
Affiliation(s)
- Yiwen Zhou
- Humboldt-Universität zu Berlin, Institue of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany
| | - Michael Rothe
- Lipidomix GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Wolf-Hagen Schunck
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Liliane Ruess
- Humboldt-Universität zu Berlin, Institue of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany
| | - Ralph Menzel
- Humboldt-Universität zu Berlin, Institue of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany.
| |
Collapse
|
10
|
Favor OK, Chauhan PS, Pourmand E, Edwards AM, Wagner JG, Lewandowski RP, Heine LK, Harkema JR, Lee KSS, Pestka JJ. Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice. Front Immunol 2023; 14:1124910. [PMID: 36875087 PMCID: PMC9978350 DOI: 10.3389/fimmu.2023.1124910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/17/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Lipopolysaccharide (LPS)-accelerated autoimmune glomerulonephritis (GN) in NZBWF1 mice is a preclinical model potentially applicable for investigating lipidome-modulating interventions against lupus. LPS can be expressed as one of two chemotypes: smooth LPS (S-LPS) or rough LPS (R-LPS) which is devoid of O-antigen polysaccharide sidechain. Since these chemotypes differentially affect toll-like receptor 4 (TLR4)-mediated immune cell responses, these differences may influence GN induction. Methods We initially compared the effects of subchronic intraperitoneal (i.p.) injection for 5 wk with 1) Salmonella S-LPS, 2) Salmonella R-LPS, or 3) saline vehicle (VEH) (Study 1) in female NZBWF1 mice. Based on the efficacy of R-LPS in inducing GN, we next used it to compare the impact of two lipidome-modulating interventions, ω-3 polyunsaturated fatty acid (PUFA) supplementation and soluble epoxide hydrolase (sEH) inhibition, on GN (Study 2). Specifically, effects of consuming ω-3 docosahexaenoic acid (DHA) (10 g/kg diet) and/or the sEH inhibitor 1-(4-trifluoro-methoxy-phenyl)-3-(1-propionylpiperidin-4-yl) urea (TPPU) (22.5 mg/kg diet ≈ 3 mg/kg/day) on R-LPS triggering were compared. Results In Study 1, R-LPS induced robust elevations in blood urea nitrogen, proteinuria, and hematuria that were not evident in VEH- or S-LPS-treated mice. R-LPS-treated mice further exhibited kidney histopathology including robust hypertrophy, hyperplasia, thickened membranes, lymphocytic accumulation containing B and T cells, and glomerular IgG deposition consistent with GN that was not evident in VEH- or SLPS-treated groups. R-LPS but not S-LPS induced spleen enlargement with lymphoid hyperplasia and inflammatory cell recruitment in the liver. In Study 2, resultant blood fatty acid profiles and epoxy fatty acid concentrations reflected the anticipated DHA- and TPPU-mediated lipidome changes, respectively. The relative rank order of R-LPS-induced GN severity among groups fed experimental diets based on proteinuria, hematuria, histopathologic scoring, and glomerular IgG deposition was: VEH/CON< R-LPS/DHA ≈ R-LPS/TPPU<<< R-LPS/TPPU+DHA ≈ R-LPS/CON. In contrast, these interventions had modest-to- negligible effects on R-LPS-induced splenomegaly, plasma antibody responses, liver inflammation, and inflammation-associated kidney gene expression. Discussion We show for the first time that absence of O-antigenic polysaccharide in R-LPS is critical to accelerated GN in lupus-prone mice. Furthermore, intervention by lipidome modulation through DHA feeding or sEH inhibition suppressed R-LPS-induced GN; however, these ameliorative effects were greatly diminished upon combining the treatments.
Collapse
Affiliation(s)
- Olivia K. Favor
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Preeti S. Chauhan
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Elham Pourmand
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Angel M. Edwards
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - James G. Wagner
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Ryan P. Lewandowski
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Lauren K. Heine
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jack R. Harkema
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Kin Sing Stephen Lee
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
11
|
Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. Int J Mol Sci 2023; 24:ijms24021637. [PMID: 36675152 PMCID: PMC9863938 DOI: 10.3390/ijms24021637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
It is commonly believed that the inactivation of inflammation is mainly due to the decay or cessation of inducers. In reality, in connection with the development of atherosclerosis, spontaneous decay of inducers is not observed. It is now known that lipid mediators originating from polyunsaturated fatty acids (PUFAs), which are important constituents of all cell membranes, can act in the inflamed tissue and bring it to resolution. In fact, PUFAs, such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are precursors to both pro-inflammatory and anti-inflammatory compounds. In this review, we describe the lipid mediators of vascular inflammation and resolution, and their biochemical activity. In addition, we highlight data from the literature that often show a worsening of atherosclerotic disease in subjects deficient in lipid mediators of inflammation resolution, and we also report on the anti-proteasic and anti-thrombotic properties of these same lipid mediators. It should be noted that despite promising data observed in both animal and in vitro studies, contradictory clinical results have been observed for omega-3 PUFAs. Many further studies will be required in order to clarify the observed conflicts, although lifestyle habits such as smoking or other biochemical factors may often influence the normal synthesis of lipid mediators of inflammation resolution.
Collapse
|
12
|
Cho C, Aliwarga T, Wiley AM, Totah RA. Cardioprotective mechanisms of cytochrome P450 derived oxylipins from ω-3 and ω-6 PUFAs. ADVANCES IN PHARMACOLOGY 2023; 97:201-227. [DOI: 10.1016/bs.apha.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
13
|
ElKhatib MAW, Isse FA, El-Kadi AOS. Effect of inflammation on cytochrome P450-mediated arachidonic acid metabolism and the consequences on cardiac hypertrophy. Drug Metab Rev 2022; 55:50-74. [PMID: 36573379 DOI: 10.1080/03602532.2022.2162075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The incidence of heart failure (HF) is generally preceded by cardiac hypertrophy (CH), which is the enlargement of cardiac myocytes in response to stress. During CH, the metabolism of arachidonic acid (AA), which is present in the cell membrane phospholipids, is modulated. Metabolism of AA gives rise to hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs) via cytochrome P450 (CYP) ω-hydroxylases and CYP epoxygenases, respectively. A plethora of studies demonstrated the involvement of CYP-mediated AA metabolites in the pathogenesis of CH. Also, inflammation is known to be a characteristic hallmark of CH. In this review, our aim is to highlight the impact of inflammation on CYP-derived AA metabolites and CH. Inflammation is shown to modulate the expression of various CYP ω-hydroxylases and CYP epoxygenases and their respective metabolites in the heart. In general, HETEs such as 20-HETE and mid-chain HETEs are pro-inflammatory, while EETs are characterized by their anti-inflammatory and cardioprotective properties. Several mechanisms are implicated in inflammation-induced CH, including the modulation of NF-κB and MAPK. This review demonstrated the inflammatory modulation of cardiac CYPs and their metabolites in the context of CH and the anti-inflammatory strategies that can be employed in the treatment of CH and HF.
Collapse
Affiliation(s)
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | | |
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
|
Nippert AR, Chiang PP, Del Franco AP, Newman EA. Astrocyte regulation of cerebral blood flow during hypoglycemia. J Cereb Blood Flow Metab 2022; 42:1534-1546. [PMID: 35296178 PMCID: PMC9274859 DOI: 10.1177/0271678x221089091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 12/13/2022]
Abstract
Hypoglycemia triggers increases in cerebral blood flow (CBF), augmenting glucose supply to the brain. We have tested whether astrocytes, which can regulate vessel tone, contribute to this CBF increase. We hypothesized that hypoglycemia-induced adenosine signaling acts to increase astrocyte Ca2+ activity, which then causes the release of prostaglandins (PGs) and epoxyeicosatrienoic acids (EETs), leading to the dilation of brain arterioles and blood flow increases. We used an awake mouse model to investigate the effects of insulin-induced hypoglycemia on arterioles and astrocytes in the somatosensory cortex. During insulin-induced hypoglycemia, penetrating arterioles dilated and astrocyte Ca2+ signaling increased when blood glucose dropped below a threshold of ∼50 mg/dL. Application of the A2A adenosine receptor antagonist ZM-241385 eliminated hypoglycemia-evoked astrocyte Ca2+ increases and reduced arteriole dilations by 44% (p < 0.05). SC-560 and miconazole, which block the production of the astrocyte vasodilators PGs and EETs respectively, reduced arteriole dilations in response to hypoglycemia by 89% (p < 0.001) and 76% (p < 0.001). Hypoglycemia-induced arteriole dilations were decreased by 65% (p < 0.001) in IP3R2 knockout mice, which have reduced astrocyte Ca2+ signaling compared to wild-type. These results support the hypothesis that astrocytes contribute to hypoglycemia-induced increases in CBF by releasing vasodilators in a Ca2+-dependent manner.
