1
|
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
|
2
|
Sakaguchi R, Takahashi N, Yoshida T, Ogawa N, Ueda Y, Hamano S, Yamaguchi K, Sawamura S, Yamamoto S, Hara Y, Kawamoto T, Suzuki R, Nakao A, Mori MX, Furukawa T, Shimizu S, Inoue R, Mori Y. Dynamic remodeling of TRPC5 channel-caveolin-1-eNOS protein assembly potentiates the positive feedback interaction between Ca 2+ and NO signals. J Biol Chem 2024; 300:107705. [PMID: 39178948 PMCID: PMC11420454 DOI: 10.1016/j.jbc.2024.107705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024] Open
Abstract
The cell signaling molecules nitric oxide (NO) and Ca2+ regulate diverse biological processes through their closely coordinated activities directed by signaling protein complexes. However, it remains unclear how dynamically the multicomponent protein assemblies behave within the signaling complexes upon the interplay between NO and Ca2+ signals. Here we demonstrate that TRPC5 channels activated by the stimulation of G-protein-coupled ATP receptors mediate Ca2+ influx, that triggers NO production from endothelial NO synthase (eNOS), inducing secondary activation of TRPC5 via cysteine S-nitrosylation and eNOS in vascular endothelial cells. Mutations in the caveolin-1-binding domains of TRPC5 disrupt its association with caveolin-1 and impair Ca2+ influx and NO production, suggesting that caveolin-1 serves primarily as the scaffold for TRPC5 and eNOS to assemble into the signal complex. Interestingly, during ATP receptor activation, eNOS is dissociated from caveolin-1 and in turn directly associates with TRPC5, which accumulates at the plasma membrane dependently on Ca2+ influx and calmodulin. This protein reassembly likely results in a relief of eNOS from the inhibitory action of caveolin-1 and an enhanced TRPC5 S-nitrosylation by eNOS localized in the proximity, thereby facilitating the secondary activation of Ca2+ influx and NO production. In isolated rat aorta, vasodilation induced by acetylcholine was significantly suppressed by the TRPC5 inhibitor AC1903. Thus, our study provides evidence that dynamic remodeling of the protein assemblies among TRPC5, eNOS, caveolin-1, and calmodulin determines the ensemble of Ca2+ mobilization and NO production in vascular endothelial cells.
Collapse
Affiliation(s)
- Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan; Laboratory of Biomaterials and Chemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Nobuaki Takahashi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Advanced Biomedical Engineering Research Unit, Kyoto University, Kyoto, Japan
| | - Takashi Yoshida
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Nozomi Ogawa
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yoshifumi Ueda
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Satoshi Hamano
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kaori Yamaguchi
- Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Seishiro Sawamura
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Shinichiro Yamamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Yuji Hara
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Department of Integrative Physiology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tomoya Kawamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Ryosuke Suzuki
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Akito Nakao
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Masayuki X Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Laboratory of Biomaterials and Chemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shunichi Shimizu
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University, Fukuoka, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan; Advanced Biomedical Engineering Research Unit, Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Szczesny-Malysiak E, Bartkowiak A, Dybas J. Label-free tracking of cytochrome C oxidation state in live cells by resonance Raman imaging. FEBS Lett 2024; 598:1981-1988. [PMID: 38740560 DOI: 10.1002/1873-3468.14905] [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: 02/09/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
Free interconversion of cytochrome C (CytC) between native ferrous (Cyt-FeII) and oxidized ferric (CytC-FeIII) states is necessary to maintain the respiratory function of mitochondria. Disturbances in CytC-FeIII to total CytC ratio may indicate mitochondrial dysfunction and apoptosis. Thus, tracking CytC oxidation state delivers important information about cellular physiology. In this work, we propose a novel methodology based on resonance Raman (rR) imaging optimized uniquely to track and qualitatively analyze the transition of Cyt-FeII to CytC-FeIII within live cells without affecting their morphology. None of the commonly used excitation lines allows such clear-cut differentiation, contrary to the 405 nm applied in this work. The presented methodology provides a novel pathway in the label-free detection of ferrous and ferric heme proteins.
Collapse
Affiliation(s)
- Ewa Szczesny-Malysiak
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Amanda Bartkowiak
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Jakub Dybas
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| |
Collapse
|
4
|
Ablooglu AJ, Chen WS, Xie Z, Desai A, Paul S, Lack JB, Scott LA, Eisch AR, Dudek AZ, Parikh SM, Druey KM. Intrinsic endothelial hyperresponsiveness to inflammatory mediators drives acute episodes in models of Clarkson disease. J Clin Invest 2024; 134:e169137. [PMID: 38502192 DOI: 10.1172/jci169137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
Clarkson disease, or monoclonal gammopathy-associated idiopathic systemic capillary leak syndrome (ISCLS), is a rare, relapsing-remitting disorder featuring the abrupt extravasation of fluids and proteins into peripheral tissues, which in turn leads to hypotensive shock, severe hemoconcentration, and hypoalbuminemia. The specific leakage factor(s) and pathways in ISCLS are unknown, and there is no effective treatment for acute flares. Here, we characterize an autonomous vascular endothelial defect in ISCLS that was recapitulated in patient-derived endothelial cells (ECs) in culture and in a mouse model of disease. ISCLS-derived ECs were functionally hyperresponsive to permeability-inducing factors like VEGF and histamine, in part due to increased endothelial nitric oxide synthase (eNOS) activity. eNOS blockade by administration of N(γ)-nitro-l-arginine methyl ester (l-NAME) ameliorated vascular leakage in an SJL/J mouse model of ISCLS induced by histamine or VEGF challenge. eNOS mislocalization and decreased protein phosphatase 2A (PP2A) expression may contribute to eNOS hyperactivation in ISCLS-derived ECs. Our findings provide mechanistic insights into microvascular barrier dysfunction in ISCLS and highlight a potential therapeutic approach.
Collapse
Affiliation(s)
- Ararat J Ablooglu
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Wei-Sheng Chen
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Zhihui Xie
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Abhishek Desai
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Subrata Paul
- Integrative Data Sciences Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Justin B Lack
- Integrative Data Sciences Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Linda A Scott
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - A Robin Eisch
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Arkadiusz Z Dudek
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samir M Parikh
- Division of Nephrology, Departments of Internal Medicine and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| |
Collapse
|
5
|
Smeir M, Chumala P, Katselis GS, Liu L. Lymphocyte-Specific Protein 1 Regulates Expression and Stability of Endothelial Nitric Oxide Synthase. Biomolecules 2024; 14:111. [PMID: 38254711 PMCID: PMC10813790 DOI: 10.3390/biom14010111] [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: 11/04/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Nitric oxide (NO), synthesized by endothelial nitric oxide synthase (eNOS), plays a critical role in blood pressure regulation. Genome-wide association studies have identified genetic susceptibility loci for hypertension in human lymphocyte-specific protein 1 (LSP1) gene. LSP1 is recognized as modulator of leukocyte extravasation, and endothelial permeability, however, the role of LSP1 in regulation of NO signaling within endothelial cells (ECs) remains unknown. The present study investigated the role of LSP1 in the regulation of eNOS expression and activity utilizing human macrovascular ECs in vitro and LSP1 knockout (KO) mice. In ECs, specific CRISPR-Cas9 genomic editing deleted LSP1 and caused downregulation of eNOS expression. LSP1 gain-of-function through adenovirus-mediated gene transfer was associated with enhanced expression of eNOS. Co-immunoprecipitation and confocal fluorescence microscopy revealed that eNOS and LSP1 formed a protein complex under basal conditions in ECs. Furthermore, LSP1 deficiency in mice promoted significant upregulation and instability of eNOS. Utilizing a mass-spectrometry-based bottom-up proteomics approach, we identified novel truncated forms of eNOS in immunoprecipitates from LSP1 KO aortae. Our experimental data suggest an important role of endothelial LSP1 in regulation of eNOS expression and activity within human ECs and murine vascular tissues.
