1
|
Wang J, Yang F, Chen R, Yang X, Wang J, Zhang H. Hydrogel Composite Incorporating Deferoxamine-Loaded Gelatin-Based Microspheres Enhance Angiogenesis Ability of Dental Pulp Stem Cells. ACS OMEGA 2025; 10:12579-12589. [PMID: 40191326 PMCID: PMC11966253 DOI: 10.1021/acsomega.5c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
Fast reconstruction of the pulpal vasculature is crucial for effective pulp regeneration. Dental pulp stem cells (DPSCs) are promising candidates for pulp regeneration because of their potential for multilineage differentiation and vasculogenic properties. Deferoxamine (DFO) has been shown to stimulate angiogenesis during wound healing and bone regeneration; however, the effects of DFO on the angiogenic potential of DPSCs remain unknown. Moreover, its usefulness is restricted by a limited half-life and challenges in achieving localized tissue enrichment. This study aimed to develop a sustained-release injectable hydrogel composite as a drug delivery system and to investigate its influence on DPSCs. Herein, gelatin-based microspheres (GMSs) were loaded with DFO, and temperature-sensitive injectable hydrogels incorporating collagen and chitosan were synthesized to enable controlled DFO release. The experimental findings demonstrated that the DFO-loaded GMSs (DFO-GMSs) hydrogel composite possessed favorable physical properties and biocompatibility, enabling sustained DFO delivery for up to 15 days. DFO effectively stimulated DPSC migration, promoted the secretion of angiogenesis-related factors, and induced tube formation in vitro. These results suggest that the DFO-GMSs hydrogel composite significantly increased the migration and angiogenic potential of DPSCs, highlighting its promise for tissue regeneration applications.
Collapse
Affiliation(s)
- Jie Wang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Fan Yang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Ruting Chen
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
- Department
of Stomatology, Yangjiang People’s
Hospital, Affiliated Yangjiang Hospital of Guangdong Medical University, Yangjiang529500, China
| | - Xinyue Yang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Jingjing Wang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Hongyan Zhang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| |
Collapse
|
2
|
Choi IY, Kim YJ, Kim SY, Lee MK, Seol GH. Rb 1 restores palmitic acid-induced reduction of Ca 2+ influx by activating PLC in EA cells and PLD in MOVAS cells. Biomed Pharmacother 2025; 184:117927. [PMID: 39970733 DOI: 10.1016/j.biopha.2025.117927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/07/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025] Open
Abstract
Recent interest has focused on the role of Ca2+ in regulating health problems, including cardiovascular disease and colorectal cancer. The inverse correlation between colon cancer and serum Ca2+ underlines the importance of understanding intracellular Ca2+ dynamics. Studies are also evaluating the contributions of abnormalities in Ca²⁺ homeostasis and intracellular dysfunction to the pathogenesis of metabolic syndrome as a precursor of cardiovascular disease. In this study, we investigated the changes of Ca2+ dynamics and ginsenoside Rb1 (Rb1)-induced recovery in two vascular cell lines exposed to palmitic acid (PA), the most abundant active ingredient in palm oil. The mechanism underlying the Rb1-induced recovery was examined in a store operated Ca2+ entry model by Ca2+ store depletion. PA reduced the Ca2+ influx in both EA.hy926 (EA) and MOVAS cells, and this change was restored by Rb1. In EA cells, the Rb1-induced restoration was abolished by U73122 or 2-APB. In MOVAS cells, meanwhile, the effect of Rb1 was abolished by FIPI, U73122 and U73343. Under normal conditions, Rb1 itself altered phospholipid signaling (PLC in EA cells and PLD in MOVAS cells), but did not affect Ca2+ homeostasis. These differences resulted in differences in downstream actions, as KB-R7943 and nifedipine inhibited Rb1-mediated Ca2+ influx recovery only in MOVAS cells. In conclusion, Rb1 rescues the PA-induced Ca2+ influx by appropriately activating PLC in EA cells and PLD in MOVAS cells. This demonstrates that Ca2+ dynamics are elaborately regulated via intracellular Ca2+ signaling networks, suggesting a potential strategy for maintaining vascular Ca2+ homeostasis in hyperlipidemic environments.
Collapse
Affiliation(s)
- In-Young Choi
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Yoo Jin Kim
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea; BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul, Republic of Korea
| | - So Young Kim
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Min Kyung Lee
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea; BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Villadangos L, Serrador JM. Subcellular Localization Guides eNOS Function. Int J Mol Sci 2024; 25:13402. [PMID: 39769167 PMCID: PMC11678294 DOI: 10.3390/ijms252413402] [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: 11/04/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide synthases (NOS) are enzymes responsible for the cellular production of nitric oxide (NO), a highly reactive signaling molecule involved in important physiological and pathological processes. Given its remarkable capacity to diffuse across membranes, NO cannot be stored inside cells and thus requires multiple controlling mechanisms to regulate its biological functions. In particular, the regulation of endothelial nitric oxide synthase (eNOS) activity has been shown to be crucial in vascular homeostasis, primarily affecting cardiovascular disease and other pathophysiological processes of importance for human health. Among other factors, the subcellular localization of eNOS plays an important role in regulating its enzymatic activity and the bioavailability of NO. The aim of this review is to summarize pioneering studies and more recent publications, unveiling some of the factors that influence the subcellular compartmentalization of eNOS and discussing their functional implications in health and disease.
Collapse
Affiliation(s)
| | - Juan M. Serrador
- Interactions with the Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas (CSIC)—Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
4
|
Park J, Sahyoun C, Frangieh J, Réthoré L, Proux C, Grimaud L, Vessières E, Bourreau J, Mattei C, Henrion D, Marionneau C, Fajloun Z, Legendre C, Legros C. Veratridine Induces Vasorelaxation in Mouse Cecocolic Mesenteric Arteries. Toxins (Basel) 2024; 16:533. [PMID: 39728791 PMCID: PMC11679225 DOI: 10.3390/toxins16120533] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The vegetal alkaloid toxin veratridine (VTD) is a selective voltage-gated Na+ (NaV) channel activator, widely used as a pharmacological tool in vascular physiology. We have previously shown that NaV channels, expressed in arteries, contribute to vascular tone in mouse mesenteric arteries (MAs). Here, we aimed to better characterize the mechanisms of action of VTD using mouse cecocolic arteries (CAs), a model of resistance artery. Using wire myography, we found that VTD induced vasorelaxation in mouse CAs. This VTD-induced relaxation was insensitive to prazosin, an α1-adrenergic receptor antagonist, but abolished by atropine, a muscarinic receptor antagonist. Indeed, VTD-vasorelaxant effect was totally inhibited by the NaV channel blocker tetrodotoxin (0.3 µM), the NO synthase inhibitor L-NNA (20 µM), and low extracellular Na+ concentration (14.9 mM) and was partially blocked by the NCX1 antagonist SEA0400 (45.4% at 1 µM). Thus, we assumed that the VTD-induced vasorelaxation in CAs was due to acetylcholine release by parasympathetic neurons, which induced NO synthase activation mediated by the NCX1-Ca2+ entry mode in endothelial cells (ECs). We demonstrated NCX1 expression in ECs by RT-qPCR and immunohisto- and western immunolabelling. VTD did not induce an increase in intracellular Ca2+ ([Ca2+]i), while SEA0400 partially blocked acetylcholine-triggered [Ca2+]i elevations in Mile Sven 1 ECs. Altogether, these results illustrate that VTD activates NaV channels in parasympathetic neurons and then vasorelaxation in resistance arteries, which could explain arterial hypotension after VTD intoxication.
Collapse
Affiliation(s)
- Joohee Park
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Christina Sahyoun
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
- Laboratory of Applied Biotechnology (LBA3B), Department of Cell Culture, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon;
| | - Jacinthe Frangieh
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
- Laboratory of Applied Biotechnology (LBA3B), Department of Cell Culture, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon;
| | - Léa Réthoré
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Coralyne Proux
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Linda Grimaud
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Emilie Vessières
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Jennifer Bourreau
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - César Mattei
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Daniel Henrion
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Céline Marionneau
- Nantes Université, CNRS, INSERM, l’Institut du thorax, 44000 Nantes, France;
| | - Ziad Fajloun
- Laboratory of Applied Biotechnology (LBA3B), Department of Cell Culture, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon;
- Department of Biology, Faculty of Sciences 3, Campus Michel Slayman Ras Maska, Lebanese University, Tripoli 1352, Lebanon
| | - Claire Legendre
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| | - Christian Legros
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, 49000 Angers, France; (J.P.); (C.S.); (J.F.); (L.R.); (C.P.); (L.G.); (E.V.); (J.B.); (C.M.); (D.H.); (C.L.)
| |
Collapse
|
5
|
O’Hare N, Millican K, Ebong EE. Unraveling neurovascular mysteries: the role of endothelial glycocalyx dysfunction in Alzheimer's disease pathogenesis. Front Physiol 2024; 15:1394725. [PMID: 39027900 PMCID: PMC11254711 DOI: 10.3389/fphys.2024.1394725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
While cardiovascular disease, cancer, and human immunodeficiency virus (HIV) mortality rates have decreased over the past 20 years, Alzheimer's Disease (AD) deaths have risen by 145% since 2010. Despite significant research efforts, effective AD treatments remain elusive due to a poorly defined etiology and difficulty in targeting events that occur too downstream of disease onset. In hopes of elucidating alternative treatment pathways, now, AD is commonly being more broadly defined not only as a neurological disorder but also as a progression of a variety of cerebrovascular pathologies highlighted by the breakdown of the blood-brain barrier. The endothelial glycocalyx (GCX), which is an essential regulator of vascular physiology, plays a crucial role in the function of the neurovascular system, acting as an essential vascular mechanotransducer to facilitate ultimate blood-brain homeostasis. Shedding of the cerebrovascular GCX could be an early indication of neurovascular dysfunction and may subsequently progress neurodegenerative diseases like AD. Recent advances in in vitro modeling, gene/protein silencing, and imaging techniques offer new avenues of scrutinizing the GCX's effects on AD-related neurovascular pathology. Initial studies indicate GCX degradation in AD and other neurodegenerative diseases and have begun to demonstrate a possible link to GCX loss and cerebrovascular dysfunction. This review will scrutinize the GCX's contribution to known vascular etiologies of AD and propose future work aimed at continuing to uncover the relationship between GCX dysfunction and eventual AD-associated neurological deterioration.
Collapse
Affiliation(s)
- Nicholas O’Hare
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E. Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
6
|
Lee RJ, Adappa ND, Palmer JN. Akt activator SC79 stimulates antibacterial nitric oxide generation in human nasal epithelial cells in vitro. Int Forum Allergy Rhinol 2024; 14:1147-1162. [PMID: 38197521 PMCID: PMC11219270 DOI: 10.1002/alr.23318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND The role of Akt in nasal immunity is unstudied. Akt phosphorylates and activates endothelial nitric oxide synthase (eNOS) expressed in epithelial ciliated cells. Nitric oxide (NO) production by ciliated cells can have antibacterial and antiviral effects. Increasing nasal NO may be a useful antipathogen strategy in chronic rhinosinusitis (CRS). We previously showed that small-molecule Akt activator SC79 induces nasal cell NO production and suppresses IL-8 via the transcription factor Nrf-2. We hypothesized that SC79 NO production may additionally have antibacterial effects. METHODS NO production was measured using fluorescent dye DAF-FM. We tested effects of SC79 during co-culture of Pseudomonas aeruginosa with primary nasal epithelial cells, using CFU counting and live-dead staining to quantify bacterial killing. Pharmacology determined the mechanism of SC79-induced NO production and tested dependence on Akt. RESULTS SC79 induced dose-dependent, Akt-dependent NO production in nasal epithelial cells. The NO production required eNOS and Akt. The NO released into the airway surface liquid killed P. aeruginosa. No toxicity (LDH release) or inflammatory effects (IL8 transcription) were observed over 24 h. CONCLUSIONS Together, these data suggest multiple immune pathways are stimulated by SC79, with antipathogen effects. This in vitro pilot study suggests that a small-molecule Akt activator may have clinical utility in CRS or respiratory other infection settings, warranting future in vivo studies.