Collapse
Affiliation(s)
- Amy R Nippert
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | | | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
16
|
Paraoxonase 1 hydrolysis of EPA-derived lactone impairs endothelial-mediated vasodilation. Prostaglandins Other Lipid Mediat 2022; 162:106665. [PMID: 35817276 DOI: 10.1016/j.prostaglandins.2022.106665] [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: 05/11/2022] [Revised: 06/26/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022]
Abstract
Human serum paraoxonase-1 (PON1) is a lactonase that plays a significant role in anti-atherosclerotic high-density lipoprotein (HDL) activity. PON1 is also localized in endothelial cell membranes, where it is enzymatically active and regulates endothelial signals. PON1 has a high specificity for lipophilic lactones and has been shown to hydrolyze and regulate lactone lipid mediators derived from arachidonic polyunsaturated fatty acids (PUFA). Previously, we showed that an arachidonic acid lactone metabolite (AA-L) dose-dependently dilates PON1 gene deletion (PON1KO) mouse mesenteric arteries significantly more than wild-type arteries. In contrast, preincubation with HDL or rePON1 reduced AA-L-dependent vasodilation. Recently we showed that an additional δ-lactone metabolite derived from the eicosapentaenoic acid lactone, 5,6-δ-DiHETE lactone (EPA-L) reduced blood pressure by dilating microvessels of hypertensive rats. However, whether PON1 regulates the activity of the EPA-L lipid mediator is unknown. AIM To demonstrate that PON1 hydrolyzes EPA-L and to reveal the effect of this hydrolysis on endothelial-dependent vascular dilation. METHODS AND RESULTS In vascular reactivity experiments, EPA-L dose-dependently dilated PON1KO mouse mesenteric arteries significantly more than wild-type mesenteric arteries. This dilation was not affected by nitric oxide inhibition. PON1 impaired the cellular calcium increase mediated by EPA-L in endothelial cells, though this impairment decreased with PON1 internalization to the cell. CONCLUSION These findings support that PUFA-lactones are physiological substrates of PON1, and that PON1 activity in the endothelial membrane affects the dilation of microvessels that is induced by these endothelial-derived hyperpolarizing PUFA-lactones.
Collapse
|
17
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
18
|
O'Brien F, Staunton CA, Barrett-Jolley R. Systemic application of the transient receptor potential vanilloid-type 4 antagonist GSK2193874 induces tail vasodilation in a mouse model of thermoregulation. Biol Lett 2022; 18:20220129. [PMID: 35702981 PMCID: PMC9198786 DOI: 10.1098/rsbl.2022.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/25/2022] [Indexed: 11/12/2022] Open
Abstract
In humans, skin is a primary thermoregulatory organ, with vasodilation leading to rapid body cooling, whereas in Rodentia the tail performs an analogous function. Many thermodetection mechanisms are likely to be involved including transient receptor potential vanilloid-type 4 (TRPV4), an ion channel with thermosensitive properties. Previous studies have shown that TRPV4 is a vasodilator by local action in blood vessels, so here, we investigated whether constitutive TRPV4 activity affects Mus muscularis tail vascular tone and thermoregulation. We measured tail blood flow by pressure plethysmography in lightly sedated M. muscularis (CD1 strain) at a range of ambient temperatures, with and without intraperitoneal administration of the blood-brain barrier crossing TRPV4 antagonist GSK2193874. We also measured heart rate (HR) and blood pressure. As expected for a thermoregulatory organ, we found that tail blood flow increased with temperature. However, unexpectedly, we found that GSK2193874 increased tail blood flow at all temperatures, and we observed changes in HR variability. Since local TRPV4 activation causes vasodilation that would increase tail blood flow, these data suggest that increases in tail blood flow resulting from the TRPV4 antagonist may arise from a site other than the blood vessels themselves, perhaps in central cardiovascular control centres.
Collapse
Affiliation(s)
- Fiona O'Brien
- Department of Musculoskeletal Ageing, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Caroline A. Staunton
- Department of Musculoskeletal Ageing, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Ageing, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
19
|
Malacarne PF, Bezzenberger J, Lopez M, Warwick T, Müller N, Brandes RP, Rezende F. Epoxyeicosatrienoic Acid and Prostanoid Crosstalk at the Receptor and Intracellular Signaling Levels to Maintain Vascular Tone. Int J Mol Sci 2022; 23:ijms23115939. [PMID: 35682616 PMCID: PMC9180422 DOI: 10.3390/ijms23115939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are signaling lipids produced by the cytochrome P450-(CYP450)-mediated epoxygenation of arachidonic acid. EETs have numerous biological effects on the vascular system, but aspects including their species specificity make their effects on vascular tone controversial. CYP450 enzymes require the 450-reductase (POR) for their activity. We set out to determine the contribution of endothelial CYP450 to murine vascular function using isolated aortic ring preparations from tamoxifen-inducible endothelial cell-specific POR knockout mice (ecPOR-/-). Constrictor responses to phenylephrine were similar between control (CTR) and ecPOR-/- mice. Contrastingly, sensitivity to the thromboxane receptor agonist U46619 and prostaglandin E2 (PGE2) was increased following the deletion of POR. Ex vivo incubation with a non-hydrolyzable EET (14,15-EE-8(Z)-E, EEZE) reversed the increased sensitivity to U46619 to the levels of CTR. EETs had no effect on vascular tone in phenylephrine-preconstricted vessels, but dilated vessels contracted with U46619 or PGE2. As U46619 acts through RhoA-dependent kinase, this system was analyzed. The deletion of POR affected the expression of genes in this pathway and the inhibition of Rho-GTPase with SAR407899 decreased sensitivity to U46619. These data suggest that EET and prostanoid crosstalk at the receptor level and that lack of EET production sensitizes vessels to vasoconstriction via the induction of the Rho kinase system.
Collapse
Affiliation(s)
- Pedro Felipe Malacarne
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Justus Bezzenberger
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Melina Lopez
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Niklas Müller
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-6996; Fax: +49-69-6301-7668
| |
Collapse
|
20
|
Liu T, Dogan I, Rothe M, Kunz JV, Knauf F, Gollasch M, Luft FC, Gollasch B. Hemodialysis and Plasma Oxylipin Biotransformation in Peripheral Tissue. Metabolites 2022; 12:metabo12010034. [PMID: 35050156 PMCID: PMC8781597 DOI: 10.3390/metabo12010034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Factors causing the increased cardiovascular morbidity and mortality in hemodialysis (HD) patients are largely unknown. Oxylipins are a superclass of lipid mediators with potent bioactivities produced from oxygenation of polyunsaturated fatty acids. We previously assessed the impact of HD on oxylipins in arterial blood plasma and found that HD increases several oxylipins. To study the phenomenon further, we now evaluated the differences in arterial and venous blood oxylipins from patients undergoing HD. We collected arterial and venous blood samples in upper extremities from 12 end-stage renal disease (ESRD) patients before and after HD and measured oxylipins in plasma by LC-MS/MS tandem mass spectrometry. Comparison between cytochrome P450 (CYP), lipoxygenase (LOX), and LOX/CYP ω/(ω-1)-hydroxylase metabolites levels from arterial and venous blood showed no arteriovenous differences before HD but revealed arteriovenous differences in several CYP metabolites immediately after HD. These changes were explained by metabolites in the venous blood stream of the upper limb. Decreased soluble epoxide hydrolase (sEH) activity contributed to the release and accumulation of the CYP metabolites. However, HD did not affect arteriovenous differences of the majority of LOX and LOX/CYP ω/(ω-1)-hydroxylase metabolites. The HD treatment itself causes changes in CYP epoxy metabolites that could have deleterious effects in the circulation.
Collapse
Affiliation(s)
- Tong Liu
- Experimental and Clinical Research Center (ECRC), Charité Medical Faculty and Max Delbrück Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (T.L.); (M.G.); (F.C.L.)
| | - Inci Dogan
- LIPIDOMIX GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (I.D.); (M.R.)
| | - Michael Rothe
- LIPIDOMIX GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (I.D.); (M.R.)
| | - Julius V. Kunz
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (J.V.K.); (F.K.)
| | - Felix Knauf
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (J.V.K.); (F.K.)
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), Charité Medical Faculty and Max Delbrück Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (T.L.); (M.G.); (F.C.L.)
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Friedrich C. Luft
- Experimental and Clinical Research Center (ECRC), Charité Medical Faculty and Max Delbrück Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (T.L.); (M.G.); (F.C.L.)
| | - Benjamin Gollasch
- Experimental and Clinical Research Center (ECRC), Charité Medical Faculty and Max Delbrück Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (T.L.); (M.G.); (F.C.L.)