Collapse
Affiliation(s)
- Musstafa Smeir
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada;
| | - Paulos Chumala
- Department of Medicine, Canadian Center for Rural and Agricultural Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (P.C.); (G.S.K.)
| | - George S. Katselis
- Department of Medicine, Canadian Center for Rural and Agricultural Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (P.C.); (G.S.K.)
| | - Lixin Liu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada;
| |
Collapse
|
6
|
Narasaki S, Noguchi S, Urabe T, Harada K, Hide I, Tanaka S, Yanase Y, Kajimoto T, Uchida K, Tsutsumi YM, Sakai N. Identification of protein kinase C domains involved in its translocation induced by propofol. Eur J Pharmacol 2023; 955:175806. [PMID: 37230321 DOI: 10.1016/j.ejphar.2023.175806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Propofol is widely used for general anesthesia and sedation; however, the mechanisms of its anesthetic and adverse effects are not fully understood. We have previously shown that propofol activates protein kinase C (PKC) and induces its translocation in a subtype-specific manner. The purpose of this study was to identify the PKC domains involved in propofol-induced PKC translocation. The regulatory domains of PKC consist of C1 and C2 domains, and the C1 domain is subdivided into the C1A and C1B subdomains. Mutant PKCα and PKCδ with each domain deleted were fused with green fluorescent protein (GFP) and expressed in HeLa cells. Propofol-induced PKC translocation was observed by time-lapse imaging using a fluorescence microscope. The results showed that persistent propofol-induced PKC translocation to the plasma membrane was abolished by the deletion of both C1 and C2 domains in PKCα and by the deletion of the C1B domain in PKCδ. Therefore, propofol-induced PKC translocation involves the C1 and C2 domains of PKCα and the C1B domain of PKCδ. We also found that treatment with calphostin C, a C1 domain inhibitor, abolished propofol-induced PKCδ translocation. In addition, calphostin C inhibited the propofol-induced phosphorylation of endothelial nitric oxide synthase (eNOS). These results suggest that it may be possible to modulate the exertion of propofol effects by regulating the PKC domains involved in propofol-induced PKC translocation.
Collapse
Affiliation(s)
- Soshi Narasaki
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan; Dept of Anesthesiology & Critical Care, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Soma Noguchi
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Tomoaki Urabe
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan; Dept of Anesthesiology & Critical Care, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Kana Harada
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Izumi Hide
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Shigeru Tanaka
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Yuhki Yanase
- Dept of Pharmacotherapy, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Taketoshi Kajimoto
- Div of Biochem, Dept of Biochem and Mol Biol, Kobe Univ Grad Sch of Med, Japan
| | - Kazue Uchida
- Dept of Dermatology, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Yasuo M Tsutsumi
- Dept of Anesthesiology & Critical Care, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan
| | - Norio Sakai
- Dept of Mol & Pharmacol Neurosci, Grad Sch of Biomed & Health Sci, Hiroshima Univ, Japan.
| |
Collapse
|
7
|
Montanaro R, Vellecco V, Torregrossa R, Casillo GM, Manzo OL, Mitidieri E, Bucci M, Castaldo S, Sorrentino R, Whiteman M, Smimmo M, Carriero F, Terrazzano G, Cirino G, d'Emmanuele di Villa Bianca R, Brancaleone V. Hydrogen sulfide donor AP123 restores endothelial nitric oxide-dependent vascular function in hyperglycemia via a CREB-dependent pathway. Redox Biol 2023; 62:102657. [PMID: 36913800 PMCID: PMC10025109 DOI: 10.1016/j.redox.2023.102657] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/06/2023] Open
Abstract
Diabetes is associated with severe vascular complications involving the impairment of endothelial nitric oxide synthase (eNOS) as well as cystathionine γ-lyase (CSE) activity. eNOS function is suppressed in hyperglycaemic conditions, resulting in reduced NO bioavailability, which is paralleled by reduced levels of hydrogen sulfide (H2S). Here we have addressed the molecular basis of the interplay between the eNOS and CSE pathways. We tested the impact of H2S replacement by using the mitochondrial-targeted H2S donor AP123 in isolated vessels and cultured endothelial cells in high glucose (HG) environment, at concentrations not causing any vasoactive effect per se. Aorta exposed to HG displayed a marked reduction of acetylcholine (Ach)-induced vasorelaxation that was restored by the addition of AP123 (10 nM). In HG condition, bovine aortic endothelial cells (BAEC) showed reduced NO levels, downregulation of eNOS expression, and suppression of CREB activation (p-CREB). Similar results were obtained by treating BAEC with propargylglycine (PAG), an inhibitor of CSE. AP123 treatment rescued eNOS expression, as well as NO levels, and restored p-CREB expression in both the HG environment and the presence of PAG. This effect was mediated by a PI3K-dependent activity since wortmannin (PI3K inhibitor) blunted the rescuing effects operated by the H2S donor. Experiments performed in the aorta of CSE-/- mice confirmed that reduced levels of H2S not only negatively affect the CREB pathway but also impair Ach-induced vasodilation, significantly ameliorated by AP123. We have demonstrated that the endothelial dysfunction due to HG involves H2S/PI3K/CREB/eNOS route, thus highlighting a novel aspect of the H2S/NO interplay in the vasoactive response.
Collapse
Affiliation(s)
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - Sigismondo Castaldo
- U.O.C.Ricerca Formazione & Cooperazione Internazionale, A.O.R.N." Antonio Cardarelli", Naples, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, 80131, Naples, Italy
| | | | - Martina Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Flavia Carriero
- Department of Science, University of Basilicata, Potenza, Italy
| | | | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | | |
Collapse
|
8
|
Tehrani AY, Zhao R, Donen G, Bernatchez P. Heterogenous improvements in endothelial function by sub-blood pressure lowering doses of ARBs result in major anti-aortic root remodeling effects. Nitric Oxide 2023; 131:18-25. [PMID: 36565741 DOI: 10.1016/j.niox.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Low basal nitric oxide (NO) production is associated with a dysfunctional endothelium and vascular diseases. We have shown that some angiotensin II (AngII) receptor type 1 (AT1R) blockers (ARBs), a group of clinic-approved blood pressure (BP)-lowering medications, are also capable of activating endothelial function acutely and chronically, both ex vivo and in vivo, in pleiotropic, AngII-independent fashions, which suggested that endothelial function enhancement with ARBs may be independent of their well-documented BP lowering properties. Herein, we attempt to identify the most potent ARB at activating endothelial function when administered at sub-BP-lowering doses and determine its anti-aortic root remodeling properties in a model of Marfan syndrome (MFS). Amongst the 8 clinically available ARBs tested, only telmisartan and azilsartan induced significant (70% and 49%, respectively) NO-dependent inhibition of aortic contractility when administered for 4 weeks at sub-BP lowering, EC5 doses. Low-dose telmisartan (0.47 mg/kg) attenuated MFS-associated aortic root widening, medial thickening, and elastic fiber fragmentation to the same degree as high-dose telmisartan (10 mg/kg) despite wide differences in BP lowering between the two doses. Our study suggests that telmisartan is the most potent ARB at promoting increased endothelial function at low sub-BP doses and that it retained major aortic root widening inhibition activities. ARBs may enhance endothelial function independently from BP-lowering pathways, which could lead to new therapeutic approaches.
Collapse
Affiliation(s)
- Arash Y Tehrani
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Roy Zhao
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Graham Donen
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Pascal Bernatchez
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
9
|
Li Z, He Y, He H, Zhou W, Li M, Lu A, Che T, Shen S. Purification identification and function analysis of ACE inhibitory peptide from Ulva prolifera protein. Food Chem 2023; 401:134127. [DOI: 10.1016/j.foodchem.2022.134127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
|
10
|
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]
|
11
|
Pinilla I, Maneu V, Campello L, Fernández-Sánchez L, Martínez-Gil N, Kutsyr O, Sánchez-Sáez X, Sánchez-Castillo C, Lax P, Cuenca N. Inherited Retinal Dystrophies: Role of Oxidative Stress and Inflammation in Their Physiopathology and Therapeutic Implications. Antioxidants (Basel) 2022; 11:antiox11061086. [PMID: 35739983 PMCID: PMC9219848 DOI: 10.3390/antiox11061086] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a large group of genetically and clinically heterogeneous diseases characterized by the progressive degeneration of the retina, ultimately leading to loss of visual function. Oxidative stress and inflammation play fundamental roles in the physiopathology of these diseases. Photoreceptor cell death induces an inflammatory state in the retina. The activation of several molecular pathways triggers different cellular responses to injury, including the activation of microglia to eliminate debris and recruit inflammatory cells from circulation. Therapeutical options for IRDs are currently limited, although a small number of patients have been successfully treated by gene therapy. Many other therapeutic strategies are being pursued to mitigate the deleterious effects of IRDs associated with oxidative metabolism and/or inflammation, including inhibiting reactive oxygen species’ accumulation and inflammatory responses, and blocking autophagy. Several compounds are being tested in clinical trials, generating great expectations for their implementation. The present review discusses the main death mechanisms that occur in IRDs and the latest therapies that are under investigation.
Collapse
Affiliation(s)
- Isabel Pinilla
- Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa, University Hospital, 50009 Zaragoza, Spain
- Department of Surgery, University of Zaragoza, 50009 Zaragoza, Spain
- Correspondence: (I.P.); (V.M.)