Collapse
Affiliation(s)
- Robert J. Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine
- Department of Physiology, University of Pennsylvania Perelman School of Medicine
| | - Nithin D. Adappa
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine
| | - James N. Palmer
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine
| |
Collapse
|
7
|
Lee RJ, Adappa ND, Palmer JN. Effects of Akt Activator SC79 on Human M0 Macrophage Phagocytosis and Cytokine Production. Cells 2024; 13:902. [PMID: 38891035 PMCID: PMC11171788 DOI: 10.3390/cells13110902] [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: 04/25/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Akt is an important kinase in metabolism. Akt also phosphorylates and activates endothelial and neuronal nitric oxide (NO) synthases (eNOS and nNOS, respectively) expressed in M0 (unpolarized) macrophages. We showed that e/nNOS NO production downstream of bitter taste receptors enhances macrophage phagocytosis. In airway epithelial cells, we also showed that the activation of Akt by a small molecule (SC79) enhances NO production and increases levels of nuclear Nrf2, which reduces IL-8 transcription during concomitant stimulation with Toll-like receptor (TLR) 5 agonist flagellin. We hypothesized that SC79's production of NO in macrophages might likewise enhance phagocytosis and reduce the transcription of some pro-inflammatory cytokines. Using live cell imaging of fluorescent biosensors and indicator dyes, we found that SC79 induces Akt activation, NO production, and downstream cGMP production in primary human M0 macrophages. This was accompanied by a reduction in IL-6, IL-8, and IL-12 production during concomitant stimulation with bacterial lipopolysaccharide, an agonist of pattern recognition receptors including TLR4. Pharmacological inhibitors suggested that this effect was dependent on Akt and Nrf2. Together, these data suggest that several macrophage immune pathways are regulated by SC79 via Akt. A small-molecule Akt activator may be useful in some infection settings, warranting future in vivo studies.
Collapse
Affiliation(s)
- Robert J. Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (N.D.A.); (J.N.P.)
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nithin D. Adappa
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (N.D.A.); (J.N.P.)
| | - James N. Palmer
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (N.D.A.); (J.N.P.)
| |
Collapse
|
8
|
Hong SG, Ashby JW, Kennelly JP, Wu M, Steel M, Chattopadhyay E, Foreman R, Tontonoz P, Tarling EJ, Turowski P, Gallagher-Jones M, Mack JJ. Mechanosensitive membrane domains regulate calcium entry in arterial endothelial cells to protect against inflammation. J Clin Invest 2024; 134:e175057. [PMID: 38771648 PMCID: PMC11213468 DOI: 10.1172/jci175057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/10/2024] [Indexed: 05/23/2024] Open
Abstract
Endothelial cells (ECs) in the descending aorta are exposed to high laminar shear stress, and this supports an antiinflammatory phenotype. High laminar shear stress also induces flow-aligned cell elongation and front-rear polarity, but whether these are required for the antiinflammatory phenotype is unclear. Here, we showed that caveolin-1-rich microdomains polarize to the downstream end of ECs that are exposed to continuous high laminar flow. These microdomains were characterized by high membrane rigidity, filamentous actin (F-actin), and raft-associated lipids. Transient receptor potential vanilloid (TRPV4) ion channels were ubiquitously expressed on the plasma membrane but mediated localized Ca2+ entry only at these microdomains where they physically interacted with clustered caveolin-1. These focal Ca2+ bursts activated endothelial nitric oxide synthase within the confines of these domains. Importantly, we found that signaling at these domains required both cell body elongation and sustained flow. Finally, TRPV4 signaling at these domains was necessary and sufficient to suppress inflammatory gene expression and exogenous activation of TRPV4 channels ameliorated the inflammatory response to stimuli both in vitro and in vivo. Our work revealed a polarized mechanosensitive signaling hub in arterial ECs that dampened inflammatory gene expression and promoted cell resilience.
Collapse
Affiliation(s)
- Soon-Gook Hong
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| | | | - John P. Kennelly
- Molecular Biology Institute
- Department of Pathology and Laboratory Medicine, and
| | - Meigan Wu
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| | | | | | - Rob Foreman
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, California, USA
| | - Peter Tontonoz
- Molecular Biology Institute
- Department of Pathology and Laboratory Medicine, and
| | | | - Patric Turowski
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Marcus Gallagher-Jones
- Correlated Imaging, Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, United Kingdom
| | - Julia J. Mack
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| |
Collapse
|
9
|
Abolfazli S, Mortazavi P, Kheirandish A, Butler AE, Jamialahmadi T, Sahebkar A. Regulatory effects of curcumin on nitric oxide signaling in the cardiovascular system. Nitric Oxide 2024; 143:16-28. [PMID: 38141926 DOI: 10.1016/j.niox.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
The continuously rising prevalence of cardiovascular disease (CVD) globally substantially impacts the economic growth of developing countries. Indeed, one of the leading causes of death worldwide is unfavorable cardiovascular events. Reduced nitric oxide (NO) generation is the pathogenic foundation of endothelial dysfunction, which is regarded as the first stage in the development of a number of CVDs. Nitric oxide exerts an array of biological effects, including vasodilation, the suppression of vascular smooth muscle cell proliferation and the functional control of cardiac cells. Numerous treatment strategies aim to increase NO synthesis or upregulate downstream NO signaling pathways. The major component of Curcuma longa, curcumin, has long been utilized in traditional medicine to treat various illnesses, especially CVDs. Curcumin improves CV function as well as having important pleiotropic effects, such as anti-inflammatory and antioxidant, through its ability to increase the bioavailability of NO and to positively impact NO-related signaling pathways. In this review, we discuss the scientific literature relating to curcumin's positive effects on NO signaling and vascular endothelial function.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Parham Mortazavi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Kheirandish
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, PO Box, 15503, Adliya, Bahrain
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Hong SG, Ashby JW, Kennelly JP, Wu M, Chattopadhyay E, Foreman R, Tontonoz P, Turowski P, Gallagher-Jones M, Mack JJ. Polarized Mechanosensitive Signaling Domains Protect Arterial Endothelial Cells Against Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542500. [PMID: 37292837 PMCID: PMC10246006 DOI: 10.1101/2023.05.26.542500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Endothelial cells (ECs) in the descending aorta are exposed to high laminar shear stress, which supports an anti-inflammatory phenotype that protects them from atherosclerosis. High laminar shear stress also supports flow-aligned cell elongation and front-rear polarity, but whether this is required for athero-protective signaling is unclear. Here, we show that Caveolin-1-rich microdomains become polarized at the downstream end of ECs exposed to continuous high laminar flow. These microdomains are characterized by higher membrane rigidity, filamentous actin (F-actin) and lipid accumulation. Transient receptor potential vanilloid-type 4 (Trpv4) ion channels, while ubiquitously expressed, mediate localized Ca 2+ entry at these microdomains where they physically interact with clustered Caveolin-1. The resultant focal bursts in Ca 2+ activate the anti-inflammatory factor endothelial nitric oxide synthase (eNOS) within the confines of these domains. Importantly, we find that signaling at these domains requires both cell body elongation and sustained flow. Finally, Trpv4 signaling at these domains is necessary and sufficient to suppress inflammatory gene expression. Our work reveals a novel polarized mechanosensitive signaling hub that induces an anti-inflammatory response in arterial ECs exposed to high laminar shear stress.
Collapse
|
11
|
Festa J, Hussain A, Al-Hareth Z, Singh H, Da Boit M. Anthocyanins and Vascular Health: A Matter of Metabolites. Foods 2023; 12:foods12091796. [PMID: 37174334 PMCID: PMC10178014 DOI: 10.3390/foods12091796] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Anthocyanins are a subgroup of flavonoid polyphenols previously investigated for improving cardiovascular health and preventing the development of endothelial dysfunction. However, their poor bioavailability raises the question of whether the observed biological activity is due to their metabolites. Phenolic metabolites can reach higher plasma concentrations and can persist in the circulation for periods much longer than their original anthocyanin form; therefore, the biological activity and health promoting effects of anthocyanins may differ from their metabolites. To address this, recent studies have facilitated different cell models, in vivo studies and explored physiologically relevant concentrations to better understand their mechanisms of action. The criteria were chosen based on previous reports demonstrating that anthocyanins can improve endothelial function via modulation of the Akt-endothelial nitric oxide synthase pathway and transcription factors Nrf2 and NF-κB, which made it critical to assess the phenolic metabolites' modes of action via these pathways. This review demonstrates how phenolic metabolites differ in bioactivity from their precursor anthocyanin, demonstrating improved endothelial function in response to inflammatory mediators at concentrations that are tolerated in vivo. The review highlights the crucial need for further studies to focus on improving the bioavailability of metabolites in isolation and explore the effect of metabolites in mixtures.
Collapse
Affiliation(s)
- Joseph Festa
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Aamir Hussain
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Zakia Al-Hareth
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
- Pandemic Sciences Institute, Old Road Campus, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
| | - Harprit Singh
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Mariasole Da Boit
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| |
Collapse
|
12
|
Modulation of Endothelial Function by TMAO, a Gut Microbiota-Derived Metabolite. Int J Mol Sci 2023; 24:ijms24065806. [PMID: 36982880 PMCID: PMC10054148 DOI: 10.3390/ijms24065806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Endothelial function is essential in the maintenance of systemic homeostasis, whose modulation strictly depends on the proper activity of tissue-specific angiocrine factors on the physiopathological mechanisms acting at both single and multi-organ levels. Several angiocrine factors take part in the vascular function itself by modulating vascular tone, inflammatory response, and thrombotic state. Recent evidence has outlined a strong relationship between endothelial factors and gut microbiota-derived molecules. In particular, the direct involvement of trimethylamine N-oxide (TMAO) in the development of endothelial dysfunction and its derived pathological outcomes, such as atherosclerosis, has come to light. Indeed, the role of TMAO in the modulation of factors strictly related to the development of endothelial dysfunction, such as nitric oxide, adhesion molecules (ICAM-1, VCAM-1, and selectins), and IL-6, has been widely accepted. The aim of this review is to present the latest studies that describe a direct role of TMAO in the modulation of angiocrine factors primarily involved in the development of vascular pathologies.
Collapse
|
13
|
Cho JM, Park SK, Kwon OS, Taylor La Salle D, Cerbie J, Fermoyle CC, Morgan D, Nelson A, Bledsoe A, Bharath LP, Tandar M, Kunapuli SP, Richardson RS, Anandh Babu PV, Mookherjee S, Kishore BK, Wang F, Yang T, Boudina S, Trinity JD, Symons JD. Activating P2Y1 receptors improves function in arteries with repressed autophagy. Cardiovasc Res 2023; 119:252-267. [PMID: 35420120 PMCID: PMC10236004 DOI: 10.1093/cvr/cvac061] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 02/19/2022] [Accepted: 03/23/2022] [Indexed: 11/14/2022] Open
Abstract
AIM The importance of endothelial cell (EC) autophagy to vascular homeostasis in the context of health and disease is evolving. Earlier, we reported that intact EC autophagy is requisite to maintain shear-stress-induced nitric oxide (NO) generation via glycolysis-dependent purinergic signalling to endothelial NO synthase (eNOS). Here, we illustrate the translational and functional significance of these findings. METHODS AND RESULTS First, we assessed translational relevance using older male humans and mice that exhibit blunted EC autophagy and impaired arterial function vs. adult controls. Active hyperaemia evoked by rhythmic handgrip exercise-elevated radial artery shear-rate similarly from baseline in adult and older subjects for 60 min. Compared with baseline, indexes of autophagy initiation, p-eNOSS1177 activation, and NO generation, occurred in radial artery ECs obtained from adult but not older volunteers. Regarding mice, indexes of autophagy and p-eNOSS1177 activation were robust in ECs from adult but not older animals that completed 60-min treadmill-running. Furthermore, 20 dyne • cm2 laminar shear stress × 45-min increased autophagic flux, glycolysis, ATP production, and p-eNOSS1177 in primary arterial ECs obtained from adult but not older mice. Concerning functional relevance, we next questioned whether the inability to initiate EC autophagy, glycolysis, and p-eNOSS1177in vitro precipitates arterial dysfunction ex vivo. Compromised intraluminal flow-mediated vasodilation displayed by arteries from older vs. adult mice was recapitulated in vessels from adult mice by (i) NO synthase inhibition; (ii) acute autophagy impairment using 3-methyladenine (3-MA); (iii) EC Atg3 depletion (iecAtg3KO mice); (iv) purinergic 2Y1-receptor (P2Y1-R) blockade; and (v) germline depletion of P2Y1-Rs. Importantly, P2Y1-R activation using 2-methylthio-ADP (2-Me-ADP) improved vasodilatory capacity in arteries from (i) adult mice treated with 3-MA; (ii) adult iecAtg3KO mice; and (iii) older animals with repressed EC autophagy. CONCLUSIONS Arterial dysfunction concurrent with pharmacological, genetic, and age-associated EC autophagy compromise is improved by activating P2Y1-Rs.