- HELIOS Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-540-249
| |
Collapse
|
21
|
Muñoz M, López-Oliva E, Pinilla E, Rodríguez C, Martínez MP, Contreras C, Gómez A, Benedito S, Sáenz-Medina J, Rivera L, Prieto D. Differential contribution of renal cytochrome P450 enzymes to kidney endothelial dysfunction and vascular oxidative stress in obesity. Biochem Pharmacol 2022; 195:114850. [PMID: 34822809 DOI: 10.1016/j.bcp.2021.114850] [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: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022]
Abstract
Arachidonic acid (AA)-derived cytochrome P450 (CYP) derivatives, epoxyeicosatrienoic acids (EETs) and 20-hidroxyeicosatetranoic acid (20-HETE), play a key role in kidney tubular and vascular functions and blood pressure. Altered metabolism of CYP epoxygenases and CYP hydroxylases has differentially been involved in the pathogenesis of metabolic disease-associated vascular complications, although the mechanisms responsible for the vascular injury are unclear. The present study aimed to assess whether obesity-induced changes in CYP enzymes may contribute to oxidative stress and endothelial dysfunction in kidney preglomerular arteries. Endothelial function and reactive oxygen species (ROS) production were assessed in interlobar arteries of obese Zucker rats (OZR) and their lean counterparts lean Zucker rats (LZR) and the effects of CYP2C and CYP4A inhibitors sulfaphenazole and HET0016, respectively, were examined on the endothelium-dependent relaxations and O2- and H2O2 levels of preglomerular arteries. Non-nitric oxide (NO) non-prostanoid endothelium-derived hyperpolarization (EDH)-type responses were preserved but resistant to the CYP epoxygenase blocker sulfaphenazole in OZR in contrast to those in LZR. Sulfaphenazole did not further inhibit reduced arterial H2O2 levels, and CYP2C11/CYP2C23 enzymes were downregulated in intrarenal arteries from OZR. Renal EDH-mediated relaxations were preserved in obese rats by the enhanced activity and expression of endothelial calcium-activated potassium channels (KCa). CYP4A blockade restored impaired NO-mediated dilatation and inhibited augmented O2- production in kidney arteries from OZR. The current data demonstrate that both decreased endothelial CYP2C11/ CYP2C23-derived vasodilator H2O2 and augmented CYP4A-derived 20-HETE contribute to endothelial dysfunction and vascular oxidative stress in obesity. CYP4A inhibitors ameliorate arterial oxidative stress and restore endothelial function which suggests its therapeutic potential for the vascular complications of obesity-associated kidney injury.
Collapse
Affiliation(s)
- Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Elvira López-Oliva
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Estéfano Pinilla
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Claudia Rodríguez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - María Pilar Martínez
- Departamento de Anatomía y Embriología, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Cristina Contreras
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Alfonso Gómez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Sara Benedito
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Javier Sáenz-Medina
- Departamento de Urología, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Luis Rivera
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
22
|
Peters EC, Gee MT, Pawlowski LN, Kath AM, Polk FD, Vance CJ, Sacoman JL, Pires PW. Amyloid- β disrupts unitary calcium entry through endothelial NMDA receptors in mouse cerebral arteries. J Cereb Blood Flow Metab 2022; 42:145-161. [PMID: 34465229 PMCID: PMC8721780 DOI: 10.1177/0271678x211039592] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/07/2023]
Abstract
Transient increases in intracellular Ca2+ activate endothelium-dependent vasodilatory pathways. This process is impaired in cerebral amyloid angiopathy, where amyloid-β(1-40) accumulates around blood vessels. In neurons, amyloid-β impairs the Ca2+-permeable N-methyl-D-aspartate receptor (NMDAR), a mediator of endothelium-dependent dilation in arteries. We hypothesized that amyloid-β(1-40) reduces NMDAR-elicited Ca2+ signals in mouse cerebral artery endothelial cells, blunting dilation. Cerebral arteries isolated from 4-5 months-old, male and female cdh5:Gcamp8 mice were used for imaging of unitary Ca2+ influx through NMDAR (NMDAR sparklets) and intracellular Ca2+ transients. The NMDAR agonist NMDA (10 µmol/L) increased frequency of NMDAR sparklets and intracellular Ca2+ transients in endothelial cells; these effects were prevented by NMDAR antagonists D-AP5 and MK-801. Next, we tested if amyloid-β(1-40) impairs NMDAR-elicited Ca2+ transients. Cerebral arteries incubated with amyloid-β(1-40) (5 µmol/L) exhibited reduced NMDAR sparklets and intracellular Ca2+ transients. Lastly, we observed that NMDA-induced dilation of pial arteries is reduced by acute intraluminal amyloid-β(1-40), as well as in a mouse model of Alzheimer's disease, the 5x-FAD, linked to downregulation of Grin1 mRNA compared to wild-type littermates. These data suggest that endothelial NMDAR mediate dilation via Ca2+-dependent pathways, a process disrupted by amyloid-β(1-40) and impaired in 5x-FAD mice.
Collapse
Affiliation(s)
- Emily C Peters
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Michael T Gee
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Lukas N Pawlowski
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Allison M Kath
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Felipe D Polk
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Christopher J Vance
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Juliana L Sacoman
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Paulo W Pires
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
- Sarver Heart Center, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| |
Collapse
|
23
|
Frömel T, Naeem Z, Pirzeh L, Fleming I. Cytochrome P450-derived fatty acid epoxides and diols in angiogenesis and stem cell biology. Pharmacol Ther 2021; 234:108049. [PMID: 34848204 DOI: 10.1016/j.pharmthera.2021.108049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/04/2021] [Accepted: 11/24/2021] [Indexed: 10/19/2022]
Abstract
Cytochrome P450 (CYP) enzymes are frequently referred to as the third pathway for the metabolism of arachidonic acid. While it is true that these enzymes generate arachidonic acid epoxides i.e. the epoxyeicosatrienoic acids (EETs), they are able to accept a wealth of ω-3 and ω-6 polyunsaturated fatty acids (PUFAs) to generate a large range of regio- and stereo-isomers with distinct biochemical properties and physiological actions. Probably the best studied are the EETs which have well documented effects on vascular reactivity and angiogenesis. CYP enzymes can also participate in crosstalk with other PUFA pathways and metabolize prostaglandin G2 and H2, which are the precursors of effector prostaglandins, to affect macrophage function and lymphangiogenesis. The activity of the PUFA epoxides is thought to be kept in check by the activity of epoxide hydrolases. However, rather than being inactive, the diols generated have been shown to regulate neutrophil activation, stem and progenitor cell proliferation and Notch signaling in addition to acting as exercise-induced lipokines. Excessive production of PUFA diols has also been implicated in pathologies such as severe respiratory distress syndromes, including COVID-19, and diabetic retinopathy. This review highlights some of the recent findings related to this pathway that affect angiogenesis and stem cell biology.
Collapse
Affiliation(s)
- Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Zumer Naeem
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Lale Pirzeh
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK) Partner Site Rhein-Main, Frankfurt am Main, Germany; The Cardio-Pulmonary Institute, Frankfurt am Main, Germany.
| |
Collapse
|
24
|
Alaeddine LM, Harb F, Hamza M, Dia B, Mogharbil N, Azar NS, Noureldein MH, El Khoury M, Sabra R, Eid AA. Pharmacological regulation of cytochrome P450 metabolites of arachidonic acid attenuates cardiac injury in diabetic rats. Transl Res 2021; 235:85-101. [PMID: 33746109 DOI: 10.1016/j.trsl.2021.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/13/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a well-established complication of type 1 and type 2 diabetes associated with a high rate of morbidity and mortality. DCM is diagnosed at advanced and irreversible stages. Therefore, it is of utmost need to identify novel mechanistic pathways involved at early stages to prevent or reverse the development of DCM. In vivo experiments were performed on type 1 diabetic rats (T1DM). Functional and structural studies of the heart were executed and correlated with mechanistic assessments exploring the role of cytochromes P450 metabolites, the 20-hydroxyeicosatetraenoic acids (20-HETEs) and epoxyeicosatrienoic acids (EETs), and their crosstalk with other homeostatic signaling molecules. Our data displays that hyperglycemia results in CYP4A upregulation and CYP2C11 downregulation in the left ventricles (LV) of T1DM rats, paralleled by a differential alteration in their metabolites 20-HETEs (increased) and EETs (decreased). These changes are concomitant with reductions in cardiac outputs, LV hypertrophy, fibrosis, and increased activation of cardiac fetal and hypertrophic genes. Besides, pro-fibrotic cytokine TGF-ß overexpression and NADPH (Nox4) dependent-ROS overproduction are also correlated with the observed cardiac functional and structural modifications. Of interest, these observations are attenuated when T1DM rats are treated with 12-(3-adamantan-1-yl-ureido) dodecanoic acid (AUDA), which blocks EETs metabolism, or N-hydroxy-N'-(4-butyl-2-methylphenol)Formamidine (HET0016), which inhibits 20-HETEs formation. Taken together, our findings confer pioneering evidence about a potential interplay between CYP450-derived metabolites and Nox4/TGF-β axis leading to DCM. Pharmacologic interventions targeting the inhibition of 20-HETEs synthesis or the activation of EETs synthesis may offer novel therapeutic approaches to treat DCM.