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Correspondence: (I.P.); (V.M.)
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Laura Fernández-Sánchez
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
| | - Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Pedro Lax
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Nicolás Cuenca
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| |
Collapse
|
12
|
Schneckmann R, Suvorava T, Hundhausen C, Schuler D, Lorenz C, Freudenberger T, Kelm M, Fischer JW, Flögel U, Grandoch M. Endothelial Hyaluronan Synthase 3 Augments Postischemic Arteriogenesis Through CD44/eNOS Signaling. Arterioscler Thromb Vasc Biol 2021; 41:2551-2562. [PMID: 34380333 DOI: 10.1161/atvbaha.121.315478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objective: The dominant driver of arteriogenesis is elevated shear stress sensed by the endothelial glycocalyx thereby promoting arterial outward remodeling. Hyaluronan, a critical component of the endothelial glycocalyx, is synthesized by 3 HAS isoenzymes (hyaluronan synthases 1-3) at the plasma membrane. Considering further the importance of HAS3 for smooth muscle cell and immune cell functions we aimed to evaluate its role in collateral artery growth. Approach and Results: Male Has3-deficient (Has3-KO) mice were subjected to hindlimb ischemia. Blood perfusion was monitored by laser Doppler perfusion imaging and endothelial function was assessed by measurement of flow-mediated dilation in vivo. Collateral remodeling was monitored by high resolution magnetic resonance angiography. A neutralizing antibody against CD44 (clone KM201) was injected intraperitoneally to analyze hyaluronan signaling in vivo. After hindlimb ischemia, Has3-KO mice showed a reduced arteriogenic response with decreased collateral remodeling and impaired perfusion recovery. While postischemic leukocyte infiltration was unaffected, a diminished flow-mediated dilation pointed towards an impaired endothelial cell function. Indeed, endothelial AKT (protein kinase B)-dependent eNOS (endothelial nitric oxide synthase) phosphorylation at Ser1177 was substantially reduced in Has3-KO thigh muscles. Endothelial-specific Has3-KO mice mimicked the hindlimb ischemia-induced phenotype of impaired perfusion recovery as observed in global Has3-deficiency. Mechanistically, blocking selectively the hyaluronan binding site of CD44 reduced flow-mediated dilation, thereby suggesting hyaluronan signaling through CD44 as the underlying signaling pathway. Conclusions: In summary, HAS3 contributes to arteriogenesis in hindlimb ischemia by hyaluronan/CD44-mediated stimulation of eNOS phosphorylation at Ser1177. Thus, strategies augmenting endothelial HAS3 or CD44 could be envisioned to enhance vascularization under pathological conditions.
Collapse
Affiliation(s)
- Rebekka Schneckmann
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Tatsiana Suvorava
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Christian Hundhausen
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Dominik Schuler
- Clinic for Cardiology, Pneumology and Angiology (D.S., M.K.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Christin Lorenz
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Till Freudenberger
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Malte Kelm
- Clinic for Cardiology, Pneumology and Angiology (D.S., M.K.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.K., J.W.F.)
| | - Jens W Fischer
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, University Hospital Düsseldorf, Heinrich-Heine-University, Germany (M.K., J.W.F.)
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology (U.F.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| | - Maria Grandoch
- Institute for Pharmacology and Clinical Pharmacology, Medical Faculty (R.S., T.S., C.H., C.L., T.F., J.W.F., M.G.), University Clinics and Heinrich-Heine University Düsseldorf, Germany
| |
Collapse
|
13
|
Leo F, Suvorava T, Heuser SK, Li J, LoBue A, Barbarino F, Piragine E, Schneckmann R, Hutzler B, Good ME, Fernandez BO, Vornholz L, Rogers S, Doctor A, Grandoch M, Stegbauer J, Weitzberg E, Feelisch M, Lundberg JO, Isakson BE, Kelm M, Cortese-Krott MM. Red Blood Cell and Endothelial eNOS Independently Regulate Circulating Nitric Oxide Metabolites and Blood Pressure. Circulation 2021; 144:870-889. [PMID: 34229449 PMCID: PMC8529898 DOI: 10.1161/circulationaha.120.049606] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 06/22/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Current paradigms suggest that nitric oxide (NO) produced by endothelial cells (ECs) through endothelial nitric oxide synthase (eNOS) in the vessel wall is the primary regulator of blood flow and blood pressure. However, red blood cells (RBCs) also carry a catalytically active eNOS, but its role is controversial and remains undefined. This study aimed to elucidate the functional significance of RBC eNOS compared with EC eNOS for vascular hemodynamics and nitric oxide metabolism. METHODS We generated tissue-specific loss- and gain-of-function models for eNOS by using cell-specific Cre-induced gene inactivation or reactivation. We created 2 founder lines carrying a floxed eNOS (eNOSflox/flox) for Cre-inducible knockout (KO), and gene construct with an inactivated floxed/inverted exon (eNOSinv/inv) for a Cre-inducible knock-in (KI), which respectively allow targeted deletion or reactivation of eNOS in erythroid cells (RBC eNOS KO or RBC eNOS KI mice) or in ECs (EC eNOS KO or EC eNOS KI mice). Vascular function, hemodynamics, and nitric oxide metabolism were compared ex vivo and in vivo. RESULTS The EC eNOS KOs exhibited significantly impaired aortic dilatory responses to acetylcholine, loss of flow-mediated dilation, and increased systolic and diastolic blood pressure. RBC eNOS KO mice showed no alterations in acetylcholine-mediated dilation or flow-mediated dilation but were hypertensive. Treatment with the nitric oxide synthase inhibitor Nγ-nitro-l-arginine methyl ester further increased blood pressure in RBC eNOS KOs, demonstrating that eNOS in both ECs and RBCs contributes to blood pressure regulation. Although both EC eNOS KOs and RBC eNOS KOs had lower plasma nitrite and nitrate concentrations, the levels of bound NO in RBCs were lower in RBC eNOS KOs than in EC eNOS KOs. Reactivation of eNOS in ECs or RBCs rescues the hypertensive phenotype of the eNOSinv/inv mice, whereas the levels of bound NO were restored only in RBC eNOS KI mice. CONCLUSIONS These data reveal that eNOS in ECs and RBCs contribute independently to blood pressure homeostasis.
Collapse
Affiliation(s)
- Francesca Leo
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Cardiology Pneumology and Angiology (T.S., M.K., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sophia K. Heuser
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Junjie Li
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anthea LoBue
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Frederik Barbarino
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Pharmacy, University of Pisa, Italy (F.P.)
| | - Eugenia Piragine
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rebekka Schneckmann
- Department of Pharmacology and Clinical Pharmacology (R.S., M.G.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Beate Hutzler
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Miranda E. Good
- Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville (M.E.G., B.E.I.)
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (M.E.G.)
| | - Bernadette O. Fernandez
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (B.O.F.)
| | - Lukas Vornholz
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stephen Rogers
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore (S.R., A.D.)
| | - Allan Doctor
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore (S.R., A.D.)
| | - Maria Grandoch
- Department of Pharmacology and Clinical Pharmacology (R.S., M.G.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology (J.S.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (E.W., J.O.L., M.M.C.-K.)
| | - Martin Feelisch
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Pharmacology and Clinical Pharmacology (R.S., M.G.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Nephrology (J.S.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Cardiology Pneumology and Angiology (T.S., M.K., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf (M.K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Pharmacy, University of Pisa, Italy (F.P.)
- Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville (M.E.G., B.E.I.)
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (M.E.G.)
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (B.O.F.)
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore (S.R., A.D.)
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (E.W., J.O.L., M.M.C.-K.)
| | - Jon O. Lundberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (E.W., J.O.L., M.M.C.-K.)