Collapse
Affiliation(s)
- Jae Min Cho
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Seul-Ki Park
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery & Center on Aging, University of Connecticut School of Medicine, Storrs, CT, USA
| | - David Taylor La Salle
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - James Cerbie
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Caitlin C Fermoyle
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - David Morgan
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Ashley Nelson
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Amber Bledsoe
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Leena P Bharath
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Megan Tandar
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Russell S Richardson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | | | - Sohom Mookherjee
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Bellamkonda K Kishore
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Nephrology Research, George E. Whalen VA Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA
| | - Fei Wang
- Nephrology Research, George E. Whalen VA Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA
| | - Tianxin Yang
- Nephrology Research, George E. Whalen VA Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joel D Trinity
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - John David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Role of Vitamin D Deficiency in the Pathogenesis of Cardiovascular and Cerebrovascular Diseases. Nutrients 2023; 15:nu15020334. [PMID: 36678205 PMCID: PMC9864832 DOI: 10.3390/nu15020334] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/12/2023] Open
Abstract
Deficiency in vitamin D (VitD), a lipid-soluble vitamin and steroid hormone, affects approximately 24% to 40% of the population of the Western world. In addition to its well-documented effects on the musculoskeletal system, VitD also contributes importantly to the promotion and preservation of cardiovascular health via modulating the immune and inflammatory functions and regulating cell proliferation and migration, endothelial function, renin expression, and extracellular matrix homeostasis. This brief overview focuses on the cardiovascular and cerebrovascular effects of VitD and the cellular, molecular, and functional changes that occur in the circulatory system in VitD deficiency (VDD). It explores the links among VDD and adverse vascular remodeling, endothelial dysfunction, vascular inflammation, and increased risk for cardiovascular and cerebrovascular diseases. Improved understanding of the complex role of VDD in the pathogenesis of atherosclerotic cardiovascular diseases, stroke, and vascular cognitive impairment is crucial for all cardiologists, dietitians, and geriatricians, as VDD presents an easy target for intervention.
Collapse
|
15
|
Bandala C, Cárdenas-Rodríguez N, Reyes-Long S, Cortés-Algara A, Contreras-García IJ, Cruz-Hernández TR, Alfaro-Rodriguez A, Cortes-Altamirano JL, Perez-Santos M, Anaya-Ruiz M, Lara-Padilla E. Estrogens as a Possible Therapeutic Strategy for the Management of Neuroinflammation and Neuroprotection in COVID-19. Curr Neuropharmacol 2023; 21:2110-2125. [PMID: 37326113 PMCID: PMC10556364 DOI: 10.2174/1570159x21666230616103850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 06/17/2023] Open
Abstract
The Coronavirus disease 2019 (COVID-19) affects several tissues, including the central and peripheral nervous system. It has also been related to signs and symptoms that suggest neuroinflammation with possible effects in the short, medium, and long term. Estrogens could have a positive impact on the management of the disease, not only due to its already known immunomodulator effect, but also activating other pathways that may be important in the pathophysiology of COVID-19, such as the regulation of the virus receptor and its metabolites. In addition, they can have a positive effect on neuroinflammation secondary to pathologies other than COVID-19. The aim of this study is to analyze the molecular mechanisms that link estrogens with their possible therapeutic effect for neuroinflammation related to COVID-19. Advanced searches were performed in scientific databases as Pub- Med, ProQuest, EBSCO, the Science Citation index, and clinical trials. Estrogens have been shown to participate in the immune modulation of the response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to this mechanism, we propose that estrogens can regulate the expression and activity of the Angiotensin-converting enzyme 2 (ACE2), reestablishing its cytoprotective function, which may be limited by its interaction with SARS-CoV-2. In this proposal, estrogens and estrogenic compounds could increase the synthesis of Angiotensin-(1-7) (Ang-(1-7)) that acts through the Mas receptor (MasR) in cells that are being attacked by the virus. Estrogens can be a promising, accessible, and low-cost treatment for neuroprotection and neuroinflammation in patients with COVID-19, due to its direct immunomodulatory capacity in decreasing cytokine storm and increasing cytoprotective capacity of the axis ACE2/Ang (1-7)/MasR.
Collapse
Affiliation(s)
- Cindy Bandala
- Higher School of Medicine, National Polytechnic Institute, Mexico City, 11340, Mexico
| | - Noemí Cárdenas-Rodríguez
- Higher School of Medicine, National Polytechnic Institute, Mexico City, 11340, Mexico
- Neuroscience Laboratory, National Institute of Pediatrics, Mexico City, 04530, Mexico
| | - Samuel Reyes-Long
- Basic Neurosciences, National Institute of Rehabilitation LGII, Mexico City, 14389, Mexico
| | - Alfredo Cortés-Algara
- Higher School of Medicine, National Polytechnic Institute, Mexico City, 11340, Mexico
- Department of Robotic Surgery and Laparoscopy in Gynecology, Centro Médico Nacional 20 de Noviembre, Mexico City, CP, Mexico
| | | | | | | | - José Luis Cortes-Altamirano
- Basic Neurosciences, National Institute of Rehabilitation LGII, Mexico City, 14389, Mexico
- Research Department, Ecatepec Valley State University, Valle de Anahuac, Ecatepec, 55210, Mexico State, Mexico
| | - Martín Perez-Santos
- Directorate of Innovation and Knowledge Transfer, Meritorious Autonomous University of Puebla, 72570, Puebla
| | - Maricruz Anaya-Ruiz
- Cell Biology Laboratory, Oriente Biomedical Research Center, Mexican Social Security Institute, Metepec, 74360, Puebla
| | - Eleazar Lara-Padilla
- Higher School of Medicine, National Polytechnic Institute, Mexico City, 11340, Mexico
| |
Collapse
|
16
|
mTOR contributes to endothelium-dependent vasorelaxation by promoting eNOS expression and preventing eNOS uncoupling. Commun Biol 2022; 5:726. [PMID: 35869262 PMCID: PMC9307829 DOI: 10.1038/s42003-022-03653-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/24/2022] [Indexed: 11/08/2022] Open
Abstract
Clinically used inhibitors of mammalian target of rapamycin (mTOR) negatively impacts endothelial-dependent vasodilatation (EDD) through unidentified mechanisms. Here we show that either the endothelium-specific deletion of Mtor to inhibit both mTOR complexes, or depletion of Raptor or Rictor to disrupt mTORC1 or mTORC2, causes impaired EDD, accompanied by reduced NO in the serum of mice. Consistently, inhibition of mTOR decreases NO production by human and mouse EC. Specifically, inhibition of mTORC1 suppresses eNOS gene expression, due to impairment in p70S6K-mediated posttranscriptional regulation of the transcription factor KLF2 expression. In contrast to mTORC1 inhibition, a positive-feedback between MAPK (p38 and JNK) activation and Nox2 upregulation contributes to the excessive generation of reactive oxygen species (ROS), which causes eNOS uncoupling and decreased NO bioavailability in mTORC2-inhibited EC. Adeno-associated virus-mediated EC-specific overexpression of KLF2 or suppression of Nox2 restores EDD function in endothelial mTORC1- or mTORC2-inhibited mice. The endothelium-specific inhibition of either of mammalian target of rapamycin (mTOR) complexes impairs endothelial-dependent vasodilatation (EDD), accompanied by decreased nitric oxide bioavailability in both human and mice endothelial cells.
Collapse
|
17
|
Kotlyarov S, Kotlyarova A. The Importance of the Plasma Membrane in Atherogenesis. MEMBRANES 2022; 12:1036. [PMID: 36363591 PMCID: PMC9698587 DOI: 10.3390/membranes12111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Atherosclerotic cardiovascular diseases are an important medical problem due to their high prevalence, impact on quality of life and prognosis. The pathogenesis of atherosclerosis is an urgent medical and social problem, the solution of which may improve the quality of diagnosis and treatment of patients. Atherosclerosis is a complex chain of events, which proceeds over many years and in which many cells in the bloodstream and the vascular wall are involved. A growing body of evidence suggests that there are complex, closely linked molecular mechanisms that occur in the plasma membranes of cells involved in atherogenesis. Lipid transport, innate immune system receptor function, and hemodynamic regulation are linked to plasma membranes and their biophysical properties. A better understanding of these interrelationships will improve diagnostic quality and treatment efficacy.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
18
|
Wenninger N, Bernhart C, Kappaun W, Kollau A, Kalcher K, Ortner A. High-performance amperometric determination of nitric oxide released by endothelial cells using flow injection analysis. Talanta 2022. [DOI: 10.1016/j.talanta.2022.123810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
19
|
Whitehead AK, Fried ND, Li Z, Neelamegam K, Pearson CS, LaPenna KB, Sharp TE, Lefer DJ, Lazartigues E, Gardner JD, Yue X. Alpha7 nicotinic acetylcholine receptor mediates chronic nicotine inhalation-induced cardiopulmonary dysfunction. Clin Sci (Lond) 2022; 136:973-987. [PMID: 35678315 PMCID: PMC10199464 DOI: 10.1042/cs20220083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/27/2022] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Cigarette smoking remains the leading modifiable risk factor for cardiopulmonary diseases; however, the effects of nicotine alone on cardiopulmonary function remain largely unknown. Previously, we have shown that chronic nicotine vapor inhalation in mice leads to the development of pulmonary hypertension (PH) with right ventricular (RV) remodeling. The present study aims to further examine the cardiopulmonary effects of nicotine and the role of the α7 nicotinic acetylcholine receptor (α7-nAChR), which is widely expressed in the cardiovascular system. Wild-type (WT) and α7-nAChR knockout (α7-nAChR-/-) mice were exposed to room air (control) or nicotine vapor daily for 12 weeks. Consistent with our previous study, echocardiography and RV catheterization reveal that male WT mice developed increased RV systolic pressure with RV hypertrophy and dilatation following 12-week nicotine vapor exposure; in contrast, these changes were not observed in male α7-nAChR-/- mice. In addition, chronic nicotine inhalation failed to induce PH and RV remodeling in female mice regardless of genotype. The effects of nicotine on the vasculature were further examined in male mice. Our results show that chronic nicotine inhalation led to impaired acetylcholine-mediated vasodilatory response in both thoracic aortas and pulmonary arteries, and these effects were accompanied by altered endothelial nitric oxide synthase phosphorylation (enhanced inhibitory phosphorylation at threonine 495) and reduced plasma nitrite levels in WT but not α7-nAChR-/- mice. Finally, RNA sequencing revealed up-regulation of multiple inflammatory pathways in thoracic aortas from WT but not α7-nAChR-/- mice. We conclude that the α7-nAChR mediates chronic nicotine inhalation-induced PH, RV remodeling and vascular dysfunction.
Collapse
Affiliation(s)
- Anna K. Whitehead
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Nicholas D. Fried
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Zhen Li
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Kandasamy Neelamegam
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Charlotte S. Pearson
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Kyle B. LaPenna
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Thomas E. Sharp
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Department of Medicine Section of Cardiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - David J. Lefer
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Southeast Louisiana Veterans Health Care Systems, New Orleans, LA 70119, U.S.A
| | - Jason D. Gardner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| |
Collapse
|
20
|
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
|
21
|
Kemps H, Dessy C, Dumas L, Sonveaux P, Alders L, Van Broeckhoven J, Font LP, Lambrichts S, Foulquier S, Hendrix S, Brône B, Lemmens R, Bronckaers A. Extremely low frequency electromagnetic stimulation reduces ischemic stroke volume by improving cerebral collateral blood flow. J Cereb Blood Flow Metab 2022; 42:979-996. [PMID: 35209740 PMCID: PMC9125494 DOI: 10.1177/0271678x221084410] [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] [Indexed: 11/16/2022]
Abstract
Extremely low frequency electromagnetic stimulation (ELF-EMS) has been considered as a neuroprotective therapy for ischemic stroke based on its capacity to induce nitric oxide (NO) signaling. Here, we examined whether ELF-EMS reduces ischemic stroke volume by stimulating cerebral collateral perfusion. Moreover, the pathway responsible for ELF-EMS-induced NO production was investigated. ELF-EMS diminished infarct growth following experimental stroke in collateral-rich C57BL/6 mice, but not in collateral-scarce BALB/c mice, suggesting that decreased lesion sizes after ELF-EMS results from improved collateral blood flow. In vitro analysis demonstrated that ELF-EMS increased endothelial NO levels by stimulating the Akt-/eNOS pathway. Furthermore, ELF-EMS augmented perfusion in the hind limb of healthy mice, which was mediated by enhanced Akt-/eNOS signaling. In healthy C57BL/6 mouse brains, ELF-EMS treatment increased cerebral blood flow in a NOS-dependent manner, whereas no improvement in cerebrovascular perfusion was observed in collateral-sparse BALB/c mice. In addition, ELF-EMS enhanced cerebral blood flow in both the contra- and ipsilateral hemispheres of C57BL/6 mice subjected to experimental ischemic stroke. In conclusion, we showed that ELF-EMS enhances (cerebro)vascular perfusion by stimulating NO production, indicating that ELF-EMS could be an attractive therapeutic strategy for acute ischemic stroke by improving cerebral collateral blood flow.