Collapse
Affiliation(s)
- Lynn M Alaeddine
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Frederic Harb
- Department of Biology, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Maysaa Hamza
- Department of Pharmacology and Toxicology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Batoul Dia
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Nahed Mogharbil
- Department of Pharmacology and Toxicology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Nadim S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohamed H Noureldein
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Mirella El Khoury
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Ramzi Sabra
- Department of Pharmacology and Toxicology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
25
|
Chen M, Li X. Role of TRPV4 channel in vasodilation and neovascularization. Microcirculation 2021; 28:e12703. [PMID: 33971061 DOI: 10.1111/micc.12703] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/02/2021] [Indexed: 12/12/2022]
Abstract
The transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+ -permeable nonselective cation channel, is widely distributed in the circulatory system, particularly in vascular endothelial cells (ECs) and smooth muscle cells (SMCs). The TRPV4 channel is activated by various endogenous and exogenous stimuli, including shear stress, low intravascular pressure, and arachidonic acid. TRPV4 has a role in mediating vascular tone and arterial blood pressure. The activation of the TRPV4 channel induces Ca2+ influx, thereby resulting in endothelium-dependent hyperpolarization and SMC relaxation through SKCa and IKCa activation on ECs or through BKCa activation on SMCs. Ca2+ binds to calmodulin, which leads to the production of nitric oxide, causing vasodilation. Furthermore, the TRPV4 channel plays an important role in angiogenesis and arteriogenesis and is critical for tumor angiogenesis and growth, since it promotes or inhibits the development of various types of cancer. The TRPV4 channel is involved in the active growth of collateral arteries induced by flow shear stress, which makes it a promising therapeutic target in the occlusion or stenosis of the main arteries. In this review, we explore the role and the potential mechanism of action of the TRPV4 channel in the regulation of vascular tone and in the induction of neovascularization to provide a reference for future research.
Collapse
Affiliation(s)
- Miao Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xiucun Li
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Anatomy and Histoembryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
26
|
Intrinsic exercise capacity induces divergent vascular plasticity via arachidonic acid-mediated inflammatory pathways in female rats. Vascul Pharmacol 2021; 140:106862. [PMID: 33872803 DOI: 10.1016/j.vph.2021.106862] [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: 12/03/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 12/31/2022]
Abstract
Metabolic syndrome prevalence has increased among US adults, particularly among non-hispanic white and black women. Sedentary behavior often leads to chronic inflammation, a triggering factor of metabolic syndrome. Given that intrinsic exercise capacity is genetically inherited, we questioned if low-grade chronic inflammation would be present in a female rat model of low intrinsic exercise capacity-induced metabolic syndrome, while beneficial increase of resolution of inflammation would be present in a female rat model of high intrinsic exercise capacity. In the vascular system, two primary markers for inflammation and resolution of inflammation are cyclooxygenase (COX) and lipoxygenase (LOX), respectively. Our study focused on the novel hypothesis that untrained, inherited exercise capacity induces divergent vascular plasticity via changes in the delicate balance between COX and LOX inflammatory mediators. We used divergent rat strains with low (LCR) and high (HCR) aerobic running capacity. By using animals with contrasting intrinsic exercise capacities, it is possible to determine the exact triggers that lead to inherited vascular plasticity in female rats. We observed that female LCR displayed increased periovarian fat pad and body weight, which is congruent with their obesity-presenting phenotype. Furthermore, LCR presented with vascular hypocontractility and increased COX and LOX-derived pro-inflammatory factors. On the other hand, HCR presented with a "shutdown" of COX-induced vasoconstriction and enhanced resolution of inflammation to maintain vascular tone and homeostasis. In conclusion, LCR display low-grade chronic inflammation via increased COX activity. These results provide mechanistic clues as to why lower intrinsic aerobic capacity correlates with a predisposition to risk of vascular disease. Conversely, being born with higher intrinsic aerobic capacity is a significant factor for improved vascular physiology in female rats.
Collapse
|
27
|
Lai J, Chen C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Front Physiol 2021; 12:642470. [PMID: 33716791 PMCID: PMC7943617 DOI: 10.3389/fphys.2021.642470] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid by cytochrome P450 (CYP) epoxygenases, which include four regioisomers: 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET. Each of them possesses beneficial effects against inflammation, fibrosis, and apoptosis, which could combat cardiovascular diseases. Numerous studies have demonstrated that elevation of EETs by overexpression of CYP2J2, inhibition of sEH, or treatment with EET analogs showed protective effects in various cardiovascular diseases, including hypertension, myocardial infarction, and heart failure. As is known to all, cardiac remodeling is the major pathogenesis of cardiovascular diseases. This review will begin with the introduction of EETs and their protective effects in cardiovascular diseases. In the following, the roles of EETs in cardiac remodeling, with a particular emphasis on myocardial hypertrophy, apoptosis, fibrosis, inflammation, and angiogenesis, will be summarized. Finally, it is suggested that upregulation of EETs is a potential therapeutic strategy for cardiovascular diseases. The EET-related drug development against cardiac remodeling is also discussed, including the overexpression of CYP2J2, inhibition of sEH, and the analogs of EET.
Collapse
Affiliation(s)
- Jinsheng Lai
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Ashby JW, Mack JJ. Endothelial Control of Cerebral Blood Flow. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1906-1916. [PMID: 33713686 DOI: 10.1016/j.ajpath.2021.02.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/19/2022]
Abstract
Since constant perfusion of blood throughout the brain is critical for neuronal health, the regulation of cerebral blood flow is complex and highly controlled. This regulation is controlled, in part, by the cerebral endothelium. In this review, multiple modes of endothelium-derived blood flow regulation is discussed, including chemical control of vascular tone, heterotypic and homotypic cell-cell interactions, second messenger signaling, and cellular response to physical forces and inflammatory mediators. Because cerebral small vessel disease is often associated with endothelial dysfunction and a compromised blood-brain barrier, understanding the endothelial factors that regulate vessel function to maintain cerebral blood flow and prevent vascular permeability may provide insights into disease prevention and treatment.
Collapse
Affiliation(s)
- Julianne W Ashby
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Julia J Mack
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California.
| |
Collapse
|
29
|
Kinins and Kinin Receptors in Cardiovascular and Renal Diseases. Pharmaceuticals (Basel) 2021; 14:ph14030240. [PMID: 33800422 PMCID: PMC8000381 DOI: 10.3390/ph14030240] [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: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
This review addresses the physiological role of the kallikrein–kinin system in arteries, heart and kidney and the consequences of kallikrein and kinin actions in diseases affecting these organs, especially ischemic and diabetic diseases. Emphasis is put on pharmacological and genetic studies targeting kallikrein; ACE/kininase II; and the two kinin receptors, B1 (B1R) and B2 (B2R), distinguished through the work of Domenico Regoli and his collaborators. Potential therapeutic interest and limitations of the pharmacological manipulation of B1R or B2R activity in cardiovascular and renal diseases are discussed. This discussion addresses either the activation or inhibition of these receptors, based on recent clinical and experimental studies.
Collapse
|
30
|
TRPing to the Point of Clarity: Understanding the Function of the Complex TRPV4 Ion Channel. Cells 2021; 10:cells10010165. [PMID: 33467654 PMCID: PMC7830798 DOI: 10.3390/cells10010165] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
The transient receptor potential vanilloid 4 channel (TRPV4) belongs to the mammalian TRP superfamily of cation channels. TRPV4 is ubiquitously expressed, activated by a disparate array of stimuli, interacts with a multitude of proteins, and is modulated by a range of post-translational modifications, the majority of which we are only just beginning to understand. Not surprisingly, a great number of physiological roles have emerged for TRPV4, as have various disease states that are attributable to the absence, or abnormal functioning, of this ion channel. This review will highlight structural features of TRPV4, endogenous and exogenous activators of the channel, and discuss the reported roles of TRPV4 in health and disease.