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville (M.E.G., B.E.I.)
| | - Malte Kelm
- Department of Cardiology Pneumology and Angiology (T.S., M.K., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf (M.K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Miriam M. Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology (F.L., T.S., S.K.H., J.L., A.L.B., F.B., E.P., B.H., L.V., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Cardiology Pneumology and Angiology (T.S., M.K., M.M.C.-K.), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (E.W., J.O.L., M.M.C.-K.)
| |
Collapse
|
14
|
Zhou Y, Ariotti N, Rae J, Liang H, Tillu V, Tee S, Bastiani M, Bademosi AT, Collins BM, Meunier FA, Hancock JF, Parton RG. Caveolin-1 and cavin1 act synergistically to generate a unique lipid environment in caveolae. J Cell Biol 2021; 220:211716. [PMID: 33496726 PMCID: PMC7844427 DOI: 10.1083/jcb.202005138] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/20/2020] [Accepted: 12/21/2020] [Indexed: 01/09/2023] Open
Abstract
Caveolae are specialized domains of the vertebrate cell surface with a well-defined morphology and crucial roles in cell migration and mechanoprotection. Unique compositions of proteins and lipids determine membrane architectures. The precise caveolar lipid profile and the roles of the major caveolar structural proteins, caveolins and cavins, in selectively sorting lipids have not been defined. Here, we used quantitative nanoscale lipid mapping together with molecular dynamic simulations to define the caveolar lipid profile. We show that caveolin-1 (CAV1) and cavin1 individually sort distinct plasma membrane lipids. Intact caveolar structures composed of both CAV1 and cavin1 further generate a unique lipid nano-environment. The caveolar lipid sorting capability includes selectivities for lipid headgroups and acyl chains. Because lipid headgroup metabolism and acyl chain remodeling are tightly regulated, this selective lipid sorting may allow caveolae to act as transit hubs to direct communications among lipid metabolism, vesicular trafficking, and signaling.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston, TX
| | - Nicholas Ariotti
- University of New South Wales Sydney, Mark Wainwright Analytical Center, Sydney, New South Wales, Australia.,University of New South Wales Sydney, Department of Pathology, School of Medical Sciences, Kensington, Sydney, New South Wales, Australia
| | - James Rae
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Hong Liang
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston, TX
| | - Vikas Tillu
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Shern Tee
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - Michele Bastiani
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Adekunle T Bademosi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Brett M Collins
- University of New South Wales Sydney, Department of Pathology, School of Medical Sciences, Kensington, Sydney, New South Wales, Australia
| | - Frederic A Meunier
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - John F Hancock
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston, TX.,Program in Cell and Regulatory Biology, University of Texas Graduate School of Biomedical Sciences, Houston, TX
| | - Robert G Parton
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia.,The University of Queensland, Centre for Microscopy and Microanalysis, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
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
|
16
|
Alghanem AF, Abello J, Maurer JM, Kumar A, Ta CM, Gunasekar SK, Fatima U, Kang C, Xie L, Adeola O, Riker M, Elliot-Hudson M, Minerath RA, Grueter CE, Mullins RF, Stratman AN, Sah R. The SWELL1-LRRC8 complex regulates endothelial AKT-eNOS signaling and vascular function. eLife 2021; 10:61313. [PMID: 33629656 PMCID: PMC7997661 DOI: 10.7554/elife.61313] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
The endothelium responds to numerous chemical and mechanical factors in regulating vascular tone, blood pressure, and blood flow. The endothelial volume-regulated anion channel (VRAC) has been proposed to be mechanosensitive and thereby sense fluid flow and hydrostatic pressure to regulate vascular function. Here, we show that the leucine-rich repeat-containing protein 8a, LRRC8A (SWELL1), is required for VRAC in human umbilical vein endothelial cells (HUVECs). Endothelial LRRC8A regulates AKT-endothelial nitric oxide synthase (eNOS) signaling under basal, stretch, and shear-flow stimulation, forms a GRB2-Cav1-eNOS signaling complex, and is required for endothelial cell alignment to laminar shear flow. Endothelium-restricted Lrrc8a KO mice develop hypertension in response to chronic angiotensin-II infusion and exhibit impaired retinal blood flow with both diffuse and focal blood vessel narrowing in the setting of type 2 diabetes (T2D). These data demonstrate that LRRC8A regulates AKT-eNOS in endothelium and is required for maintaining vascular function, particularly in the setting of T2D.
Collapse
Affiliation(s)
- Ahmad F Alghanem
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States.,Eastern Region, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Al Hasa, Saudi Arabia
| | - Javier Abello
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, United States
| | - Joshua M Maurer
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Chau My Ta
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Susheel K Gunasekar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Urooj Fatima
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Chen Kang
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Litao Xie
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Oluwaseun Adeola
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Megan Riker
- Department of Ophthalmology, University of Iowa, Carver College of Medicine, Iowa City, United States
| | - Macaulay Elliot-Hudson
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Rachel A Minerath
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Chad E Grueter
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Robert F Mullins
- Department of Ophthalmology, University of Iowa, Carver College of Medicine, Iowa City, United States
| | - Amber N Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, United States
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States.,Center for Cardiovascular Research, Washington University, St Louis, United States
| |
Collapse
|
17
|
Bhatia V, Elnagary L, Dakshinamurti S. Tracing the path of inhaled nitric oxide: Biological consequences of protein nitrosylation. Pediatr Pulmonol 2021; 56:525-538. [PMID: 33289321 DOI: 10.1002/ppul.25201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/28/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a comprehensive regulator of vascular and airway tone. Endogenous NO produced by nitric oxide synthases regulates multiple signaling cascades, including activation of soluble guanylate cyclase to generate cGMP, relaxing smooth muscle cells. Inhaled NO is an established therapy for pulmonary hypertension in neonates, and has been recently proposed for the treatment of hypoxic respiratory failure and acute respiratory distress syndrome due to COVID-19. In this review, we summarize the effects of endogenous and exogenous NO on protein S-nitrosylation, which is the selective and reversible covalent attachment of a nitrogen monoxide group to the thiol side chain of cysteine. This posttranslational modification targets specific cysteines based on the acid/base sequence of surrounding residues, with significant impacts on protein interactions and function. S-nitrosothiol (SNO) formation is tightly compartmentalized and enzymatically controlled, but also propagated by nonenzymatic transnitrosylation of downstream protein targets. Redox-based nitrosylation and denitrosylation pathways dynamically regulate the equilibrium of SNO-proteins. We review the physiological roles of SNO proteins, including nitrosohemoglobin and autoregulation of blood flow through hypoxic vasodilation, and pathological effects of nitrosylation including inhibition of critical vasodilator enzymes; and discuss the intersection of NO source and dose with redox environment, in determining the effects of protein nitrosylation.
Collapse
Affiliation(s)
- Vikram Bhatia
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Lara Elnagary
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Shyamala Dakshinamurti
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada.,Section of Neonatology, Departments of Pediatrics and Physiology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
18
|
Krishnan Y, Zou J, Jani MS. Quantitative Imaging of Biochemistry in Situ and at the Nanoscale. ACS CENTRAL SCIENCE 2020; 6:1938-1954. [PMID: 33274271 PMCID: PMC7706076 DOI: 10.1021/acscentsci.0c01076] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Indexed: 05/12/2023]
Abstract
Biochemical reactions in eukaryotic cells occur in subcellular, membrane-bound compartments called organelles. Each kind of organelle is characterized by a unique lumenal chemical composition whose stringent regulation is vital to proper organelle function. Disruption of the lumenal ionic content of organelles is inextricably linked to disease. Despite their vital roles in cellular homeostasis, there are large gaps in our knowledge of organellar chemical composition largely from a lack of suitable probes. In this Outlook, we describe how, using organelle-targeted ratiometric probes, one can quantitatively image the lumenal chemical composition and biochemical activity inside organelles. We discuss how excellent fluorescent detection chemistries applied largely to the cytosol may be expanded to study organelles by chemical imaging at subcellular resolution in live cells. DNA-based reporters are a new and versatile platform to enable such approaches because the resultant probes have precise ratiometry and accurate subcellular targeting and are able to map multiple chemicals simultaneously. Quantitatively mapping lumenal ions and biochemical activity can drive the discovery of new biology and biomedical applications.