Collapse
Affiliation(s)
- Hannelore Kemps
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurent Dumas
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Lotte Alders
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Jana Van Broeckhoven
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Lena Perez Font
- Centro Nacional de Electromagnetismo Aplicado (CNEA), Universidad de Oriente, Santiago de Cuba, Cuba
| | - Sara Lambrichts
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands.,CARIM, School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Sven Hendrix
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium.,Medical School Hamburg, Hamburg, Germany
| | - Bert Brône
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Robin Lemmens
- KU Leuven, - University of Leuven, Department of Neurosciences, Experimental Neurology, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| |
Collapse
|
22
|
Carey RM, Hariri BM, Adappa ND, Palmer JN, Lee RJ. HSP90 Modulates T2R Bitter Taste Receptor Nitric Oxide Production and Innate Immune Responses in Human Airway Epithelial Cells and Macrophages. Cells 2022; 11:1478. [PMID: 35563784 PMCID: PMC9101439 DOI: 10.3390/cells11091478] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Bitter taste receptors (T2Rs) are G protein-coupled receptors (GPCRs) expressed in various cell types including ciliated airway epithelial cells and macrophages. T2Rs in these two innate immune cell types are activated by bitter products, including those secreted by Pseudomonas aeruginosa, leading to Ca2+-dependent activation of endothelial nitric oxide (NO) synthase (eNOS). NO enhances mucociliary clearance and has direct antibacterial effects in ciliated epithelial cells. NO also increases phagocytosis by macrophages. Using biochemistry and live-cell imaging, we explored the role of heat shock protein 90 (HSP90) in regulating T2R-dependent NO pathways in primary sinonasal epithelial cells, primary monocyte-derived macrophages, and a human bronchiolar cell line (H441). Immunofluorescence showed that H441 cells express eNOS and T2Rs and that the bitter agonist denatonium benzoate activates NO production in a Ca2+- and HSP90-dependent manner in cells grown either as submerged cultures or at the air-liquid interface. In primary sinonasal epithelial cells, we determined that HSP90 inhibition reduces T2R-stimulated NO production and ciliary beating, which likely limits pathogen clearance. In primary monocyte-derived macrophages, we found that HSP-90 is integral to T2R-stimulated NO production and phagocytosis of FITC-labeled Escherichia coli and pHrodo-Staphylococcus aureus. Our study demonstrates that HSP90 serves as an innate immune modulator by regulating NO production downstream of T2R signaling by augmenting eNOS activation without impairing upstream Ca2+ signaling. These findings suggest that HSP90 plays an important role in airway antibacterial innate immunity and may be an important target in airway diseases such as chronic rhinosinusitis, asthma, or cystic fibrosis.
Collapse
Affiliation(s)
- Ryan M. Carey
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - Benjamin M. Hariri
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - Nithin D. Adappa
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - James N. Palmer
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - Robert J. Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
23
|
Trimethylamine N-Oxide (TMAO) Impairs Purinergic Induced Intracellular Calcium Increase and Nitric Oxide Release in Endothelial Cells. Int J Mol Sci 2022; 23:ijms23073982. [PMID: 35409341 PMCID: PMC8999849 DOI: 10.3390/ijms23073982] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
Trimethylamine N-oxide (TMAO) is a diet derived compound directly introduced through foodstuff, or endogenously synthesized from its precursors, primarily choline, L-carnitine, and ergothioneine. New evidence outlines high TMAO plasma concentrations in patients with overt cardiovascular disease, but its direct role in pathological development is still controversial. The purpose of the study was to evaluate the role of TMAO in affecting key intracellular factors involved in endothelial dysfunction development, such as reactive oxygen species, mitochondrial health, calcium balance, and nitric oxide release using bovine aortic endothelial cells (BAE-1). Cell viability and oxidative stress indicators were monitored after acute and prolonged TMAO treatment. The role of TMAO in interfering with the physiological purinergic vasodilatory mechanism after ATP stimulation was defined through measurements of the rise of intracellular calcium, nitric oxide release, and eNOS phosphorylation at Ser1179 (eNOSSer1179). TMAO was not cytotoxic for BAE-1 and it did not induce the rise of reactive oxygen species and impairment of mitochondrial membrane potential, either in the basal condition or in the presence of a stressor. In contrast, TMAO modified the purinergic response affecting intracellular ATP-induced calcium increase, nitric oxide release, and eNOSSer1179. Results obtained suggest a possible implication of TMAO in impairing the endothelial-dependent vasodilatory mechanism.
Collapse
|
24
|
Therapeutic Potential of Seaweed-Derived Bioactive Compounds for Cardiovascular Disease Treatment. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cardiovascular diseases are closely related to hypertension, type 2 diabetes mellitus, obesity, and hyperlipidemia. Many studies have reported that an unhealthy diet and sedentary lifestyle are critical factors that enhance these diseases. Recently, many bioactive compounds isolated from marine seaweeds have been studied for their benefits in improving human health. In particular, several unique bioactive metabolites such as polyphenols, polysaccharides, peptides, carotene, and sterol are the most effective components responsible for these activities. This review summarizes the current in vitro, in vivo, and clinical studies related to the protective effects of bioactive compounds isolated from seaweeds against cardiovascular disorders, including anti-diabetic, anti-hypertensive, anti-hyperlipidemia, and anti-obesity effects. Therefore, this present review summarizes these concepts and provides a basis for further in-depth research.
Collapse
|
25
|
Ferulic Acid Alleviates Atherosclerotic Plaques by Inhibiting VSMC Proliferation Through the NO/p21 Signaling pathway. J Cardiovasc Transl Res 2022; 15:865-875. [PMID: 34993756 PMCID: PMC9622559 DOI: 10.1007/s12265-021-10196-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/13/2021] [Indexed: 11/21/2022]
Abstract
The benefits and risks of inhibiting the proliferation and migration of vascular smooth muscle cells (VSMCs) in atherosclerosis (AS) remain a subject of debate. In this study, we investigated the effect of ferulic acid (FA) on the proliferation and migration of VSMCs induced by platelet-derived growth factor (PDGF) and the associated mechanism and used ApoE-/- mice to study whether the effect of FA on VSMC proliferation and migration is beneficial in alleviating AS plaques. It was found that FA not only reduced blood lipid levels but also promoted the production of nitric oxide (NO) by MOVAS cells through the endothelial nitric oxide synthase (eNOS) pathway, inhibited the migration and proliferation of VSMCs induced by PDGF, promoted the expression of p21 in VSMCs, and exerted a therapeutic effect against AS.
Collapse
|
26
|
Zheng Y, Wang J, Zhao T, Wang L, Wang J. Modulation of the VEGF/AKT/eNOS signaling pathway to regulate liver angiogenesis to explore the anti-hepatic fibrosis mechanism of curcumol. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114480. [PMID: 34358654 DOI: 10.1016/j.jep.2021.114480] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/20/2021] [Accepted: 07/30/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcuma is a common Chinese herbal medicine that is used in the clinical treatment of chronic liver disease. Studies have found that curcumol is the main active ingredient of curcuma and has good hepatoprotective and anti-inflammatory effects. However, there are few reports on the molecular mechanism underlying the anti-liver fibrosis effect of curcumol. AIM To explore the effect of curcumol on liver angiogenesis, and to reveal the mechanism of curcumol against liver fibrosis. MATERIALS AND METHODS We used liver collagenase perfusion combined with Percoll density gradient sedimentation to separate primary liver sinusoidal endothelial cells, and then applied a leptin-activated cell pathological model. The cells were divided into four treatment groups as follows: blank group, model group, curcumol group, and solafini group. MTT was used to detect the cell proliferation rate in each group, and RT-PCR and western blotting were used to detect the expressions of VEGF, AKT, eNOS, CD31, and vWF. A fluorescent probe was used to detect NO expression, and scanning electron microscopy was used to observe changes in the cell fenestration structure. Angiogenesis assays were used to observe blood vessel formation in each group. RESULTS The results of the MTT test found that the proliferation rate of each group was higher. The results of the molecular biology tests found that curcumol inhibited the activity of the VEGF/AKT/eNOS pathway, thereby increasing fenestration of sinusoidal endothelial cells and inhibiting liver angiogenesis. These differences were statistically significant compared with the model group. CONCLUSIONS Curcumol inhibits the activity of the VEGF/AKT/eNOS signaling pathway, regulates the structure of hepatic sinusoidal endothelial cells, and inhibits liver angiogenesis, which together may explain its anti-liver fibrosis mechanism.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese, Medicine, Nanning, Guangxi, 530222, China
| | - Jiaru Wang
- College of Nursing,Guangdong Medical University, Dongguan, Guangdong, 523000, China
| | - Tiejian Zhao
- Department of Physiology, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Lei Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese, Medicine, Nanning, Guangxi, 530222, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese, Medicine, Nanning, Guangxi, 530222, China.
| |
Collapse
|
27
|
Malikova E, Kmecova Z, Doka G, Pivackova LB, Balis P, Trubacova S, Velasova E, Krenek P, Klimas J. Pioglitazone restores phosphorylation of downregulated caveolin-1 in right ventricle of monocrotaline-induced pulmonary hypertension. Clin Exp Hypertens 2021; 44:101-112. [PMID: 34747283 DOI: 10.1080/10641963.2021.1996589] [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] [Indexed: 10/25/2022]
Abstract
BACKGROUND Caveolin-1 (cav-1) plays a role in pulmonary arterial hypertension (PAH). Monocrotaline (MCT)-induced PAH is characterized by a loss of cav-1 in pulmonary arteries; however, less is known regarding its role in the hypertrophied right ventricle (RV). We aimed to characterize the role of cav-1 and Hsp90 in the RV of MCT-induced PAH and their impact on endothelial nitric oxide synthase (eNOS). Additionally, we focused on restoration of cav-1 expression with pioglitazone administration. METHODS Male 12-week-old Wistar rats were injected subcutaneously with monocrotaline (60 mg/kg). Selected proteins (cav-1, eNOS, pSer1177eNOS, Hsp90) and mRNAs (cav-1α, cav-1β, eNOS) were determined in the RV and left ventricle (LV) 4 weeks later. In a separate MCT-induced PAH study, pioglitazone (10 mg/kg/d, orally) administration started on day 14 after MCT. RESULTS MCT induced RV hypertrophy and lung enlargement. Cav-1 and pTyr14cav-1 were decreased in RV. Caveolin-1α (cav-1α) and caveolin-1β (cav-1β) mRNAs were decreased in both ventricles. Hsp90 protein was increased in RV. eNOS and pSer1177eNOS proteins were unchanged in the ventricles. eNOS mRNA was reduced in RV. Pioglitazone treatment increased oxygen saturation and pTyr14cav-1 vs. MCT group. CONCLUSIONS Restoration of pTyr14cav-1 did not lead to amelioration of the disease, nor did it prevent RV hypertrophy and fibrosis, which was indicated by an increase in Acta2, Nppb, Col3a1, and Tgfβ1 mRNA.