Collapse
|
31
|
Mori A, Yano E, Sakamoto K, Ishii K, Nakahara T. Role of Epoxyeicosatrienoic Acids in Acetylcholine-Induced Dilation of Rat Retinal Arterioles in Vivo. Biol Pharm Bull 2021; 44:82-87. [PMID: 33390554 DOI: 10.1248/bpb.b20-00635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CYP epoxygenase-derived epoxyeicosatrienoic acids (EETs) contribute to endothelium-dependent hyperpolarization (EDH)-related dilation in multiple vascular beds. The present study aimed to determine the role of EETs in the acetylcholine (ACh)-induced dilation of retinal arterioles in rats in vivo. The vasodilator responses were assessed by determining the change in diameter of the retinal arterioles on images of the ocular fundus. The intravitreal injection of 17-octadecynoic acid (1.4 nmol/eye), an inhibitor of CYP epoxygenase, and 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EE-5(Z)-E; 2 nmol/eye), an antagonist of EETs, reduced the ACh (0.3-10 µg/kg/min)-induced dilation of the retinal arterioles. The EET antagonist attenuated the vasodilator response to ACh under blockade of nitric oxide (NO) synthases and cyclooxygenases with NG-nitro-L-arginine methyl ester (30 mg/kg) plus indomethacin (5 mg/kg). Intravitreal injection of 14,15-EET (0.5 nmol/eye) dilated retinal arterioles and the response was prevented by iberiotoxin, an inhibitor of large-conductance Ca2+-activated K+ (BKCa) channels (20 pmol/eye). These results suggest that ACh stimulates the production of EETs, thereby dilating the retinal arterioles via activation of BKCa channels. CYP epoxygenase-derived EETs may be involved in the EDH-related component of the ACh-induced dilation of the retinal arterioles.
Collapse
Affiliation(s)
- Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences
| | - Erika Yano
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences
| | - Kunio Ishii
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences
| |
Collapse
|
32
|
Hu Y, Chen M, Wang M, Li X. Flow-mediated vasodilation through mechanosensitive G protein-coupled receptors in endothelial cells. Trends Cardiovasc Med 2021; 32:61-70. [PMID: 33406458 DOI: 10.1016/j.tcm.2020.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022]
Abstract
Currently, endothelium-dependent vasodilatation involves three main mechanisms: production of nitric oxide (NO) by endothelial nitric oxide synthase (eNOS), synthesis of prostanoids by cyclooxygenase, and/or opening of calcium-sensitive potassium channels. Researchers have proposed multiple mechanosensors that may be involved in flow-mediated vasodilation (FMD), including G protein-coupled receptors (GPCRs), ion channels, and intercellular junction proteins, among others. However, GPCRs are considered the major mechanosensors that play a pivotal role in shear stress signal transduction. Among mechanosensitive GPCRs, G protein-coupled receptor 68, histamine H1 receptors, sphingosine-1-phosphate receptor 1, and bradykinin B2 receptors have been identified as endothelial sensors of flow shear stress regulating flow-mediated vasodilation. Thus, this review aims to expound on the mechanism whereby flow shear stress promotes vasodilation through the proposed mechanosensitive GPCRs in ECs.
Collapse
Affiliation(s)
- Yong Hu
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.247, Beiyuan Street, Jinan, Shandong Province, 250031, China.
| | - Miao Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun, Jilin Province, 130021, China.
| | - Meili Wang
- Department of Obstetrics, Maternal and Child Health Care Hospital of Shandong Province, Shandong University, NO.238, Jingshi East Road, Jinan, Shandong, 250012, China.
| | - Xiucun Li
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.247, Beiyuan Street, Jinan, Shandong Province, 250031, China; Department of Anatomy and Histoembryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, NO.44, Wenhua West Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
33
|
Discovery of novel urea-diarylpyrazole hybrids as dual COX-2/sEH inhibitors with improved anti-inflammatory activity and highly reduced cardiovascular risks. Eur J Med Chem 2020; 205:112662. [DOI: 10.1016/j.ejmech.2020.112662] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 11/21/2022]
|
34
|
Krüger-Genge A, Jung F, Hufert F, Jung EM, Küpper JH, Storsberg J. Effects of gut microbial metabolite trimethylamine N-oxide (TMAO) on platelets and endothelial cells. Clin Hemorheol Microcirc 2020; 76:309-316. [PMID: 32925010 DOI: 10.3233/ch-209206] [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: 11/15/2022]
Abstract
Thrombotic events result from different pathologies and are the underlying causes of severe diseases like stroke or myocardial infarction. Recent basic research now revealed a link between food uptake, food conversion and gut metabolism. Gut microbial production of trimethylamine N-oxide (TMAO) from dietary nutrients like choline, lecithin and L-carnitine was associated with the development of cardiovascular diseases. Within this review we give a systematic overview about the influence of TMAO on blood components like platelets and endothelial cells which both are involved as key players in thrombotic processes. In summary, a mechanistic correlation between the gut microbiome, TMAO and cardiovascular diseases becomes obvious and emphasizes to the significance of the intestinal microbiome.
Collapse
Affiliation(s)
- A Krüger-Genge
- Fraunhofer-Institute for Applied Polymer Research (IAP), Potsdam-Golm, Germany
| | - F Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Senftenberg, Germany
| | - F Hufert
- Institute for Microbiology and Virology, Medizinische Hochschule Brandenburg Theodor Fontane, Senftenberg, Germany
| | - E-M Jung
- Department of Radiology and Interdisciplinary Ultrasound Department, University Hospital Regensburg, Regensburg, Germany
| | - J-H Küpper
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Senftenberg, Germany
| | - J Storsberg
- Fraunhofer-Institute for Applied Polymer Research (IAP), Potsdam-Golm, Germany
| |
Collapse
|
35
|
Matin N, Fisher C, Lansdell TA, Hammock BD, Yang J, Jackson WF, Dorrance AM. Soluble epoxide hydrolase inhibition improves cognitive function and parenchymal artery dilation in a hypertensive model of chronic cerebral hypoperfusion. Microcirculation 2020; 28:e12653. [PMID: 32767848 DOI: 10.1111/micc.12653] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/30/2020] [Accepted: 07/30/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Parenchymal arterioles (PAs) regulate perfusion of the cerebral microcirculation, and impaired PA endothelium-dependent dilation occurs in dementia models mimicking chronic cerebral hypoperfusion (CCH). Epoxyeicosatrienoic acids (EETs) are vasodilators; their actions are potentiated by soluble epoxide hydrolase (sEH) inhibition. We hypothesized that chronic sEH inhibition with trifluoromethoxyphenyl-3 (1-propionylpiperidin-4-yl) urea (TPPU) would prevent cognitive dysfunction and improve PA dilation in a hypertensive CCH model. METHODS Bilateral carotid artery stenosis (BCAS) was used to induce CCH in twenty-week-old male stroke-prone spontaneously hypertensive rats (SHSRP) that were treated with vehicle or TPPU for 8 weeks. Cognitive function was assessed by novel object recognition. PA dilation and structure were assessed by pressure myography, and mRNA expression in brain tissue was assessed by qRT-PCR. RESULTS TPPU did not enhance resting cerebral perfusion, but prevented CCH-induced memory deficits. TPPU improved PA endothelium-dependent dilation but reduced the sensitivity of PAs to a nitric oxide donor. TPPU treatment had no effect on PA structure or biomechanical properties. TPPU treatment increased brain mRNA expression of brain derived neurotrophic factor, doublecortin, tumor necrosis factor-alpha, sEH, and superoxide dismutase 3, CONCLUSIONS: These data suggest that sEH inhibitors may be viable treatments for cognitive impairments associated with hypertension and CCH.