Collapse
Affiliation(s)
| | - Junyi Zou
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
- Grossman Institute of Neuroscience,
Quantitative Biology and Human Behavior, University of Chicago, Chicago, Illinois 60637, United States
| | - Maulik S. Jani
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
- Grossman Institute of Neuroscience,
Quantitative Biology and Human Behavior, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
19
|
Luchetti F, Crinelli R, Nasoni MG, Benedetti S, Palma F, Fraternale A, Iuliano L. LDL receptors, caveolae and cholesterol in endothelial dysfunction: oxLDLs accomplices or victims? Br J Pharmacol 2020; 178:3104-3114. [PMID: 32986849 DOI: 10.1111/bph.15272] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Oxidized LDLs (oxLDLs) and oxysterols play a key role in endothelial dysfunction and the development of atherosclerosis. The loss of vascular endothelium function negatively impacts vasomotion, cell growth, adhesiveness and barrier functions. While for some of these disturbances, a reasonable explanation can be provided from a mechanistic standpoint, for many others, the molecular mediators that are involved are unknown. Caveolae, specific plasma membrane domains, have recently emerged as targets and mediators of oxLDL-induced endothelial dysfunction. Caveolae and their associated protein caveolin-1 (Cav-1) are involved in oxLDLs/LDLs transcytosis, mainly through the scavenger receptor class B type 1 (SR-B1 or SCARB1). In contrast, oxLDLs endocytosis is mediated by the lectin-like oxidized LDL receptor 1 (LOX-1), whose activity depends on an intact caveolae system. In addition, LOX-1 regulates the expression of Cav-1 and vice versa. On the other hand, oxLDLs may affect cholesterol plasma membrane content/distribution thus influencing caveolae architecture, Cav-1 localization and the associated signalling. Overall, the evidence indicate that caveolae have both active and passive roles in oxLDL-induced endothelial cell dysfunction. First, as mediators of lipid uptake and transfer in the subendothelial space and, later, as targets of changes in composition/dynamics of plasma membrane lipids resulting from increased levels of circulating oxLDLs. Gaining a better understanding of how oxLDLs interact with endothelial cells and modulate caveolae-mediated signalling pathways, leading to endothelial dysfunction, is crucial to find new targets for intervention to tackle atherosclerosis and the related clinical entities. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Serena Benedetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesco Palma
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | - Luigi Iuliano
- Department of Medico-Surgical Sciences and Biotechnologies Vascular Biology, Atherothrombosis & Mass Spectrometry, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
20
|
Sun Y, Lu Y, Saredy J, Wang X, Drummer Iv C, Shao Y, Saaoud F, Xu K, Liu M, Yang WY, Jiang X, Wang H, Yang X. ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol 2020; 37:101696. [PMID: 32950427 PMCID: PMC7767745 DOI: 10.1016/j.redox.2020.101696] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are critical for the progression of cardiovascular diseases, inflammations and tumors. However, the mechanisms of how ROS sense metabolic stress, regulate metabolic pathways and initiate proliferation, inflammation and cell death responses remain poorly characterized. In this analytic review, we concluded that: 1) Based on different features and functions, eleven types of ROS can be classified into seven functional groups: metabolic stress-sensing, chemical connecting, organelle communication, stress branch-out, inflammasome-activating, dual functions and triple functions ROS. 2) Among the ROS generation systems, mitochondria consume the most amount of oxygen; and nine types of ROS are generated; thus, mitochondrial ROS systems serve as the central hub for connecting ROS with inflammasome activation, trained immunity and immunometabolic pathways. 3) Increased nuclear ROS production significantly promotes cell death in comparison to that in other organelles. Nuclear ROS systems serve as a convergent hub and decision-makers to connect unbearable and alarming metabolic stresses to inflammation and cell death. 4) Balanced ROS levels indicate physiological homeostasis of various metabolic processes in subcellular organelles and cytosol, while imbalanced ROS levels present alarms for pathological organelle stresses in metabolic processes. Based on these analyses, we propose a working model that ROS systems are a new integrated network for sensing homeostasis and alarming stress in metabolic processes in various subcellular organelles. Our model provides novel insights on the roles of the ROS systems in bridging metabolic stress to inflammation, cell death and tumorigenesis; and provide novel therapeutic targets for treating those diseases. (Word count: 246).
Collapse
Affiliation(s)
- Yu Sun
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Yifan Lu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Jason Saredy
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xianwei Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Charles Drummer Iv
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ying Shao
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Fatma Saaoud
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Keman Xu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ming Liu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - William Y Yang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
21
|
Ottolini M, Daneva Z, Chen YL, Cope EL, Kasetti RB, Zode GS, Sonkusare SK. Mechanisms underlying selective coupling of endothelial Ca 2+ signals with eNOS vs. IK/SK channels in systemic and pulmonary arteries. J Physiol 2020; 598:3577-3596. [PMID: 32463112 DOI: 10.1113/jp279570] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Endothelial cell TRPV4 (TRPV4EC ) channels exert a dilatory effect on the resting diameter of resistance mesenteric and pulmonary arteries. Functional intermediate- and small-conductance K+ (IK and SK) channels and endothelial nitric oxide synthase (eNOS) are present in the endothelium of mesenteric and pulmonary arteries. TRPV4EC sparklets preferentially couple with IK/SK channels in mesenteric arteries and with eNOS in pulmonary arteries. TRPV4EC channels co-localize with IK/SK channels in mesenteric arteries but not in pulmonary arteries, which may explain TRPV4EC -IK/SK channel coupling in mesenteric arteries and its absence in pulmonary arteries. The presence of the nitric oxide-scavenging protein, haemoglobin α, limits TRPV4EC -eNOS signalling in mesenteric arteries. Spatial proximity of TRPV4EC channels with eNOS and the absence of haemoglobin α favour TRPV4EC -eNOS signalling in pulmonary arteries. ABSTRACT Spatially localized Ca2+ signals activate Ca2+ -sensitive intermediate- and small-conductance K+ (IK and SK) channels in some vascular beds and endothelial nitric oxide synthase (eNOS) in others. The present study aimed to uncover the signalling organization that determines selective Ca2+ signal to vasodilatory target coupling in the endothelium. Resistance-sized mesenteric arteries (MAs) and pulmonary arteries (PAs) were used as prototypes for arteries with predominantly IK/SK channel- and eNOS-dependent vasodilatation, respectively. Ca2+ influx signals through endothelial transient receptor potential vanilloid 4 (TRPV4EC ) channels played an important role in controlling the baseline diameter of both MAs and PAs. TRPV4EC channel activity was similar in MAs and PAs. However, the TRPV4 channel agonist GSK1016790A (10 nm) selectively activated IK/SK channels in MAs and eNOS in PAs, revealing preferential TRPV4EC -IK/SK channel coupling in MAs and TRPV4EC -eNOS coupling in PAs. IK/SK channels co-localized with TRPV4EC channels at myoendothelial projections (MEPs) in MAs, although they lacked the spatial proximity necessary for their activation by TRPV4EC channels in PAs. Additionally, the presence of the NO scavenging protein haemoglobin α (Hbα) within nanometer proximity to eNOS limits TRPV4EC -eNOS signalling in MAs. By contrast, co-localization of TRPV4EC channels and eNOS at MEPs, and the absence of Hbα, favour TRPV4EC -eNOS coupling in PAs. Thus, our results reveal that differential spatial organization of signalling elements determines TRPV4EC -IK/SK vs. TRPV4EC -eNOS coupling in resistance arteries.
Collapse
Affiliation(s)
- Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA.,Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, USA
| | - Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA
| | - Eric L Cope
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA
| | - Ramesh B Kasetti
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Gulab S Zode
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA.,Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, USA.,Department of Molecular Physiology and Biological Physics, University of Virginia-School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
22
|
Wei X, Guan L, Fan P, Liu X, Liu R, Liu Y, Bai H. Direct Current Electric Field Stimulates Nitric Oxide Production and Promotes NO-Dependent Angiogenesis: Involvement of the PI3K/Akt Signaling Pathway. J Vasc Res 2020; 57:195-205. [PMID: 32375152 DOI: 10.1159/000506517] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/13/2020] [Indexed: 02/05/2023] Open
Abstract
Electric fields (EFs) promote angiogenesis in vitro and in vivo. These results indicate the feasibility of the application of EFs to modulate angiogenesis. Nitric oxide (NO) derived from endothelial nitric oxide synthase (eNOS) is an important regulator of angiogenesis. However, the role of direct current EFs in eNOS activity and expression in association with angiogenesis of endothelial cells has not been investigated. In the present study, we stimulated human umbilical vein endothelial cells (HUVECs) with EFs and evaluated the activity and expression of eNOS. EFs induced significant phosphorylation of eNOS, upregulation of the expression of eNOS protein, and an increase in NO production from HUVECs. L-NAME, a specific inhibitor of eNOS, abolished EF-induced HUVEC angiogenesis. EFs stimulated Akt activation. Inhibition of PI3K activity inhibited EF-mediated Akt and eNOS activation and inhibited NO production in the endothelial cells. Moreover, EFs stimulated HUVEC proliferation and enhanced the S phase cell population of the cell cycle. We conclude that EFs stimulate eNOS activation and NO production via a PI3K/Akt-dependent pathway. Thus, activation of eNOS appears to be one of the key signaling pathways necessary for EF-mediated angiogenesis. These novel findings suggest that NO signaling may have an important role in EF-mediated endothelial cell function.