Collapse
Affiliation(s)
- Eva Malikova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| | - Zuzana Kmecova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| | - Gabriel Doka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| | - Lenka Bies Pivackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| | - Peter Balis
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute of Normal and Pathological Physiology, Bratislava, Slovakia
| | - Simona Trubacova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Eva Velasova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovakia
| |
Collapse
|
28
|
Kotlyarov S. Diversity of Lipid Function in Atherogenesis: A Focus on Endothelial Mechanobiology. Int J Mol Sci 2021; 22:11545. [PMID: 34768974 PMCID: PMC8584259 DOI: 10.3390/ijms222111545] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the most important problems in modern medicine. Its high prevalence and social significance determine the need for a better understanding of the mechanisms of the disease's development and progression. Lipid metabolism and its disorders are one of the key links in the pathogenesis of atherosclerosis. Lipids are involved in many processes, including those related to the mechanoreception of endothelial cells. The multifaceted role of lipids in endothelial mechanobiology and mechanisms of atherogenesis are discussed in this review. Endothelium is involved in ensuring adequate vascular hemodynamics, and changes in blood flow characteristics are detected by endothelial cells and affect their structure and function.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
29
|
Troy AM, Cheng HM. Human microvascular reactivity: a review of vasomodulating stimuli and non-invasive imaging assessment. Physiol Meas 2021; 42. [PMID: 34325417 DOI: 10.1088/1361-6579/ac18fd] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/29/2021] [Indexed: 11/11/2022]
Abstract
The microvasculature serves an imperative function in regulating perfusion and nutrient exchange throughout the body, adaptively altering blood flow to preserve hemodynamic and metabolic homeostasis. Its normal functioning is vital to tissue health, whereas its dysfunction is present in many chronic conditions, including diabetes, heart disease, and cognitive decline. As microvascular dysfunction often appears early in disease progression, its detection can offer early diagnostic information. To detect microvascular dysfunction, one uses imaging to probe the microvasculature's ability to react to a stimulus, also known as microvascular reactivity (MVR). An assessment of MVR requires an integrated understanding of vascular physiology, techniques for stimulating reactivity, and available imaging methods to capture the dynamic response. Practical considerations, including compatibility between the selected stimulus and imaging approach, likewise require attention. In this review, we provide a comprehensive foundation necessary for informed imaging of MVR, with a particular focus on the challenging endeavor of assessing microvascular function in deep tissues.
Collapse
Affiliation(s)
- Aaron M Troy
- Institute of Biomedical Engineering, University of Toronto, Toronto, CANADA
| | | |
Collapse
|
30
|
Li H, Nguyen H, Meda Venkata SP, Koh JY, Kowluru A, Li L, Rossi NF, Chen W, Wang JM. Novel Role of GPR35 (G-Protein-Coupled Receptor 35) in the Regulation of Endothelial Cell Function and Blood Pressure. Hypertension 2021; 78:816-830. [PMID: 34275335 PMCID: PMC8357038 DOI: 10.1161/hypertensionaha.120.15423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. GPR35 (G-protein–coupled receptor 35) is a poorly characterized receptor that has garnered increased interest as a therapeutic target through its implications in a range of inflammatory and cardiovascular diseases, but its biological functions stay largely unknown. The current study evaluated the effect of GPR35 on endothelial cell (EC) functions and hemodynamic homeostasis. In primary human aortic ECs, the expression of GPR35 was manipulated by transfections of adenovirus carrying either GPR35 cDNA or shRNA against GPR35, using adenovirus carrying β-gal as control. Mouse aortic ECs were isolated and cultured from GPR35 knockout and wild-type control mice. Our results indicated that genetic inhibition of GPR35 in human and mouse ECs significantly promoted cell proliferation, migration, and tube formation in vitro. The GCH1 (guanosine triphosphate cyclohydrolase I)-mediated biosynthesis of tetrahydrobiopterin was enhanced, reducing intracellular superoxide. Knocking down GCH1 or eNOS (endothelial nitric oxide synthase) significantly blunted the robust angiogenesis induced by GPR35 suppression. Male GPR35 knockout mice demonstrated reduced basal arterial blood pressure and an attenuated onset of hypertension in deoxycorticosterone acetate-salt induced hypertensive model compared with male GPR35 wild-type control mice in vivo, with concomitant improved endothelium-dependent vasodilation and decreased superoxide in isolated aortas. The difference in arterial blood pressure was absent between female GPR35 wild-type control and female GPR35 knockout mice. Our study provides novel insights into the roles of GPR35 in endothelial function and vascular tone modulation that critically contribute to the pathophysiology of blood pressure elevation. Antagonizing GPR35 activity might represent a potentially effective therapeutic approach to restore EC function and hemodynamic homeostasis.
Collapse
Affiliation(s)
- Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (H.L., H.N., S.P.M.V., J.Y.K., A.K., J.-M.W.), Wayne State University, Detroit, MI
| | - Huong Nguyen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (H.L., H.N., S.P.M.V., J.Y.K., A.K., J.-M.W.), Wayne State University, Detroit, MI
| | - Sai Pranathi Meda Venkata
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (H.L., H.N., S.P.M.V., J.Y.K., A.K., J.-M.W.), Wayne State University, Detroit, MI
| | - Jia Yi Koh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (H.L., H.N., S.P.M.V., J.Y.K., A.K., J.-M.W.), Wayne State University, Detroit, MI
| | - Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (H.L., H.N., S.P.M.V., J.Y.K., A.K., J.-M.W.), Wayne State University, Detroit, MI.,John D. Dingell VA Medical Center, Detroit, MI (A.K., N.F.R.)
| | - Li Li
- Departments of Internal Medicine (L.L., N.F.R.), Wayne State University, Detroit, MI
| | - Noreen F Rossi
- Departments of Internal Medicine (L.L., N.F.R.), Wayne State University, Detroit, MI.,John D. Dingell VA Medical Center, Detroit, MI (A.K., N.F.R.)
| | - Wei Chen
- Department of Oncology (W.C.), Wayne State University, Detroit, MI.,School of Medicine, Karmanos Cancer Institute (W.C.), Wayne State University, Detroit, MI
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (H.L., H.N., S.P.M.V., J.Y.K., A.K., J.-M.W.), Wayne State University, Detroit, MI.,Centers for Molecular Medicine and Genetics (J.-M.W.), Wayne State University, Detroit, MI
| |
Collapse
|
31
|
Janaszak-Jasiecka A, Siekierzycka A, Płoska A, Dobrucki IT, Kalinowski L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021; 11:biom11070982. [PMID: 34356605 PMCID: PMC8301841 DOI: 10.3390/biom11070982] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. The initial stage of CVDs is characterized by endothelial dysfunction, defined as the limited bioavailability of nitric oxide (NO). Thus, any factors that interfere with the synthesis or metabolism of NO in endothelial cells are involved in CVD pathogenesis. It is well established that hypoxia is both the triggering factor as well as the accompanying factor in cardiovascular disease, and diminished tissue oxygen levels have been reported to influence endothelial NO bioavailability. In endothelial cells, NO is produced by endothelial nitric oxide synthase (eNOS) from L-Arg, with tetrahydrobiopterin (BH4) as an essential cofactor. Here, we discuss the mechanisms by which hypoxia affects NO bioavailability, including regulation of eNOS expression and activity. What is particularly important is the fact that hypoxia contributes to the depletion of cofactor BH4 and deficiency of substrate L-Arg, and thus elicits eNOS uncoupling-a state in which the enzyme produces superoxide instead of NO. eNOS uncoupling and the resulting oxidative stress is the major driver of endothelial dysfunction and atherogenesis. Moreover, hypoxia induces impairment in mitochondrial respiration and endothelial cell activation; thus, oxidative stress and inflammation, along with the hypoxic response, contribute to the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Laboratory of Trace Elements Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Iwona T. Dobrucki
- University of Illinois at Urbana-Champaign Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL 61801, USA;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
- Correspondence:
| |
Collapse
|
32
|
Angolano C, Kaczmarek E, Essayagh S, Daniel S, Choi LY, Tung B, Sauvage G, Lee A, Kipper FC, Arvelo MB, Moll HP, Ferran C. A20/TNFAIP3 Increases ENOS Expression in an ERK5/KLF2-Dependent Manner to Support Endothelial Cell Health in the Face of Inflammation. Front Cardiovasc Med 2021; 8:651230. [PMID: 34026871 PMCID: PMC8138474 DOI: 10.3389/fcvm.2021.651230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/18/2021] [Indexed: 11/13/2022] Open
Abstract
Rationale: Decreased expression and activity of endothelial nitric oxide synthase (eNOS) in response to inflammatory and metabolic insults is the hallmark of endothelial cell (EC) dysfunction that preludes the development of atherosclerosis and hypertension. We previously reported the atheroprotective properties of the ubiquitin-editing and anti-inflammatory protein A20, also known as TNFAIP3, in part through interrupting nuclear factor-kappa B (NF-κB) and interferon signaling in EC and protecting these cells from apoptosis. However, A20's effect on eNOS expression and function remains unknown. In this study, we evaluated the impact of A20 overexpression or knockdown on eNOS expression in EC, at baseline and after tumor necrosis factor (TNF) treatment, used to mimic inflammation. Methods and Results: A20 overexpression in human coronary artery EC (HCAEC) significantly increased basal eNOS mRNA (qPCR) and protein (western blot) levels and prevented their downregulation by TNF. Conversely, siRNA-induced A20 knockdown decreased eNOS mRNA levels, identifying A20 as a physiologic regulator of eNOS expression. By reporter assays, using deletion and point mutants of the human eNOS promoter, and knockdown of eNOS transcriptional regulators, we demonstrated that A20-mediated increase of eNOS was transcriptional and relied on increased expression of the transcription factor Krüppel-like factor (KLF2), and upstream of KLF2, on activation of extracellular signal-regulated kinase 5 (ERK5). Accordingly, ERK5 knockdown or inhibition significantly abrogated A20's ability to increase KLF2 and eNOS expression. In addition, A20 overexpression in HCAEC increased eNOS phosphorylation at Ser-1177, which is key for the function of this enzyme. Conclusions: This is the first report demonstrating that overexpression of A20 in EC increases eNOS transcription in an ERK5/KLF2-dependent manner and promotes eNOS activating phosphorylation. This effect withstands eNOS downregulation by TNF, preventing EC dysfunction in the face of inflammation. This novel function of A20 further qualifies its therapeutic promise to prevent/treat atherosclerosis.
Collapse
Affiliation(s)
- Cleide Angolano
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Elzbieta Kaczmarek
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Sanah Essayagh
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Soizic Daniel
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Lynn Y. Choi
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Brian Tung
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Gabriel Sauvage
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Andy Lee
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Franciele C. Kipper
- The Division of Neurosurgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Maria B. Arvelo
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Herwig P. Moll
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Christiane Ferran
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- The Transplant Institute and the Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Mustapha S, Mohammed M, Azemi AK, Yunusa I, Shehu A, Mustapha L, Wada Y, Ahmad MH, Ahmad WANW, Rasool AHG, Mokhtar SS. Potential Roles of Endoplasmic Reticulum Stress and Cellular Proteins Implicated in Diabesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8830880. [PMID: 33995826 PMCID: PMC8099518 DOI: 10.1155/2021/8830880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
The role of the endoplasmic reticulum (ER) has evolved from protein synthesis, processing, and other secretory pathways to forming a foundation for lipid biosynthesis and other metabolic functions. Maintaining ER homeostasis is essential for normal cellular function and survival. An imbalance in the ER implied stressful conditions such as metabolic distress, which activates a protective process called unfolded protein response (UPR). This response is activated through some canonical branches of ER stress, i.e., the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6). Therefore, chronic hyperglycemia, hyperinsulinemia, increased proinflammatory cytokines, and free fatty acids (FFAs) found in diabesity (a pathophysiological link between obesity and diabetes) could lead to ER stress. However, limited data exist regarding ER stress and its association with diabesity, particularly the implicated proteins and molecular mechanisms. Thus, this review highlights the role of ER stress in relation to some proteins involved in diabesity pathogenesis and provides insight into possible pathways that could serve as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Pulau Pinang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, College of Pharmacy, Columbia, SC, USA
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna, Nigeria
| | - Yusuf Wada
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mubarak Hussaini Ahmad
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
- School of Pharmacy Technician, Aminu Dabo College of Health Sciences and Technology, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
34
|
TRPC channel-derived calcium fluxes differentially regulate ATP and flow-induced activation of eNOS. Nitric Oxide 2021; 111-112:1-13. [PMID: 33813098 DOI: 10.1016/j.niox.2021.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/27/2021] [Indexed: 12/28/2022]
Abstract
Endothelial dysfunction, characterised by impaired nitric oxide (NO) bioavailability, arises in response to a variety of cardiovascular risk factors and precedes atherosclerosis. NO is produced by tight regulation of endothelial nitric oxide synthase (eNOS) activity in response to vasodilatory stimuli. This regulation of eNOS is mediated in part by store-operated calcium entry (SOCE). We hypothesized that both ATP- and flow-induced eNOS activation are regulated by SOCE derived from Orai1 channels and members of the transient receptor potential canonical (TRPC) channel family. Bovine aortic endothelial cells (BAECs) were pre-treated with pharmacological inhibitors of TRPC channels and Orai1 to examine their effect on calcium signaling and eNOS activation in response to flow and ATP. The peak and sustained ATP-induced calcium signal and the resulting eNOS activation were attenuated by inhibition of TRPC3, which we found to be store operated. TRPC4 blockade reduced the transient peak in calcium concentration following ATP stimulation, but did not significantly reduce eNOS activity. Simultaneous TRPC3 & 4 inhibition reduced flow-induced NO production via alterations in phosphorylation-mediated eNOS activity. Inhibition of TRPC1/6 or Orai1 failed to lower ATP-induced calcium entry or eNOS activation. Our results suggest that TRPC3 is a store-operated channel in BAECs and is the key regulator of ATP-induced eNOS activation, whereas flow stimulation also recruits TRPC4 into the pathway for the synthesis of NO.