Collapse
Affiliation(s)
- Nusrat Matin
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Courtney Fisher
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Theresa A Lansdell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Bruce D Hammock
- Department of Entomology &, University of California Comprehensive Cancer Center, Davis, CA, USA
| | - Jun Yang
- Department of Entomology &, University of California Comprehensive Cancer Center, Davis, CA, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
36
|
Pernow J, Mahdi A, Yang J, Zhou Z. Red blood cell dysfunction: a new player in cardiovascular disease. Cardiovasc Res 2020; 115:1596-1605. [PMID: 31198931 DOI: 10.1093/cvr/cvz156] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/07/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023] Open
Abstract
The primary role of red blood cells (RBCs) is to transport oxygen to the tissues and carbon dioxide to the lungs. However, emerging evidence suggests an important role of the RBC beyond being just a passive carrier of the respiratory gases. The RBCs are of importance for redox balance and are actively involved in the regulation of vascular tone, especially during hypoxic and ischaemic conditions by the release of nitric oxide (NO) bioactivity and adenosine triphosphate. The role of the RBC has gained further interest after recent discoveries demonstrating a markedly altered function of the cell in several pathological conditions. Such alterations include increased adhesion capability, increased formation of reactive oxygen species as well as altered protein content and enzymatic activities. Beyond signalling increased oxidative stress, the altered function of RBCs is characterized by reduced export of NO bioactivity regulated by increased arginase activity. Of further importance, the altered function of RBCs has important implications for several cardiovascular disease conditions. RBCs have been shown to induce endothelial dysfunction and to increase cardiac injury during ischaemia-reperfusion in diabetes mellitus. Finally, this new knowledge has led to novel therapeutic possibilities to intervene against cardiovascular disease by targeting signalling in the RBC. These novel data open up an entirely new view on the underlying pathophysiological mechanisms behind the cardiovascular disease processes in diabetes mellitus mediated by the RBC. This review highlights the current knowledge regarding the role of RBCs in cardiovascular regulation with focus on their importance for cardiovascular dysfunction in pathological conditions and therapeutic possibilities for targeting RBCs in cardiovascular disease.
Collapse
Affiliation(s)
- John Pernow
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - Ali Mahdi
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Abstract
Of the 21 members of the connexin family, 4 (Cx37, Cx40, Cx43, and Cx45) are expressed in the endothelium and/or smooth muscle of intact blood vessels to a variable and dynamically regulated degree. Full-length connexins oligomerize and form channel structures connecting the cytosol of adjacent cells (gap junctions) or the cytosol with the extracellular space (hemichannels). The different connexins vary mainly with regard to length and sequence of their cytosolic COOH-terminal tails. These COOH-terminal parts, which in the case of Cx43 are also translated as independent short isoforms, are involved in various cellular signaling cascades and regulate cell functions. This review focuses on channel-dependent and -independent effects of connexins in vascular cells. Channels play an essential role in coordinating and synchronizing endothelial and smooth muscle activity and in their interplay, in the control of vasomotor actions of blood vessels including endothelial cell reactivity to agonist stimulation, nitric oxide-dependent dilation, and endothelial-derived hyperpolarizing factor-type responses. Further channel-dependent and -independent roles of connexins in blood vessel function range from basic processes of vascular remodeling and angiogenesis to vascular permeability and interactions with leukocytes with the vessel wall. Together, these connexin functions constitute an often underestimated basis for the enormous plasticity of vascular morphology and function enabling the required dynamic adaptation of the vascular system to varying tissue demands.
Collapse
Affiliation(s)
- Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany; Biomedical Centre, Cardiovascular Physiology, LMU Munich, Planegg-Martinsried, Germany; German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
38
|
Gollasch B, Dogan I, Rothe M, Gollasch M, Luft FC. Maximal exercise and plasma cytochrome P450 and lipoxygenase mediators: a lipidomics study. Physiol Rep 2020; 7:e14165. [PMID: 31304687 PMCID: PMC6640589 DOI: 10.14814/phy2.14165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 01/15/2023] Open
Abstract
Epoxides derived from arachidonic acid (AA) are released during exercise and may contribute to vasodilation. However, exercise may also affect circulating levels of other epoxides derived from cytochromes P450 (CYP) monooxygenase and lipoxygenase (LOX) pathways, many of whose exhibit cardiovascular activity in vitro. The effects of exercise on their levels have not been documented. We tested the hypothesis that acute, maximal exercise would influence the plasma concentrations of these vasoactive substances. We measured plasma CYP and LOX mediators derived from both the n − 3 and n − 6 fatty acid (FA) classes in healthy volunteers before, during and after short‐term exhaustive exercise. Lipid mediators were profiled by means of LC–MS/MS tandem mass spectrometry. A maximal Bruce treadmill test was performed to voluntary exhaustion. Exhaustive exercise increased the circulating levels of epoxyoctadecenoic (12,13‐EpOME), dihydroxyeicosatrienoic (5,6‐DHET), dihydroxyeicosatetraenoic acids (5,6‐DiHETE, 17,18‐DiHETE), but had no effect on the majority of CYP and LOX metabolites. Although our calculations of diol/epoxide ratios revealed preferred hydrolysis of epoxyeicosatrienoic acids (EEQs) into their diols (DiHETEs), this hydrolysis was resistant to maximal exercise. Our study is the first documentation that bioactive endogenous n − 3 and n − 6 CYP lipid mediators are released by short‐term exhaustive exercise in humans. In particular, the CYP epoxy‐metabolite status, 12,13‐EpOME/DiHOME, 5,6‐EET/DHET, 5,6‐EEQ/DiHETE and 17,18‐EEQ/DiHETE may contribute to the cardiovascular response during maximal exercise.
Collapse
Affiliation(s)
- Benjamin Gollasch
- Experimental and Clinical Research Center (ECRC), a Joint Institution between the Charité University Medicine, Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany.,HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | | | | | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), a Joint Institution between the Charité University Medicine, Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), a Joint Institution between the Charité University Medicine, Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany.,Max-Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
39
|
Gollasch B, Wu G, Dogan I, Rothe M, Gollasch M, Luft FC. Effects of hemodialysis on plasma oxylipins. Physiol Rep 2020; 8:e14447. [PMID: 32562348 PMCID: PMC7305238 DOI: 10.14814/phy2.14447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/04/2020] [Accepted: 04/24/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is an important risk factor for cardiovascular and all‐cause mortality. Survival rates among end‐stage renal disease (ESRD) hemodialysis patients are poor and most deaths are related to cardiovascular disease. Oxylipins constitute a family of oxygenated natural products, formed from fatty acid by pathways involving at least one step of dioxygen‐dependent oxidation. They are derived from polyunsaturated fatty acids (PUFAs) by cyclooxygenase (COX) enzymes, by lipoxygenases (LOX) enzymes, or by cytochrome P450 epoxygenase. Oxylipins have physiological significance and some could be of regulatory importance. The effects of decreased renal function and dialysis treatment on oxylipin metabolism are unknown. We studied 15 healthy persons and 15 CKD patients undergoing regular hemodialysis treatments and measured oxylipins (HPLC‐MS lipidomics) derived from cytochrome P450 (CYP) monooxygenase and lipoxygenase (LOX)/CYP ω/(ω‐1)‐hydroxylase pathways in circulating blood. We found that all four subclasses of CYP epoxy metabolites were increased after the dialysis treatment. Rather than resulting from altered soluble epoxide hydrolase (sEH) activity, the oxylipins were released and accumulated in the circulation. Furthermore, hemodialysis did not change the majority of LOX/CYP ω/(ω‐1)‐hydroxylase metabolites. Our data support the idea that oxylipin profiles discriminate ESRD patients from normal controls and are influenced by renal replacement therapies.
Collapse
Affiliation(s)
- Benjamin Gollasch
- Experimental and Clinical Research Center (ECRC), a joint institution between the Charité University Medicine and Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany.,HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Guanlin Wu
- Experimental and Clinical Research Center (ECRC), a joint institution between the Charité University Medicine and Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany.,Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | | | | | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), a joint institution between the Charité University Medicine and Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany.,Department of Geriatrics, University of Greifswald, University District Hospital Wolgast, Greifswald, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), a joint institution between the Charité University Medicine and Max Delbrück Center (MDC) for Molecular Medicine, Berlin-Buch, Germany
| |
Collapse
|
40
|
Evangelista EA, Cho CW, Aliwarga T, Totah RA. Expression and Function of Eicosanoid-Producing Cytochrome P450 Enzymes in Solid Tumors. Front Pharmacol 2020; 11:828. [PMID: 32581794 PMCID: PMC7295938 DOI: 10.3389/fphar.2020.00828] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
Oxylipins derived from the oxidation of polyunsaturated fatty acids (PUFAs) act as important paracrine and autocrine signaling molecules. A subclass of oxylipins, the eicosanoids, have a broad range of physiological outcomes in inflammation, the immune response, cardiovascular homeostasis, and cell growth regulation. Consequently, eicosanoids are implicated in the pathophysiology of various diseases, most notably cancer, where eicosanoid mediated signaling is involved in tumor development, progression, and angiogenesis. Cytochrome P450s (CYPs) are a superfamily of heme monooxygenases generally involved in the clearance of xenobiotics while a subset of isozymes oxidize PUFAs to eicosanoids. Several eicosanoid forming CYPs are overexpressed in tumors, elevating eicosanoid levels and suggesting a key function in tumorigenesis and progression of tumors in the lung, breast, prostate, and kidney. This review summarizes the current understanding of CYPs' involvement in solid tumor etiology and progression providing supporting public data for gene expression from The Cancer Genome Atlas.