Collapse
Affiliation(s)
- Xing Wei
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Division of Peptides Related to Human Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China,
| |
Collapse
|
23
|
Jani MS, Zou J, Veetil AT, Krishnan Y. A DNA-based fluorescent probe maps NOS3 activity with subcellular spatial resolution. Nat Chem Biol 2020; 16:660-666. [PMID: 32152543 DOI: 10.1038/s41589-020-0491-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 12/05/2019] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Nitric oxide synthase 3 (NOS3) produces the gasotransmitter nitric oxide (NO), which drives critical cellular signaling pathways by S-nitrosylating target proteins. Endogenous NOS3 resides at two distinct subcellular locations: the plasma membrane and the trans-Golgi network (TGN). However, NO generation arising from the activities of both these pools of NOS3 and its relative contribution to physiology or disease is not yet resolvable. We describe a fluorescent DNA-based probe technology, NOckout, that can be targeted either to the plasma membrane or the TGN, where it can quantitatively map the activities of endogenous NOS3 at these locations in live cells. We found that, although NOS3 at the Golgi is tenfold less active than at the plasma membrane, its activity is essential for the structural integrity of the Golgi. The newfound ability to spatially map NOS3 activity provides a platform to discover selective regulators of the distinct pools of NOS3.
Collapse
Affiliation(s)
- Maulik S Jani
- Department of Chemistry, University of Chicago, Chicago, IL, USA.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL, USA
| | - Junyi Zou
- Department of Chemistry, University of Chicago, Chicago, IL, USA.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL, USA
| | - Aneesh T Veetil
- Department of Chemistry, University of Chicago, Chicago, IL, USA.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL, USA
| | - Yamuna Krishnan
- Department of Chemistry, University of Chicago, Chicago, IL, USA. .,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
24
|
Jin SW, Pham HT, Choi JH, Lee GH, Han EH, Cho YH, Chung YC, Kim YH, Jeong HG. Impressic Acid, a Lupane-Type Triterpenoid from Acanthopanax koreanum, Attenuates TNF-α-Induced Endothelial Dysfunction via Activation of eNOS/NO Pathway. Int J Mol Sci 2019; 20:ijms20225772. [PMID: 31744135 PMCID: PMC6888592 DOI: 10.3390/ijms20225772] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is one of the most reported diseases worldwide, and extensive research and trials are focused on the discovery and utilizing for novel therapeutics. Nitric oxide (NO) is produced mainly by endothelial nitric oxide synthase (eNOS) and it plays a key role in regulating vascular function including systemic blood pressure and vascular inflammation in vascular endothelium. In this study hypothesized that Impressic acid (IPA), a component isolated from Acanthopanax koreanum, acts as an enhancer of eNOS activity and NO production. IPA treatment induced eNOS phosphorylation and NO production, which was correlated with eNOS phosphorylation via the activation of JNK1/2, p38 MAPK, AMPK, and CaMKII. In addition, the induction of eNOS phosphorylation by IPA was attenuated by pharmacological inhibitor of MAPKs, AMPK, and CaMKII. Finally, IPA treatment prevented the adhesion of TNF-α-induced monocytes to endothelial cells and suppressed the TNF-α-stimulated ICAM-1 expression via activation of NF-κB, while treatment with L-NAME, the NOS inhibitor, reversed the inhibitory effect of IPA on TNF-α-induced ICAM-1 expression via activation of NF-κB. Taken together, these findings show that IPA protects against TNF-α-induced vascular endothelium dysfunction through attenuation of the NF-κB pathway by activating eNOS/NO pathway in endothelial cells.
Collapse
Affiliation(s)
- Sun Woo Jin
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (S.W.J.); (H.T.P.); (J.H.C.); (G.H.L.); (Y.H.K.)
| | - Hoa Thi Pham
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (S.W.J.); (H.T.P.); (J.H.C.); (G.H.L.); (Y.H.K.)
| | - Jae Ho Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (S.W.J.); (H.T.P.); (J.H.C.); (G.H.L.); (Y.H.K.)
| | - Gi Ho Lee
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (S.W.J.); (H.T.P.); (J.H.C.); (G.H.L.); (Y.H.K.)
| | - Eun Hee Han
- Drug & Disease Target Research Team, Division of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Cheongju 28119, Korea;
| | - Young Ho Cho
- Department of Pharmaceutics & Biotechnology, College of Medical Engineering, Konyang University, Daejeon 35365, Korea;
| | - Young Chul Chung
- Department of Food Science, International University of Korea, Jinju, 52833, Korea;
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (S.W.J.); (H.T.P.); (J.H.C.); (G.H.L.); (Y.H.K.)
| | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (S.W.J.); (H.T.P.); (J.H.C.); (G.H.L.); (Y.H.K.)
- Correspondence: ; Tel.: +82-42-821-5936
| |
Collapse
|
25
|
Koch SR, Choi H, Mace EH, Stark RJ. Toll-like receptor 3-mediated inflammation by p38 is enhanced by endothelial nitric oxide synthase knockdown. Cell Commun Signal 2019; 17:33. [PMID: 30987646 PMCID: PMC6466662 DOI: 10.1186/s12964-019-0345-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/21/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Vascular dysfunction is commonly seen during severe viral infections. Endothelial nitric oxide synthase (eNOS), has been postulated to play an important role in regulating vascular homeostasis as well as propagation of the inflammatory reaction. We hypothesized that the loss of eNOS would negatively impact toll-like receptor 3 (TLR3) signaling and worsen vascular function to viral challenge. METHODS Human microvascular endothelial cells (HMVECs) were exposed to either control or eNOS siRNA and then treated with Poly I:C, a TLR3 agonist and mimicker of dsRNA viruses. Cells were assessed for protein-protein associations, cytokine and chemokine analysis as well as transendothelial electrical resistance (TEER) as a surrogate of permeability. RESULTS HMVECs that had reduced eNOS expression had a significantly elevated increase in IL-6, IL-8 and IP-10 production after Poly I:C. In addition, the knockdown of eNOS enhanced the change in TEER after Poly I:C stimulation. Western blot analysis showed enhanced phosphorylation of p38 in sieNOS treated cells with Poly I:C compared to siControl cells. Proximity ligation assays further demonstrated direct eNOS-p38 protein-protein interactions. The addition of the p38 inhibitor, SB203580, in eNOS knockdown cells reduced both cytokine production after Poly I:C, and as well as mitigated the reduction in TEER, suggesting a direct link between eNOS and p38 in TLR3 signaling. CONCLUSIONS These results suggest that reduction of eNOS increases TLR3-mediated inflammation in human endothelial cells in a p38-dependent manner. This finding has important implications for understanding the pathogenesis of severe viral infections and the associated vascular dysfunction.
Collapse
Affiliation(s)
- Stephen R Koch
- Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, 5121 Doctors' Office Tower, Nashville, TN, 37232-9075, USA
| | - Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, 5121 Doctors' Office Tower, Nashville, TN, 37232-9075, USA
| | - Eric H Mace
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ryan J Stark
- Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, 5121 Doctors' Office Tower, Nashville, TN, 37232-9075, USA.
| |
Collapse
|
26
|
Chennoufi R, Cabrié A, Nguyen NH, Bogliotti N, Simon F, Cinquin B, Tauc P, Boucher JL, Slama-Schwok A, Xie J, Deprez E. Light-induced formation of NO in endothelial cells by photoactivatable NADPH analogues targeting nitric-oxide synthase. Biochim Biophys Acta Gen Subj 2019; 1863:1127-1137. [PMID: 30986510 DOI: 10.1016/j.bbagen.2019.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/03/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nitric-oxide synthases (NOS) catalyze the formation of NO using NADPH as electron donor. We have recently designed and synthesized a new series of two-photon absorbing and photoactivatable NADPH analogues (NT). These compounds bear one or two carboxymethyl group(s) on the 2'- or/and 3'-position(s) of the ribose in the adenosine moiety, instead of a 2'-phosphate group, and differ by the nature of the electron donor in their photoactivatable chromophore (replacing the nicotinamide moiety). Here, we addressed the ability of NTs to photoinduce eNOS-dependent NO production in endothelial cells. METHODS The cellular fate of NTs and their photoinduced effects were studied using multiphoton fluorescence imaging, cell viability assays and a BODIPY-derived NO probe for NO measurements. The eNOS dependence of photoinduced NO production was addressed using two NOS inhibitors (NS1 and L-NAME) targeting the reductase and the oxygenase domains, respectively. RESULTS We found that, two compounds, those bearing a single carboxymethyl group on the 3'-position of the ribose, colocalize with the Golgi apparatus (the main intracellular location of eNOS) and display high intracellular two-photon brightness. Furthermore, a eNOS-dependent photooxidation was observed for these two compounds only, which is accompanied by a substantial intracellular NO production accounting for specific photocytotoxic effects. CONCLUSIONS We show for the first time that NT photoactivation efficiently triggers electron flow at the eNOS level and increases the basal production of NO by endothelial cells. GENERAL SIGNIFICANCE Efficient photoactivatable NADPH analogues targeting NOS could have important implications for generating apoptosis in tumor cells or modulating NO-dependent physiological processes.