Collapse
|
35
|
Lu YA, Jiang Y, Yang HW, Hwang J, Jeon YJ, Ryu B. Diphlorethohydroxycarmalol Isolated from Ishige okamurae Exerts Vasodilatory Effects via Calcium Signaling and PI3K/Akt/eNOS Pathway. Int J Mol Sci 2021; 22:1610. [PMID: 33562632 PMCID: PMC7914902 DOI: 10.3390/ijms22041610] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide (NO) is released by endothelial cells in the blood vessel wall to enhance vasodilation. Marine polyphenols are known to have protective effects against vascular dysfunction and hypertension. The present study is the first to investigate how diphlorethohydroxycarmalol (DPHC) isolated from Ishige okamurae affects calcium levels, resulting in enhanced vasodilation. We examined calcium modulation with the well-known receptors, acetylcholine receptor (AchR) and vascular endothelial growth factor 2 (VEGFR2), which are related to NO formation, and further confirmed the vasodilatory effect of DPHC. We confirmed that DPHC stimulated NO production by increasing calcium levels and endothelial nitric oxide synthase (eNOS) expression. DPHC affected AchR and VEGFR2 expression, thereby influencing transient calcium intake. Specific antagonists, atropine and SU5416, were used to verify our findings. Furthermore, based on the results of in vivo experiments, we treated Tg(flk:EGFP) transgenic zebrafish with DPHC to confirm its vasodilatory effect. In conclusion, the present study showed that DPHC modulated calcium transit through AchR and VEGFR2, increasing endothelial-dependent NO production. Thus, DPHC, a natural marine component, can efficiently ameliorate cardiovascular diseases by improving vascular function.
Collapse
Affiliation(s)
- Yu An Lu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Yunfei Jiang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Jin Hwang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - Bomi Ryu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| |
Collapse
|
36
|
Shear Stress and RBC-NOS Serine1177 Phosphorylation in Humans: A Dose Response. Life (Basel) 2021; 11:life11010036. [PMID: 33429979 PMCID: PMC7828091 DOI: 10.3390/life11010036] [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: 12/14/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 01/01/2023] Open
Abstract
Red blood cells (RBC) express a nitric oxide synthase isoform (RBC-NOS) that appears dependent on shear stress for Serine1177 phosphorylation. Whether this protein is equally activated by varied shears in the physiological range is less described. Here, we explored RBC-NOS Serine1177 phosphorylation in response to shear stress levels reflective of in vivo conditions. Whole blood samples were exposed to specific magnitudes of shear stress (0.5, 1.5, 4.5, 13.5 Pa) for discrete exposure times (1, 10, 30 min). Thereafter, RBC-NOS Serine1177 phosphorylation was measured utilising immunofluorescence labelling. Shear stress exposure at 0.5, 1.5, and 13.5 Pa significantly increased RBC-NOS Serine1177 phosphorylation following 1 min (p < 0.0001); exposure to 4.5 Pa had no effect after 1 min. RBC-NOS Serine1177 phosphorylation was significantly increased following 10 min at each magnitude of shear stress (0.5, 1.5, 13.5 Pa, p < 0.0001; 4.5 Pa, p = 0.0042). Shear stress exposure for 30 min significantly increased RBC-NOS Serine1177 phosphorylation at 0.5 Pa and 13.5 Pa (p < 0.0001). We found that RBC-NOS phosphorylation via shear stress is non-linear and differs for a given magnitude and duration of exposure. This study provides a new understanding of the discrete relation between RBC-NOS and shear stress.
Collapse
|
37
|
Filimonova MV, Makarchuk VM, Shevchenko LI, Saburova AS, Surinova VI, Izmestieva OS, Lychagin AA, Saburov VO, Shegay PV, Kaprin AD, Ivanov SA, Filimonov AS. Radioprotective Activity of the Nitric Oxide Synthase Inhibitor T1023. Toxicological and Biochemical Properties, Cardiovascular and Radioprotective Effects. Radiat Res 2020; 194:532-543. [PMID: 34609510 DOI: 10.1667/rade-20-00046.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/03/2020] [Indexed: 11/03/2022]
Abstract
In this work, studies were performed to investigate the toxicological, biochemical, vasotropic and radiomodifying properties of the new nitric oxide synthase (NOS) inhibitor, compound T1023. Toxicological studies included the estimation of acute toxicity in mice after i.p. administration of T1023. Radiometric analysis and electron paramagnetic resonance spectroscopy were used to study NOS-inhibitory properties of T1023 in vitro and in vivo, respectively. T1023 vasoactive properties were studied in rat central hemodynamics. Radiobiological experiments were performed using endogenous and exogenous spleen colony formation as well as 30-day survival tests. The morphological changes in peripheral blood and bone marrow (BM) induced with T1023 were analyzed in mice during hematopoietic acute radiation syndrome (H-ARS). It was shown that T1023 is a sufficiently safe compound (LD10 of 317 mg/kg; LD50 of 410 mg/kg). It is an effective competitive NOS-inhibitor that is 10-to-15-fold selective to endothelial and inducible NOS (IC50 for nNOS, iNOS, eNOS: 52.3, 3.2 and 5.1 µM, respectively). Its NOS-inhibitory activity is realized in vivo and is accompanied by an increase in vascular tone. Its single i.p. administration in doses greater than 1/8 LD10 provides significant (40-50%) and long-lasting (more than 90 min) weakening of cardiac output, which can cause transient hypoxia. In radiobiological studies, T1023 proved to be a hypoxic radioprotector. Its radioprotective effect was observed only when administered prophylactically [single i.p dose, 5-120 min before total-body irradiation (TBI)] and only in doses that reduced cardiac output (1/8 LD10 and more, 40 mg/kg for mice), and was correlated in time with the dynamics of circulatory depression. Its radioprotective effect was not observed when administered in vitro and in the first 4 h after TBI. The optimal radioprotective doses of T1023 are relatively safe (1/ 5-1/4 LD10). In addition, T1023 effectively prevents H-ARS and gastrointestinal acute radiation syndrome (G-ARS) in experimental animals in vivo: dose modifying factor of 1.6-1.9. In the H-ARS mouse model, the prophylactic effect of T1023 (75 mg/kg, single i.p. injection) was accompanied by clinically significant effects. There was an express decrease in the degree of indicators of early BM devastation (by 40%) and maximal neutropenia and thrombocytopenia (2-2.5 times), in addition to a reduction in recovery time (by 30-40%). The obtained experimental results and literature data indicate that NOS inhibitors are an independent class of vasoactive radioprotectors with a specific hypoxic mechanism of action. NOS inhibitors provide new opportunities for developing effective and safe tools for the prevention of ARS.
Collapse
Affiliation(s)
- Marina V Filimonova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Victoria M Makarchuk
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Ljudmila I Shevchenko
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alina S Saburova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Valentina I Surinova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Olga S Izmestieva
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anatoly A Lychagin
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Vyacheslav O Saburov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Petr V Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergey A Ivanov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alexander S Filimonov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| |
Collapse
|
38
|
Dangudubiyyam SV, Mishra JS, Zhao H, Kumar S. Perfluorooctane sulfonic acid (PFOS) exposure during pregnancy increases blood pressure and impairs vascular relaxation mechanisms in the adult offspring. Reprod Toxicol 2020; 98:165-173. [PMID: 32980420 DOI: 10.1016/j.reprotox.2020.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
Perfluorooctanesulfonate (PFOS) is a persistent environmental agent. We examined whether PFOS exposure during pregnancy alters blood pressure in male and female offspring, and if this is related to sex-specific changes in vascular mechanisms. PFOS was administered through drinking water (50 μg/mL) to pregnant Sprague-Dawley rats from gestational day 4 until delivery. PFOS-exposure decreased maternal weight gain but did not significantly alter feed and water intake in dams. The male and female pups born to PFOS mothers were smaller in weight by 29 % and 27 %, respectively. The male PFOS offspring remained smaller through adulthood, but the female PFOS offspring exhibited catch-up growth. The blood pressure at 12 and 16 weeks of age was elevated at similar magnitude in PFOS males and females than controls. Mesenteric arterial relaxation to acetylcholine was reduced in both PFOS males and females, but the extent of decrease was greater in females. Relaxation to sodium-nitroprusside was reduced in PFOS females but unaffected in PFOS males. Vascular eNOS expression was not changed, but phospho(Ser1177)-eNOS was decreased in PFOS males. In PFOS females, both total eNOS and phospho(Ser1177)-eNOS expression were reduced. In conclusion, PFOS exposure during prenatal life (1) caused low birth weight followed by catch-up growth only in females (2) lead to hypertension of similar magnitude in both males and females; (2) decreased endothelium-dependent vascular relaxation in males but suppressed both endothelium-dependent and -independent relaxation in females. The endothelial dysfunction is associated with reduced activity of eNOS in males and decreased expression and activity of eNOS in females.
Collapse
Affiliation(s)
- Sri Vidya Dangudubiyyam
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA.
| | - Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA.
| | - Hanjie Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA.
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA; Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.
| |
Collapse
|
39
|
de Yébenes VG, Briones AM, Martos-Folgado I, Mur SM, Oller J, Bilal F, González-Amor M, Méndez-Barbero N, Silla-Castro JC, Were F, Jiménez-Borreguero LJ, Sánchez-Cabo F, Bueno H, Salaices M, Redondo JM, Ramiro AR. Aging-Associated miR-217 Aggravates Atherosclerosis and Promotes Cardiovascular Dysfunction. Arterioscler Thromb Vasc Biol 2020; 40:2408-2424. [PMID: 32847388 PMCID: PMC7505150 DOI: 10.1161/atvbaha.120.314333] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Objective: microRNAs are master regulators of gene expression with essential roles in virtually all biological processes. miR-217 has been associated with aging and cellular senescence, but its role in vascular disease is not understood. Approach and Results: We have used an inducible endothelium-specific knock-in mouse model to address the role of miR-217 in vascular function and atherosclerosis. miR-217 reduced NO production and promoted endothelial dysfunction, increased blood pressure, and exacerbated atherosclerosis in proatherogenic apoE−/− mice. Moreover, increased endothelial miR-217 expression led to the development of coronary artery disease and altered left ventricular heart function, inducing diastolic and systolic dysfunction. Conversely, inhibition of endogenous vascular miR-217 in apoE−/− mice improved vascular contractility and diminished atherosclerosis. Transcriptome analysis revealed that miR-217 regulates an endothelial signaling hub and downregulates a network of eNOS (endothelial NO synthase) activators, including VEGF (vascular endothelial growth factor) and apelin receptor pathways, resulting in diminished eNOS expression. Further analysis revealed that human plasma miR-217 is a biomarker of vascular aging and cardiovascular risk. Conclusions: Our results highlight the therapeutic potential of miR-217 inhibitors in aging-related cardiovascular disease.
Collapse
Affiliation(s)
- Virginia G de Yébenes
- Department of Vascular Physiopathology, B Lymphocyte Biology Lab (V.G.d.Y., I.M.-F., S.M.M., F.B., A.R.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 12 de Octubre Health Research Institute, Madrid, Spain (V.G.d.Y.)
| | - Ana M Briones
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Spain (A.M.B., M.G.-A., M.S.).,CIBER de Enfermedades Cardiovasculares, Spain (A.M.B., M.G.-A., M.S., J.M.R.)
| | - Inmaculada Martos-Folgado
- Department of Vascular Physiopathology, B Lymphocyte Biology Lab (V.G.d.Y., I.M.-F., S.M.M., F.B., A.R.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Sonia M Mur
- Department of Vascular Physiopathology, B Lymphocyte Biology Lab (V.G.d.Y., I.M.-F., S.M.M., F.B., A.R.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Jorge Oller
- Gene Regulation in Cardiovascular Remodelling and Inflammation Lab (J.O., N.M.-B., J.M.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Faiz Bilal
- Department of Vascular Physiopathology, B Lymphocyte Biology Lab (V.G.d.Y., I.M.-F., S.M.M., F.B., A.R.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - María González-Amor
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Spain (A.M.B., M.G.-A., M.S.).,CIBER de Enfermedades Cardiovasculares, Spain (A.M.B., M.G.-A., M.S., J.M.R.)
| | - Nerea Méndez-Barbero
- Gene Regulation in Cardiovascular Remodelling and Inflammation Lab (J.O., N.M.-B., J.M.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Juan Carlos Silla-Castro
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (J.C.S.-C., F.W., F.S.-C.)
| | - Felipe Were
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (J.C.S.-C., F.W., F.S.-C.)
| | | | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (J.C.S.-C., F.W., F.S.-C.)
| | - Héctor Bueno
- Department of Cell & Developmental Biology, Multidisciplinary Translational Cardiovascular Research (H.B.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Spain (A.M.B., M.G.-A., M.S.).,CIBER de Enfermedades Cardiovasculares, Spain (A.M.B., M.G.-A., M.S., J.M.R.)