Collapse
Affiliation(s)
- Eric A Evangelista
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Christi W Cho
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Theresa Aliwarga
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Rheem A Totah
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, United States
| |
Collapse
|
41
|
Negri S, Faris P, Rosti V, Antognazza MR, Lodola F, Moccia F. Endothelial TRPV1 as an Emerging Molecular Target to Promote Therapeutic Angiogenesis. Cells 2020; 9:cells9061341. [PMID: 32471282 PMCID: PMC7349285 DOI: 10.3390/cells9061341] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Therapeutic angiogenesis represents an emerging strategy to treat ischemic diseases by stimulating blood vessel growth to rescue local blood perfusion. Therefore, injured microvasculature may be repaired by stimulating resident endothelial cells or circulating endothelial colony forming cells (ECFCs) or by autologous cell-based therapy. Endothelial Ca2+ signals represent a crucial player in angiogenesis and vasculogenesis; indeed, several angiogenic stimuli induce neovessel formation through an increase in intracellular Ca2+ concentration. Several members of the Transient Receptor Potential (TRP) channel superfamily are expressed and mediate Ca2+-dependent functions in vascular endothelial cells and in ECFCs, the only known truly endothelial precursor. TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis, through the integration of several chemical stimuli. Herein, we first summarize TRPV1 structure and gating mechanisms. Next, we illustrate the physiological roles of TRPV1 in vascular endothelium, focusing our attention on how endothelial TRPV1 promotes angiogenesis. In particular, we describe a recent strategy to stimulate TRPV1-mediated pro-angiogenic activity in ECFCs, in the presence of a photosensitive conjugated polymer. Taken together, these observations suggest that TRPV1 represents a useful target in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Lodola
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
- Correspondence:
| |
Collapse
|
42
|
Zhang Q, Tsuji-Hosokawa A, Willson C, Watanabe M, Si R, Lai N, Wang Z, Yuan JXJ, Wang J, Makino A. Chloroquine differentially modulates coronary vasodilation in control and diabetic mice. Br J Pharmacol 2020; 177:314-327. [PMID: 31503328 DOI: 10.1111/bph.14864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Chloroquine is a traditional medicine to treat malaria. There is increasing evidence that chloroquine not only induces phagocytosis but regulates vascular tone. Few reports investigating the effect of chloroquine on vascular responsiveness of coronary arteries have been made. In this study, we examined how chloroquine affected endothelium-dependent relaxation in coronary arteries under normal and diabetic conditions. EXPERIMENTAL APPROACH We isolated coronary arteries from mice and examined endothelium-dependent relaxation (EDR). Human coronary endothelial cells and mouse coronary endothelial cells isolated from control and diabetic mouse (TALLYHO/Jng [TH] mice, a spontaneous type 2 diabetic mouse model) were used for the molecular biological or cytosolic NO and Ca2+ measurements. KEY RESULTS Chloroquine inhibited endothelium-derived NO-dependent relaxation but had negligible effect on endothelium-derived hyperpolarization (EDH)-dependent relaxation in coronary arteries of control mice. Chloroquine significantly decreased NO production in control human coronary endothelial cells partly by phosphorylating eNOSThr495 (an inhibitory phosphorylation site of eNOS) and attenuating the rise of cytosolic Ca2+ concentration after stimulation. EDR was significantly inhibited in diabetic mice in comparison to control mice. Interestingly, chloroquine enhanced EDR in diabetic coronary arteries by, specifically, increasing EDH-dependent relaxation due partly to its augmenting effect on gap junction activity in diabetic mouse coronary endothelial cells. CONCLUSIONS AND IMPLICATIONS These data indicate that chloroquine affects vascular relaxation differently under normal and diabetic conditions. Therefore, the patients' health condition such as coronary macrovascular or microvascular disease, with or without diabetes, must be taken account into the consideration when selecting chloroquine for the treatment of malaria.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Physiology, The University of Arizona, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Conor Willson
- Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Makiko Watanabe
- Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Rui Si
- Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Ning Lai
- Department of Medicine, University of California, San Diego, La Jolla, California.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ziyi Wang
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Medicine, The University of Arizona, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Jian Wang
- Department of Medicine, The University of Arizona, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Physiology, The University of Arizona, Tucson, Arizona.,Department of Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
43
|
Edwards JM, McCarthy CG, Wenceslau CF. The Obligatory Role of the Acetylcholine-Induced Endothelium-Dependent Contraction in Hypertension: Can Arachidonic Acid Resolve this Inflammation? Curr Pharm Des 2020; 26:3723-3732. [PMID: 32303165 PMCID: PMC7542659 DOI: 10.2174/1381612826666200417150121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
The endothelium produces many substances that can regulate vascular tone. Acetylcholine is a widely used pharmacological tool to assess endothelial function. In general, acetylcholine binds to G-protein coupled muscarinic receptors that mediate a transient elevation in intracellular, free calcium. This intracellular rise in calcium is responsible for triggering several cellular responses, including the synthesis of nitric oxide, endothelium- derived hyperpolarizing factor, and eicosanoids derived from arachidonic acid. Endothelial arachidonic acid metabolism is also an important signaling pathway for mediating inflammation. Therefore, in conditions with sustained and excessive inflammation such as hypertension, arachidonic acid serves as a substrate for the synthesis of several vasoconstrictive metabolites, predominantly via the cyclooxygenase and lipoxygenase enzymes. Cyclooxygenase and lipoxygenase products can then activate G-protein coupled receptors expressed on vascular smooth muscle cells to causes contractile responses. As a result, acetylcholine-induced contraction due to arachidonic acid is a commonly observed feature of endothelial dysfunction and vascular inflammation in hypertension. In this review, we will critically analyze the literature supporting this concept, as well as address the potential underlying mechanisms, including the possibility that arachidonic acid signaling is diverted away from the synthesis of pro-resolving metabolites in conditions such as hypertension.
Collapse
Affiliation(s)
- Jonnelle M. Edwards
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Cameron G. McCarthy
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Camilla F. Wenceslau
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| |
Collapse
|
44
|
Basic Concepts of the Microcirculation. Microcirculation 2020. [DOI: 10.1007/978-3-030-28199-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
Dos Santos LRB, Fleming I. Role of cytochrome P450-derived, polyunsaturated fatty acid mediators in diabetes and the metabolic syndrome. Prostaglandins Other Lipid Mediat 2019; 148:106407. [PMID: 31899373 DOI: 10.1016/j.prostaglandins.2019.106407] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 11/14/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022]
Abstract
Over the last decade, cases of metabolic syndrome and type II diabetes have increased exponentially. Exercise and ω-3 polyunsaturated fatty acid (PUFA)-enriched diets are usually prescribed but no therapy is effectively able to restore the impaired glucose metabolism, hypertension, and atherogenic dyslipidemia encountered by diabetic patients. PUFAs are metabolized by different enzymes into bioactive metabolites with anti- or pro-inflammatory activity. One important class of PUFA metabolizing enzymes are the cytochrome P450 (CYP) enzymes that can generate a series of bioactive products, many of which have been attributed protective/anti-inflammatory and insulin-sensitizing effects in animal models. PUFA epoxides are, however, further metabolized by the soluble epoxide hydrolase (sEH) to fatty acid diols. The biological actions of the latter are less well understood but while low concentrations may be biologically important, higher concentrations of diols derived from linoleic acid and docosahexaenoic acid have been linked with inflammation. One potential application for sEH inhibitors is in the treatment of diabetic retinopathy where sEH expression and activity is elevated as are levels of a diol of docosahexaenoic acid that can induce the destabilization of the retina vasculature.