Collapse
Affiliation(s)
- Rahima Chennoufi
- LBPA, CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Aimeric Cabrié
- LBPA, CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Nhi Ha Nguyen
- PPSM, CNRS UMR8531, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Nicolas Bogliotti
- PPSM, CNRS UMR8531, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Françoise Simon
- LBPA, CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Bertrand Cinquin
- LBPA, CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Patrick Tauc
- LBPA, CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Jean-Luc Boucher
- Laboratoire de "Chimie et Biochimie Pharmacologiques et Toxicologiques", CNRS UMR8601, Université Paris Descartes, 75270 Paris, France
| | - Anny Slama-Schwok
- Laboratoire de "Stabilité Génétique et Oncogénèse", CNRS UMR8200, Gustave Roussy, Université Paris-Saclay, 94607 Villejuif, France
| | - Juan Xie
- PPSM, CNRS UMR8531, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France
| | - Eric Deprez
- LBPA, CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, F-94235 Cachan, France.
| |
Collapse
|
27
|
Prasher P, Mudila H, Sharma M, Khati B. Developmental perspectives of the drugs targeting enzyme-instigated inflammation: a mini review. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02315-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
28
|
Nuno DW, Lamping KG. Dietary Fatty Acid Saturation Modulates Sphingosine-1-Phosphate-Mediated Vascular Function. J Diabetes Res 2019; 2019:2354274. [PMID: 31534971 PMCID: PMC6732604 DOI: 10.1155/2019/2354274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022] Open
Abstract
Sphingolipids, modified by dietary fatty acids, are integral components of plasma membrane and caveolae that are also vasoactive compounds. We hypothesized that dietary fatty acid saturation affects vasoconstriction to sphingosine-1-phosphate (S1P) through caveolar regulation of rho kinase. Wild type (WT) and caveolin-1-deficient (cav-1 KO) mice which lack vascular caveolae were fed a low-fat diet (LF), 60% high-saturated fat diet (lard, HF), or 60% fat diet with equal amounts of lard and n-3 polyunsaturated menhaden oil (MO). Weight gain of WT on HF and MO diets was similar while markedly blunted in cav-1 KO. Neither high-fat diet affected the expression of cav-1, rho, or rho kinase in arteries from WT. In cav-1 KO, MO increased the vascular expression of rho but had no effect on rho kinase. HF had no effect on rho or rho kinase expression in cav-1 KO. S1P produced a concentration-dependent constriction of gracilis arteries from WT on LF that was reduced with HF and restored to normal with MO. Constriction to S1P was reduced in cav-1 KO and no longer affected by a high-saturated fat diet. Inhibition of rho kinase which reduced constriction to PE independent of diet in arteries from WT and cav-1 KO only reduced constriction to S1P in arteries from WT fed MO. The data suggest that dietary fatty acids modify vascular responses to S1P by a caveolar-dependent mechanism which is enhanced by dietary n-3 polyunsaturated fats.
Collapse
Affiliation(s)
- Daniel W. Nuno
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kathryn G. Lamping
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa City Veterans Affairs Healthcare System, Iowa City, IA, USA
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
29
|
Nuno DW, Coppey LJ, Yorek MA, Lamping KG. Dietary fats modify vascular fat composition, eNOS localization within lipid rafts and vascular function in obesity. Physiol Rep 2018; 6:e13820. [PMID: 30105819 PMCID: PMC6090220 DOI: 10.14814/phy2.13820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
We tested whether dietary fatty acids alter membrane composition shifting localization of signaling pathways within caveolae to determine their role in vascular function. Wild type (WT) and caveolin-1-deficient mice (cav-1 KO), required for vascular caveolae formation, were fed low fat (LF), high saturated fat (HF, 60% kcal from lard), or high-fat diet with 50:50 lard and n-3 polyunsaturated fatty acid-enriched menhaden oil (MO). HF and MO increased body weight and fat in WT but had less effect in cav-1 KO. MO increased unsaturated fatty acids and the unsaturation index of aorta from WT and cav-1 KO. In LF WT aorta, endothelial nitric oxide synthase (eNOS) was localized to cav-1-enriched low-density fractions which shifted to actin-enriched high-density fractions with acetylcholine (ACh). HF and MO shifted eNOS to high-density fractions in WT aorta which was not affected by ACh. In cav-1 KO aorta, eNOS was localized in low-density non-caveolar fractions but not shifted by ACh or diet. Inducible NOS and cyclooxygenase 1/2 were not localized in low-density fractions or affected by diet, ACh or genotype. ACh-induced dilation of gracilis arteries from HF WT was similar to dilation in LF but the NOS component was reduced. In WT and cav-1 KO, dilation to ACh was enhanced by MO through increased role for NOS and cyclooxygenase. We conclude that dietary fats affect vascular fatty acid composition and membrane localization of eNOS but the contribution of eNOS and cyclooxygenase in ACh-mediated vascular responses is independent of lipid rafts.
Collapse
Affiliation(s)
- Daniel W. Nuno
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
| | - Lawrence J. Coppey
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
| | - Mark A. Yorek
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
- Iowa City Veterans Affairs Healthcare SystemIowa CityIowa
| | - Kathryn G. Lamping
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
- Iowa City Veterans Affairs Healthcare SystemIowa CityIowa
- Department of PharmacologyRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
| |
Collapse
|
30
|
Espinosa-Díez C, Miguel V, Vallejo S, Sánchez FJ, Sandoval E, Blanco E, Cannata P, Peiró C, Sánchez-Ferrer CF, Lamas S. Role of glutathione biosynthesis in endothelial dysfunction and fibrosis. Redox Biol 2018; 14:88-99. [PMID: 28888203 PMCID: PMC5596265 DOI: 10.1016/j.redox.2017.08.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022] Open
Abstract
Glutathione (GSH) biosynthesis is essential for cellular redox homeostasis and antioxidant defense. The rate-limiting step requires glutamate-cysteine ligase (GCL), which is composed of the catalytic (GCLc) and the modulatory (GCLm) subunits. To evaluate the contribution of GCLc to endothelial function we generated an endothelial-specific Gclc haplo-insufficient mouse model (Gclc e/+ mice). In murine lung endothelial cells (MLEC) derived from these mice we observed a 50% reduction in GCLc levels compared to lung fibroblasts from the same mice. MLEC obtained from haplo-insufficient mice showed significant reduction in GSH levels as well as increased basal and stimulated ROS levels, reduced phosphorylation of eNOS (Ser 1177) and increased eNOS S-glutathionylation, compared to MLEC from wild type (WT) mice. Studies in mesenteric arteries demonstrated impaired endothelium-dependent vasodilation in Gclc(e/+) male mice, which was corrected by pre-incubation with GSH-ethyl-ester and BH4. To study the contribution of endothelial GSH synthesis to renal fibrosis we employed the unilateral ureteral obstruction model in WT and Gclc(e/+) mice. We observed that obstructed kidneys from Gclc(e/+) mice exhibited increased deposition of fibrotic markers and reduced Nrf2 levels. We conclude that the preservation of endothelial GSH biosynthesis is not only critical for endothelial function but also in anti-fibrotic responses.
Collapse
Affiliation(s)
- Cristina Espinosa-Díez
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa", (CSIC-UAM), Madrid, Spain
| | - Verónica Miguel
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa", (CSIC-UAM), Madrid, Spain
| | - Susana Vallejo
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid and Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), Spain
| | - Francisco J Sánchez
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa", (CSIC-UAM), Madrid, Spain
| | - Elena Sandoval
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa", (CSIC-UAM), Madrid, Spain
| | - Eva Blanco
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa", (CSIC-UAM), Madrid, Spain
| | - Pablo Cannata
- Department of Pathology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spain
| | - Concepción Peiró
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid and Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid and Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), Spain
| | - Santiago Lamas
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa", (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
31
|
Blum-Johnston C, Thorpe RB, Wee C, Opsahl R, Romero M, Murray S, Brunelle A, Blood Q, Wilson R, Blood AB, Zhang L, Longo LD, Pearce WJ, Wilson SM. Long-term hypoxia uncouples Ca 2+ and eNOS in bradykinin-mediated pulmonary arterial relaxation. Am J Physiol Regul Integr Comp Physiol 2018. [PMID: 29513562 DOI: 10.1152/ajpregu.00311.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bradykinin-induced activation of the pulmonary endothelium triggers a rise in intracellular Ca2+ that activates nitric oxide (NO)-dependent vasorelaxation. Chronic hypoxia is commonly associated with increased pulmonary vascular tone, which can cause pulmonary hypertension in responsive individuals. In the present study, we tested the hypothesis that long-term high-altitude hypoxia (LTH) diminishes bradykinin-induced Ca2+ signals and inhibits endothelial nitric oxide synthase (eNOS), prostacyclin (PGI2), and large-conductance K+ (BKCa) channels in sheep, which are moderately responsive to LTH, resulting in decreased pulmonary arterial vasorelaxation. Pulmonary arteries were isolated from ewes kept near sea level (720 m) or at high altitude (3,801 m) for >100 days. Vessel force was measured with wire myography and endothelial intracellular Ca2+ with confocal microscopy. eNOS was inhibited with 100 μM NG-nitro-l-arginine methyl ester (l-NAME), PGI2 production was inhibited with 10 µM indomethacin that inhibits cyclooxygenase, and BKCa channels were blocked with 1 mM tetraethylammonium. Bradykinin-induced endothelial Ca2+ signals increased following LTH, but bradykinin relaxation decreased. Furthermore, some vessels contracted in response to bradykinin after LTH. l-NAME sensitivity decreased, suggesting that eNOS dysfunction played a role in uncoupling Ca2+ signals and bradykinin relaxation. The Ca2+ ionophore A-23187 (10 µM) elicited an enhanced Ca2+ response following LTH while relaxation was unchanged although l-NAME sensitivity increased. Additionally, BKCa function decreased during bradykinin relaxation following LTH. Western analysis showed that BKCa α-subunit expression was increased by LTH while that for the β1 subunit was unchanged. Overall, these results suggest that those even moderately responsive to LTH can have impaired endothelial function.