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Lab (J.O., N.M.-B., J.M.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Spain (A.M.B., M.G.-A., M.S.).,CIBER de Enfermedades Cardiovasculares, Spain (A.M.B., M.G.-A., M.S., J.M.R.)
| | - Almudena R Ramiro
- Department of Vascular Physiopathology, B Lymphocyte Biology Lab (V.G.d.Y., I.M.-F., S.M.M., F.B., A.R.R.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
40
|
Masaki N, Ido Y, Yamada T, Yamashita Y, Toya T, Takase B, Hamburg NM, Adachi T. Endothelial Insulin Resistance of Freshly Isolated Arterial Endothelial Cells From Radial Sheaths in Patients With Suspected Coronary Artery Disease. J Am Heart Assoc 2020; 8:e010816. [PMID: 30885039 PMCID: PMC6475050 DOI: 10.1161/jaha.118.010816] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Endothelial insulin resistance is insulin‐insensitivity in the vascular endothelium and can be observed in experimental models. This study aimed to investigate endothelial insulin resistance in patients with suspected coronary artery disease. To this end, a novel method of obtaining freshly isolated arterial endothelial cells from a radial catheter sheath was developed. Methods and Results Freshly isolated arterial endothelial cells were retrieved from catheter sheaths placed in radial arteries for coronary angiography (n=69, patient age 64±12 years). The endothelial cells were divided into groups for incubation with or without insulin, vascular endothelial growth factor, or acetylcholine. The intensity of phosphorylated endothelial nitric oxide synthase at Ser1177 (p‐eNOS) was quantified by immunofluorescence microscopy. The percentage increase of insulin‐induced phosphorylated endothelial nitric oxide synthase correlated negatively with derivatives of reactive oxygen metabolites, an oxidative stress test (r=−0.348, n=53, P=0.011), E/E′, an index of left ventricular diastolic dysfunction in Doppler echocardiography (ρ=−0.374, n=49, P=0.008), and log‐transformed brain natriuretic peptide (r=−0.266, n=62, P=0.037). Furthermore, percentage increase of insulin‐induced p‐eNOS was an independent factor for the cardio‐ankle vascular index (standardized coefficient β=−0.293, n=42, P=0.021) in the multivariate regression analysis of adaptive least absolute shrinkage and selection operator. Conclusions Our results suggested that endothelial insulin resistance is associated with oxidative stress, left ventricular diastolic dysfunction, heart failure, and arterial stiffness.
Collapse
Affiliation(s)
- Nobuyuki Masaki
- 1 Department of Intensive Care Medicine National Defense Medical College Tokorozawa Japan
| | - Yasuo Ido
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| | - Toshiyuki Yamada
- 3 Department of Cardiovascular Surgery Keio University Graduate School of Medicine Tokyo Japan
| | - Youhei Yamashita
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| | - Takumi Toya
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| | - Bonpei Takase
- 1 Department of Intensive Care Medicine National Defense Medical College Tokorozawa Japan
| | - Naomi M Hamburg
- 4 The Whitaker Cardiovascular Institute Department of Medicine Boston University School of Medicine Boston MA
| | - Takeshi Adachi
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| |
Collapse
|
41
|
Tettey CO, Yang IJ, Shin HM. Endothelium-dependent vasodilatory effect of Smilax china Linn. water extract via PI3K/Akt signaling. Arch Physiol Biochem 2020; 126:209-213. [PMID: 30269606 DOI: 10.1080/13813455.2018.1508237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Objective: The objective of this study was to investigate the pharmacological effect of Smilax china Linn. water extract (SCLWE) on vascular relaxation and its underlying biochemical mechanisms.Methodology: Isolated rat aortic rings were pre-constricted with phenylephrine (PE). This was followed by the cumulative addition of SCLWE. The effect of endothelial nitric oxide and PI3K/Akt on the SCLWE-induced vasodilation was investigated by the pretreatment of endothelium-intact aortic strips with or without NG-nitro-L-arginine methyl ester (L-NAME) or wortmanin before constriction with PE.Results: Treatment of PE (1 μM)-pre-contracted aortic strips with SCLWE induced endothelium-dependent relaxation, which was attenuated by L-NAME and wortmanin. Further studies using HUVECs indicated that nitrite production, eNOS and PI3K/PKB (Akt) phosphorylations were increased after exposure to SCLWE but was attenuated by pretreatment with wortmanin.Conclusion: These results suggest that SCLWE induces vasodilation by augmenting NO production in endothelial cells via PI3K/Akt-dependent eNOS phosphorylation.
Collapse
Affiliation(s)
- Clement Okraku Tettey
- Department of Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - In-Jun Yang
- Department of Physiology College of Korean Medicine, Dongguk University, Gyeongju, South Korea
| | - Heung-Mook Shin
- Department of Physiology College of Korean Medicine, Dongguk University, Gyeongju, South Korea
- Korea Promotion Institute for Traditional Medicine Industry, Gyeongsan, South Korea
| |
Collapse
|
42
|
Schirra C, Xia N, Schüffler A, Heck A, Hasselwander S, Förstermann U, Li H. Phosphorylation and activation of endothelial nitric oxide synthase by red fruit (Pandanus conoideus Lam) oil and its fractions. JOURNAL OF ETHNOPHARMACOLOGY 2020; 251:112534. [PMID: 31893533 DOI: 10.1016/j.jep.2019.112534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 10/03/2019] [Accepted: 12/27/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Red fruit (Pandanus conoideus Lam) oil (RFO) is utilized by inhabitants of the Papua Island to treat diseases such as infections, cancer, and cardiovascular disease, but the mechanism of action is unknown. AIM OF THE STUDY We have recently shown that RFO stimulates nitric oxide (NO) production in endothelial cells. The present study was conducted to investigate the molecular mechanism of endothelial NO synthase (eNOS) activation by RFO. MATERIALS AND METHODS NO production by endothelial cells was determined with electron paramagnetic resonance. The vascular function of isolated mouse aorta was examined using a wire myograph system. Phosphorylation of eNOS was studied with Western blot analyses. RESULTS RFO induced concentration-dependent vasodilation in isolated mouse aorta. The vasodilator effect of RFO was lost in endothelium-denuded aorta and in aorta from mice deficient in eNOS. Treatment of human EA.hy 926 endothelial cells with RFO led to an enhancement of eNOS phosphorylation at serine 1177 and NO production. The RFO-induced eNOS phosphorylation and NO production were reduced by inhibitors of Akt or AMPK, but not by an inhibitor of CaMKII. The effects of RFO were decreased by pharmacological inhibition of PI3K, indicating an involvement of the PI3K-Akt pathway. Moreover, acetone-soluble fractions and oily fractions of RFO showed higher efficacies than the RFO polar fraction in activating eNOS. CONCLUSIONS RFO contains highly active compounds that enhance NO production through Akt- or AMPK-mediated eNOS phosphorylation. The increase in endothelial NO production is likely to represent one of the molecular mechanisms responsible for the therapeutic effects of RFO.
Collapse
Affiliation(s)
- Christian Schirra
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anja Schüffler
- Institute for Biotechnology and Drug Research (IBWF gGmbH), Kaiserslautern, Germany
| | - Astrid Heck
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Solveig Hasselwander
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ulrich Förstermann
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany.
| |
Collapse
|
43
|
Plasma from Volunteers Breathing Helium Reduces Hypoxia-Induced Cell Damage in Human Endothelial Cells-Mechanisms of Remote Protection Against Hypoxia by Helium. Cardiovasc Drugs Ther 2020; 33:297-306. [PMID: 31025141 PMCID: PMC6538579 DOI: 10.1007/s10557-019-06880-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Remote ischemic preconditioning protects peripheral organs against prolonged ischemia/reperfusion injury via circulating protective factors. Preconditioning with helium protected healthy volunteers against postischemic endothelial dysfunction. We investigated whether plasma from helium-treated volunteers can protect human umbilical vein endothelial cells (HUVECs) against hypoxia in vitro through release of circulating of factors. METHODS Healthy male volunteers inhaled heliox (79% helium, 21% oxygen) or air for 30 min. Plasma was collected at baseline, directly after inhalation, 6 h and 24 h after start of the experiment. HUVECs were incubated with either 5% or 10% of the plasma for 1 or 2 h and subjected to enzymatically induced hypoxia. Cell damage was measured by LDH content. Furthermore, caveolin 1 (Cav-1), hypoxia-inducible factor (HIF1α), extracellular signal-regulated kinase (ERK)1/2, signal transducer and activator of transcription (STAT3) and endothelial nitric oxide synthase (eNOS) were determined. RESULTS Prehypoxic exposure to 10% plasma obtained 6 h after helium inhalation decreased hypoxia-induced cell damage in HUVEC. Cav-1 knockdown in HUVEC abolished this effect. CONCLUSIONS Plasma of healthy volunteers breathing helium protects HUVEC against hypoxic cell damage, possibly involving circulating Cav-1.
Collapse
|
44
|
Cantalupo A, Sasset L, Gargiulo A, Rubinelli L, Del Gaudio I, Benvenuto D, Wadsack C, Jiang XC, Bucci MR, Di Lorenzo A. Endothelial Sphingolipid De Novo Synthesis Controls Blood Pressure by Regulating Signal Transduction and NO via Ceramide. Hypertension 2020; 75:1279-1288. [PMID: 32172624 DOI: 10.1161/hypertensionaha.119.14507] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ceramides are sphingolipids that modulate a variety of cellular processes via 2 major mechanisms: functioning as second messengers and regulating membrane biophysical properties, particularly lipid rafts, important signaling platforms. Altered sphingolipid levels have been implicated in many cardiovascular diseases, including hypertension, atherosclerosis, and diabetes mellitus-related conditions; however, molecular mechanisms by which ceramides impact endothelial functions remain poorly understood. In this regard, we generated mice defective of endothelial sphingolipid de novo biosynthesis by deleting the Sptlc2 (long chain subunit 2 of serine palmitoyltransferase)-the first enzyme of the pathway. Our study demonstrated that endothelial sphingolipid de novo production is necessary to regulate (1) signal transduction in response to NO agonists and, mainly via ceramides, (2) resting eNOS (endothelial NO synthase) phosphorylation, and (3) blood pressure homeostasis. Specifically, our findings suggest a prevailing role of C16:0-Cer in preserving vasodilation induced by tyrosine kinase and GPCRs (G-protein coupled receptors), except for Gq-coupled receptors, while C24:0- and C24:1-Cer control flow-induced vasodilation. Replenishing C16:0-Cer in vitro and in vivo reinstates endothelial cell signaling and vascular tone regulation. This study reveals an important role of locally produced ceramides, particularly C16:0-, C24:0-, and C24:1-Cer in vascular and blood pressure homeostasis, and establishes the endothelium as a key source of plasma ceramides. Clinically, specific plasma ceramides ratios are independent predictors of major cardiovascular events. Our data also suggest that plasma ceramides might be indicative of the diseased state of the endothelium.
Collapse
Affiliation(s)
- Anna Cantalupo
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.)
| | - Linda Sasset
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.)
| | - Antonella Gargiulo
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.).,Department of Pharmacy, School of Medicine, University of Naples Federico II, Italy (A.G., M.R.B.)
| | - Luisa Rubinelli
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.)
| | - Ilaria Del Gaudio
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.).,Department of Obstetrics and Gynaecology, Medical University of Graz, Austria (I.d.G., C.W.).,BioTechMed-Graz, Austria (I.d.G., C.W.)
| | - Domenico Benvenuto
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.)