Collapse
Affiliation(s)
- Laila R B Dos Santos
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany; German Centre for Cardiovascular Research (DZHK) Partner Site Rhein-Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany; German Centre for Cardiovascular Research (DZHK) Partner Site Rhein-Main, Germany.
| |
Collapse
|
46
|
Hoxha M, Zappacosta B. CYP-derived eicosanoids: Implications for rheumatoid arthritis. Prostaglandins Other Lipid Mediat 2019; 146:106405. [PMID: 31838196 DOI: 10.1016/j.prostaglandins.2019.106405] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 12/18/2022]
Abstract
Today the role of cytochrome P450 metabolites in inflammatory rheumatic disease, such as rheumatoid arthritis (RA) is still poorly understood. In this review we survey the current knowledge on cytochrome P450 metabolites in rheumatoid arthritis. The balance between CYP epoxygenase- and CYP ω- hydroxylase is correlated to the regulation of NF-κB. In RA patients synovial fluid there are higher levels of IL-6, which suppresses activities of CYP enzymes, such as CYP3A, CYP2C19, CYP2C9, and CYP1A2. EETs have anti-inflammatory effects, probably attributed to the PPARγ activation. EETs inhibit bone resorption and osteoclastogenesis, and can be considered as an innovative therapeutic strategy for rheumatoid arthritis. In reference to the CYP ɷ-hydroxylase pathway, 20-HETE is a pro-inflammatory mediator. While there is scarce information on the role of 20-HETE inhibitors and its antagonists in rheumatoid arthritis, the elevation of EETs levels by sEH inhibitors is a promising therapeutic strategy for rheumatoid arthritis patients. In addition, hybrid compounds, such as sEH inhibitors/FLAP inhibitors, or sEHI combined with NSAIDs/COXIBs are also important therapeutic target. However, studies investigating the effects of inflammation and rheumatic disease on CYP-mediated eicosanoid metabolism are necessary. Obtaining a better understanding of the complex role of CYP-derived eicosanoids in inflammatory rheumatic disease, such as rheumatoid arthritis will provide valuable insight for basic and clinical researchers investigation.
Collapse
Affiliation(s)
- Malvina Hoxha
- Catholic University Our Lady of Good Counsel, Department of Chemical-Toxicological and Pharmacological Evaluation of Drugs, Rruga Dritan Hoxha, Tirana, Albania.
| | - Bruno Zappacosta
- Catholic University Our Lady of Good Counsel, Department of Chemical-Toxicological and Pharmacological Evaluation of Drugs, Rruga Dritan Hoxha, Tirana, Albania
| |
Collapse
|
47
|
Two pharmacological epoxyeicosatrienoic acid-enhancing therapies are effectively antihypertensive and reduce the severity of ischemic arrhythmias in rats with angiotensin II-dependent hypertension. J Hypertens 2019; 36:1326-1341. [PMID: 29570510 DOI: 10.1097/hjh.0000000000001708] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE We examined the effects of treatment with soluble epoxide hydrolase inhibitor (sEHi) and epoxyeicosatrienoic acids (EETs) analogue (EET-A), given alone or combined, on blood pressure (BP) and ischemia/reperfusion myocardial injury in rats with angiotensin II (ANG II)-dependent hypertension. METHODS Ren-2 transgenic rats (TGR) were used as a model of ANG II-dependent hypertension and Hannover Sprague-Dawley rats served as controls. Rats were treated for 14 days with sEHi or EET-A and BP was measured by radiotelemetry. Albuminuria, cardiac hypertrophy and concentrations of ANG II and EETs were determined. Separate groups were subjected to acute myocardial ischemia/reperfusion injury and the infarct size and ventricular arrhythmias were determined. RESULTS Treatment of TGR with sEHi and EET-A, given alone or combined, decreased BP to a similar degree, reduced albuminuria and cardiac hypertrophy to similar extent; only treatment regimens including sEHi increased myocardial and renal tissue concentrations of EETs. sEHi and EET-A, given alone or combined, suppressed kidney ANG II levels in TGR. Remarkably, infarct size did not significantly differ between TGR and Hannover Sprague-Dawley rats, but the incidence of ischemia-induced ventricular fibrillations was higher in TGR. Application of sEHi and EET-A given alone and combined sEHi and EET-A treatment were all equally effective in reducing life-threatening ventricular fibrillation in TGR. CONCLUSION The findings indicate that chronic treatment with either sEHi or EET-A exerts distinct antihypertensive and antiarrhythmic actions in our ANG II-dependent model of hypertension whereas combined administration of sEHi and EET-A does not provide additive antihypertensive or cardioprotective effects.
Collapse
|
48
|
Aiku AO, Marshall JM. Contribution of prostaglandins to exercise hyperaemia: workload, ethnicity and sex matter! J Physiol 2019; 597:4887-4900. [PMID: 31399992 DOI: 10.1113/jp278033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/06/2019] [Indexed: 01/03/2023] Open
Abstract
The contribution of prostaglandins (PGs) to exercise hyperaemia is controversial. In this review, we argue this is partly explained by differences in exercise intensity between studies. The effects of cyclooxygenase (COX) inhibition and PG assays indicate that PGs contribute more at moderate to heavy than at light workloads and are mainly released by low tissue O2 . But, the release and actions of PGs also depend on other O2 -dependent dilators including ATP, adenosine and NO. K+ may inhibit the action of PGs and other mediators by causing hyperpolarization, but contributes to the hyperaemia. Thus, at lighter loads, the influence of PGs may be blunted by K+ , while COX inhibition leads to compensatory increases in other O2 -dependent dilators. In addition, we show that other sources of variability are sex and ethnicity. Our findings indicate that exercise hyperaemia following rhythmic contractions at 60% maximum voluntary contraction, is smaller in young black African (BA) men and women than in their white European (WE) counterparts, but larger in men than in women of both ethnicities. We propose the larger absolute force in men causes greater vascular occlusion and accumulation of dilators, while blunted hyperaemia in BAs may reflect lower oxidative capacity and O2 requirement. Nevertheless, COX inhibition attenuated peak hyperaemia by ∼30% in WE, BA men and WE women, indicating PGs make a substantial contribution in all three groups. There was no effect in BA women. Lack of PG involvement may provide early evidence of endothelial dysfunction, consistent in BA women with their greater risk of cardiovascular disease.
Collapse
Affiliation(s)
- Abimbola O Aiku
- Institute of Clinical Sciences, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Janice M Marshall
- Institute of Clinical Sciences, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
49
|
Mori A, Takeda K, Sakamoto K, Nakahara T. Activation of transient receptor potential vanilloid 4 channels dilates rat retinal arterioles through nitric oxide- and BK Ca channel-dependent mechanisms in vivo. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:35-41. [PMID: 31392384 DOI: 10.1007/s00210-019-01707-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/26/2019] [Indexed: 01/19/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channel, a cation channel expressed in nearly all cell types, plays an important role in the regulation of vascular tone. In the present study, we examined the effect of GSK1016790A, an activator of TRPV4 channels, on the diameter of retinal blood vessels in rats and the underlying mechanisms. Ocular fundus images were captured with an original high-resolution digital fundus camera in vivo and diameters of retinal blood vessels were measured. Intravenous infusion of GSK1016790A (0.2-2 μg kg-1 min-1) increased retinal arteriolar diameter in a dose-dependent manner. The higher dose of GSK1016790A (2 μg kg-1 min-1) slightly decreased blood pressure. These responses to GSK1016790A were significantly attenuated by intravenous injection of GSK2193874 (0.3 mg/kg), an antagonist of TRPV4 channels. Intravitreal injection of Nω-nitro-L-arginine methyl ester, an inhibitor of nitric oxide (NO) synthase or iberiotoxin, an inhibitor of large-conductance Ca2+-activated K+ (BKCa) channel, significantly attenuated the GSK1016790A-induced increases in retinal arteriolar diameter. These results suggest that activation of TRPV4 channels dilates rat retinal arterioles through NO- and BKCa channel-dependent mechanisms in vivo.
Collapse
Affiliation(s)
- Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Kazuki Takeda
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| |
Collapse
|
50
|
Fleming I. New Lipid Mediators in Retinal Angiogenesis and Retinopathy. Front Pharmacol 2019; 10:739. [PMID: 31333461 PMCID: PMC6624440 DOI: 10.3389/fphar.2019.00739] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/07/2019] [Indexed: 12/31/2022] Open
Abstract
Retinal diseases associated with vascular destabilization and the inappropriate proliferation of retinal endothelial cells have major consequences on the retinal vascular network. In extreme cases, the development of hypoxia, the upregulation of growth factors, and the hyper-proliferation of unstable capillaries can result in bleeding and vision loss. While anti-vascular endothelial growth factor therapy and laser retinal photocoagulation can be used to treat the symptoms of late stage disease, there is currently no treatment available that can prevent disease progression. Cytochrome P450 enzymes metabolize endogenous substrates (polyunsaturated fatty acids) to bioactive fatty acid epoxides that demonstrate biological activity with generally protective/anti-inflammatory and insulin-sensitizing effects. These epoxides are further metabolized by the soluble epoxide hydrolase (sEH) to fatty acid diols, high concentrations of which have vascular destabilizing effects. Recent studies have identified increased sEH expression and activity and the subsequent generation of the docosahexaenoic acid-derived diol; 19,20-dihydroxydocosapentaenoic acid, as playing a major role in the development of diabetic retinopathy. This review summarizes current understanding of the roles of cytochrome P450 enzyme and sEH–derived PUFA mediators in retinal disease.
Collapse
Affiliation(s)
- Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK) partner site RheinMain, Frankfurt, Germany
| |
Collapse
|