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Austria (I.d.G., C.W.).,BioTechMed-Graz, Austria (I.d.G., C.W.)
| | - Xiang-Chen Jiang
- Department of Anatomy and Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (X.-C.J.)
| | - Maria Rosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Italy (A.G., M.R.B.)
| | - Annarita Di Lorenzo
- From the Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (A.C., L.S., A.G., L.R., I.d.G., D.B., A.D.L.)
| |
Collapse
|
45
|
Wang CY, Lee S, Jang HJ, Su XD, Wang HS, Kim YH, Yang SY. Inhibition potential of phenolic constituents from the aerial parts of Tetrastigma hemsleyanum against soluble epoxide hydrolase and nitric oxide synthase. J Enzyme Inhib Med Chem 2019; 34:753-760. [PMID: 30829084 PMCID: PMC6407587 DOI: 10.1080/14756366.2019.1584621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
The aerial parts of Tetrastigma hemsleyanum (APTH) have been used as a functional tea in China. The purpose of the current study was to identify the bioactive constituents with inhibitory activity against soluble epoxide hydrolase (sEH) and inducible nitric oxide synthase (iNOS), which are jointly considered potential therapeutic targets for vascular system diseases. In the present study, 39 compounds (1-39) were isolated from the APTH. Among them, compounds 8, 10, 12, 16, 17, 19, and 32 displayed potential activities, with IC50 values ranging from 4.5 to 9.5 µM, respectively, and all in non-competitive inhibition mode. Compounds 5, 10, 12, 19, and 32 displayed potent iNOS inhibitory effects, with IC50 values ranging from 15.6 to 47.3 µM. The results obtained in this work contribute to a better understanding of the pharmacological activities of T. hemsleyanum and its potential application as a functional food.
Collapse
Affiliation(s)
- Cai Yi Wang
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Sunggun Lee
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Hyun-Jae Jang
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Korea
| | - Xiang Dong Su
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Heng-Shan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, People's Republic of China
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Seo Young Yang
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| |
Collapse
|
46
|
Discordance between eNOS phosphorylation and activation revealed by multispectral imaging and chemogenetic methods. Proc Natl Acad Sci U S A 2019; 116:20210-20217. [PMID: 31527268 DOI: 10.1073/pnas.1910942116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Nitric oxide (NO) synthesized by the endothelial isoform of nitric oxide synthase (eNOS) is a critical determinant of vascular homeostasis. However, the real-time detection of intracellular NO-a free radical gas-has been difficult, and surrogate markers for eNOS activation are widely utilized. eNOS phosphorylation can be easily measured in cells by probing immunoblots with phosphospecific antibodies. Here, we pursued multispectral imaging approaches using biosensors to visualize intracellular NO and Ca2+ and exploited chemogenetic approaches to define the relationships between NO synthesis and eNOS phosphorylation in cultured endothelial cells. We found that the G protein-coupled receptor agonists adenosine triphosphate (ATP) and histamine promoted rapid increases in eNOS phosphorylation, as did the receptor tyrosine kinase agonists insulin and Vascular Endothelial Growth Factor (VEGF). Histamine and ATP also promoted robust NO formation and increased intracellular Ca2+ By contrast, neither insulin nor VEGF caused any increase whatsoever in intracellular NO or Ca2+-despite eliciting strong eNOS phosphorylation responses. Our findings demonstrate an unexpected and striking discordance between receptor-modulated eNOS phosphorylation and NO formation in endothelial cells. Previous reports in which phosphorylation of eNOS has been studied as a surrogate for enzyme activation may need to be reassessed.
Collapse
|
47
|
Lian X, Matthaeus C, Kaßmann M, Daumke O, Gollasch M. Pathophysiological Role of Caveolae in Hypertension. Front Med (Lausanne) 2019; 6:153. [PMID: 31355199 PMCID: PMC6635557 DOI: 10.3389/fmed.2019.00153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/20/2019] [Indexed: 12/02/2022] Open
Abstract
Caveolae, flask-shaped cholesterol-, and glycosphingolipid-rich membrane microdomains, contain caveolin 1, 2, 3 and several structural proteins, in particular Cavin 1-4, EHD2, pacsin2, and dynamin 2. Caveolae participate in several physiological processes like lipid uptake, mechanosensitivity, or signaling events and are involved in pathophysiological changes in the cardiovascular system. They serve as a specific membrane platform for a diverse set of signaling molecules like endothelial nitric oxide synthase (eNOS), and further maintain vascular homeostasis. Lack of caveolins causes the complete loss of caveolae; induces vascular disorders, endothelial dysfunction, and impaired myogenic tone; and alters numerous cellular processes, which all contribute to an increased risk for hypertension. This brief review describes our current knowledge on caveolae in vasculature, with special focus on their pathophysiological role in hypertension.
Collapse
Affiliation(s)
- Xiaoming Lian
- Experimental and Clinical Research Center—A Joint Cooperation Between the Charité–University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Claudia Matthaeus
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mario Kaßmann
- Experimental and Clinical Research Center—A Joint Cooperation Between the Charité–University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Daumke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center—A Joint Cooperation Between the Charité–University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Medical Clinic for Nephrology and Internal Intensive Care, Berlin, Germany
| |
Collapse
|
48
|
Wang XH, Yan CY, Liu JR. Hyperinsulinemia-induced KLF5 mediates endothelial angiogenic dysfunction in diabetic endothelial cells. J Mol Histol 2019; 50:239-251. [PMID: 31049798 DOI: 10.1007/s10735-019-09821-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/27/2019] [Indexed: 12/15/2022]
Abstract
Reduced expression of endothelial nitric oxide synthase (eNOS) is a hallmark of endothelial dysfunction in diabetes, which predisposes diabetic patients to numerous cardiovascular complications including blunted angiogenesis. The Krüppel-like factor (KLF) five has been implicated as a central regulator of cardiovascular remodeling, but its role in endothelial cells (ECs) remains poorly understood. We show here that expression of endothelial KLF5 was significantly increased in the ECs from mouse diabetes mellitus type 2 (T2DM) model, when compared to non-diabetic or T1DM mouse. KLF5 up-regulation by insulin was dependent on activation of multiple pathways, including mammalian target of rapamycin, oxidative stress and Protein kinase C pathways. Hyperinsulinemia-induced KLF5 inhibited endothelial function and migration, and thereby compromised in vitro and in vivo angiogenesis. Mechanistically, KLF5 acted in concert with the MTA1 coregulator to negatively regulate NOS3 transcription, thereby leading to the diminished eNOS levels in ECs. Conversely, potentiation of cGMP content (the essential downstream effector of eNOS signaling) by pharmacological approaches successfully rescued the endothelial proliferation and in vitro tube formation, in the HUVECs overexpressing the exogenous KLF5. Collectively, the available data suggest that the augmentation of endothelial KLF5 expression by hyperinsulinemia may represent a novel mechanism for negatively regulating eNOS expression, and may thus help to explain for the T2DM-related endothelial dysfunction at the transcriptional level.
Collapse
Affiliation(s)
- Xi-Hui Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi, People's Republic of China
| | - Chang-You Yan
- Family Planning Service Stations of Health and Family Planning Commission of Chengcheng County, Chengcheng County, Weinan, 714000, Shaanxi, People's Republic of China
| | - Jian-Rong Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi, People's Republic of China. .,Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Road, Baqiao District, Xi'an, 710038, People's Republic of China.
| |
Collapse
|
49
|
Tettey CO, Yang IJ, Shin HM. Vasodilatory effect of kaempferol-7-O-α-L-rhamnopyranoside via NO-cGMP-PKG signaling. Arch Biochem Biophys 2019; 667:1-5. [PMID: 30981710 DOI: 10.1016/j.abb.2019.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/19/2019] [Accepted: 04/10/2019] [Indexed: 01/18/2023]
Abstract
Hypertension is one of the major causes of mortality. Though a host of drugs are available for the treatment of hypertension, majority have been linked to adverse side effects, necessitating the need for research into natural compounds with fewer side effects. Kaempferol-7-O-α-L-rhamnopyroside (KR) is a glycosylated flavone with neuroprotective and anti-inflammatory effects. However, no available literature exists on its vasodilatory effect. This study examined the pharmacological effect of KR on vasodilation/vasorelaxation and its mechanism of action in endothelial cells and rat thoracic aorta. Treatment of phenylephrine (PE; 2 × 10-6 M)-pre-contracted aortic rings with KR induced endothelium-dependent relaxation, which was suppressed by NG-nitro-l-arginine methyl ester (L-NAME; 10-4 M), (nitric oxide synthase (NOS) inhibitor). Phosphorylation of eNOS in human umbilical vein endothelial cells (HUVECs) was increased after exposure to KR. Pre-treatment of aortic rings with the cyclic GMP (cGMP) inhibitors; methylene blue (MB; 10-5 M) and 1-H-[1,2,4]-oxadiazolole-[4,3-α]-quinoxalin-10-one, (ODQ; 10-6 M) suppressed the KR-induced vasodilation. Furthermore, KR also increased protein kinase G (PKG) levels whereas it suppressed levels of phosphorylated myosin light chain (MLC) and protein kinase C (PKC) in aortic rings. These results suggest that KR induces endothelium-dependent vasorelaxation via the NO-cGMP-PKG pathway.
Collapse
Affiliation(s)
- Clement O Tettey
- Department of Biomedical Sciences, University of Health and Allied Sciences, PMB 31, Ho, Ghana; Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju, South Korea
| | - In-Jun Yang
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju, South Korea
| | - Heung-Mook Shin
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju, South Korea; National Development Institute of Korean Medicine, Gyeongsan, Gyeongbuk, 712-210, South Korea.
| |
Collapse
|
50
|
Lin Q, Zhao L, Jing R, Trexler C, Wang H, Li Y, Tang H, Huang F, Zhang F, Fang X, Liu J, Jia N, Chen J, Ouyang K. Inositol 1,4,5-Trisphosphate Receptors in Endothelial Cells Play an Essential Role in Vasodilation and Blood Pressure Regulation. J Am Heart Assoc 2019; 8:e011704. [PMID: 30755057 PMCID: PMC6405661 DOI: 10.1161/jaha.118.011704] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/17/2019] [Indexed: 01/06/2023]
Abstract
Background Endothelial NO synthase plays a central role in regulating vasodilation and blood pressure. Intracellular Ca2+ mobilization is a critical modulator of endothelial NO synthase function, and increased cytosolic Ca2+ concentration in endothelial cells is able to induce endothelial NO synthase phosphorylation. Ca2+ release mediated by 3 subtypes of inositol 1,4,5-trisphosphate receptors ( IP 3Rs) from the endoplasmic reticulum and subsequent Ca2+ entry after endoplasmic reticulum Ca2+ store depletion has been proposed to be the major pathway to mobilize Ca2+ in endothelial cells. However, the physiological role of IP 3Rs in regulating blood pressure remains largely unclear. Methods and Results To investigate the role of endothelial IP 3Rs in blood pressure regulation, we first generated an inducible endothelial cell-specific IP 3R1 knockout mouse model and found that deletion of IP 3R1 in adult endothelial cells did not affect vasodilation and blood pressure. Considering all 3 subtypes of IP 3Rs are expressed in mouse endothelial cells, we further generated inducible endothelial cell-specific IP 3R triple knockout mice and found that deletion of all 3 IP 3R subtypes decreased plasma NO concentration and increased basal blood pressure. Furthermore, IP 3R deficiency reduced acetylcholine-induced vasodilation and endothelial NO synthase phosphorylation at Ser1177. Conclusions Our results reveal that IP 3R-mediated Ca2+ release in vascular endothelial cells plays an important role in regulating vasodilation and physiological blood pressure.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Blood Pressure/physiology
- Calcium/metabolism
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension/physiopathology
- Immunoblotting
- Inositol 1,4,5-Trisphosphate Receptors/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myography
- Vasodilation/physiology
Collapse
Affiliation(s)
- Qingsong Lin
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Lingyun Zhao
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Ran Jing
- Department of CardiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Christa Trexler
- Department of MedicineSchool of MedicineUniversity of California San DiegoLa JollaCA
| | - Hong Wang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Yali Li
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Huayuan Tang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Fang Huang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Fei Zhang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Xi Fang
- Department of MedicineSchool of MedicineUniversity of California San DiegoLa JollaCA
| | - Jie Liu
- Department of PathophysiologySchool of MedicineShenzhen UniversityShenzhenChina
| | - Nan Jia
- Department of CardiologyThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Ju Chen
- Department of MedicineSchool of MedicineUniversity of California San DiegoLa JollaCA
| | - Kunfu Ouyang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| |
Collapse
|