Collapse
Affiliation(s)
- Carla Blum-Johnston
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California.,Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine , Loma Linda, California
| | - Richard B Thorpe
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Chelsea Wee
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Raechel Opsahl
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Monica Romero
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine , Loma Linda, California
| | - Samuel Murray
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Alexander Brunelle
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Quintin Blood
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Rachael Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Arlin B Blood
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Lawrence D Longo
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - William J Pearce
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Sean M Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California.,Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine , Loma Linda, California
| |
Collapse
|
32
|
Yan B, Sun Y, Wang J. Depletion of ubiA prenyltransferase domain containing 1 expression promotes angiotensin II‑induced hypertrophic response in AC16 human myocardial cells via modulating the expression levels of coenzyme Q10 and endothelial nitric oxide synthase. Mol Med Rep 2017; 16:6910-6915. [PMID: 28901410 DOI: 10.3892/mmr.2017.7407] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 05/22/2017] [Indexed: 11/06/2022] Open
Abstract
UbiA prenyltransferase domain containing 1 (UBIAD1) is closely associated with cardiovascular diseases. However, at the cellular level, little is known about how UBIAD1 is expressed and functions in cardiomyocyte hypertrophy. The aim of the present study was to investigate the expression and role of UBIAD1 in angiotensin II (Ang II)‑induced hypertrophy in AC16 cardiomyoblast cells. The loss‑of‑function approach was used to knock down UBIAD1 in vehicle‑ and Ang II‑stimulated AC16 cells. The levels of atrial natriuretic factor (ANF) and caspase-3 were measured and compared between vehicle‑ and Ang II‑treated AC16 cells pretreated with control siRNA or siRNA against UBIAD1. In addition, the levels of coenzyme Q10 (CoQ10) and endothelial nitric oxide synthase (eNOS) were evaluated and compared between these groups. Ang II induced hypertrophy and apoptosis in AC16 cells, accompanied by increased expression of ANF and caspase-3, and decreased expression of UBIAD1. These effects were potentiated by UBIAD1 knockdown. In addition, Ang II treatment suppressed the expression of CoQ10 and eNOS, as well as the production of NO, and these inhibitory effects were also enhanced by UBIAD1 knockdown. Thus, silencing of UBIAD1 expression promotes a myocardial hypertrophic response to Ang II stimulation, in part, by suppressing the expression of CoQ10 and eNOS.
Collapse
Affiliation(s)
- Bingju Yan
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jun Wang
- Department of Cardiology General Surgery, First Hospital of Liaoning Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
33
|
Keeley TP, Siow RCM, Jacob R, Mann GE. A PP2A-mediated feedback mechanism controls Ca 2+-dependent NO synthesis under physiological oxygen. FASEB J 2017; 31:5172-5183. [PMID: 28760745 PMCID: PMC5690389 DOI: 10.1096/fj.201700211r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
Intracellular O2 is a key regulator of NO signaling, yet most in vitro studies are conducted in atmospheric O2 levels, hyperoxic with respect to the physiologic milieu. We investigated NO signaling in endothelial cells cultured in physiologic (5%) O2 and stimulated with histamine or shear stress. Culture of cells in 5% O2 (>5 d) decreased histamine- but not shear stress–stimulated endothelial (e)NOS activity. Unlike cells adapted to a hypoxic environment (1% O2), those cultured in 5% O2 still mobilized sufficient Ca2+ to activate AMPK. Enhanced expression and membrane targeting of PP2A-C was observed in 5% O2, resulting in greater interaction with eNOS in response to histamine. Moreover, increased dephosphorylation of eNOS in 5% O2 was Ca2+-sensitive and reversed by okadaic acid or PP2A-C siRNA. The present findings establish that Ca2+ mobilization stimulates both NO synthesis and PP2A-mediated eNOS dephosphorylation, thus constituting a novel negative feedback mechanism regulating eNOS activity not present in response to shear stress. This, coupled with enhanced NO bioavailability, underpins differences in NO signaling induced by inflammatory and physiologic stimuli that are apparent only in physiologic O2 levels. Furthermore, an explicit delineation between physiologic normoxia and genuine hypoxia is defined here, with implications for our understanding of pathophysiological hypoxia.—Keeley, T. P., Siow, R. C. M., Jacob, R., Mann, G. E. A PP2A-mediated feedback mechanism controls Ca2+-dependent NO synthesis under physiological oxygen.
Collapse
Affiliation(s)
- Thomas P Keeley
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Richard C M Siow
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Ron Jacob
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Giovanni E Mann
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
34
|
Abstract
SIGNIFICANCE The family of gasotransmitter molecules, nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), has emerged as an important mediator of numerous cellular signal transduction and pathophysiological responses. As such, these molecules have been reported to influence a diverse array of biochemical, molecular, and cell biology events often impacting one another. Recent Advances: Discrete regulation of gasotransmitter molecule formation, movement, and reaction is critical to their biological function. Due to the chemical nature of these molecules, they can move rapidly throughout cells and tissues acting on targets through reactions with metal groups, reactive chemical species, and protein amino acids. CRITICAL ISSUES Given the breadth and complexity of gasotransmitter reactions, this field of research is expanding into exciting, yet sometimes confusing, areas of study with significant promise for understanding health and disease. The precise amounts of tissue and cellular gasotransmitter levels and where they are formed, as well as how they react with molecular targets or themselves, all remain poorly understood. FUTURE DIRECTIONS Elucidation of specific molecular targets, characteristics of gasotransmitter molecule heterotypic interactions, and spatiotemporal formation and metabolism are all important to better understand their true pathophysiological importance in various organ systems. Antioxid. Redox Signal. 26, 936-960.
Collapse
Affiliation(s)
- Gopi K Kolluru
- 1 Department of Pathology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Xinggui Shen
- 1 Department of Pathology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Shuai Yuan
- 2 Department of Cellular Biology and Anatomy, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Christopher G Kevil
- 1 Department of Pathology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana.,2 Department of Cellular Biology and Anatomy, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana.,3 Department of Molecular and Cellular Physiology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| |
Collapse
|
35
|
Interplay of myosin phosphatase and protein phosphatase-2A in the regulation of endothelial nitric-oxide synthase phosphorylation and nitric oxide production. Sci Rep 2017; 7:44698. [PMID: 28300193 PMCID: PMC5353758 DOI: 10.1038/srep44698] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/13/2017] [Indexed: 01/07/2023] Open
Abstract
The inhibitory phosphorylation of endothelial nitric oxide (NO) synthase (eNOS) at Thr497 (eNOSpThr497) by protein kinase C or RhoA-activated kinase is a major regulatory determinant of eNOS activity. The signalling mechanisms involved in the dephosphorylation of eNOSpThr497 have not yet been clarified. This study identifies myosin phosphatase (MP) holoenzyme consisting of protein phosphatase-1 catalytic subunit (PP1c) and MP target subunit-1 (MYPT1) as an eNOSpThr497 phosphatase. In support of this finding are: (i) eNOS and MYPT1 interacts in various endothelial cells (ECs) and in in vitro binding assays (ii) MYPT1 targets and stimulates PP1c toward eNOSpThr497 substrate (iii) phosphorylation of MYPT1 at Thr696 (MYPT1pThr696) controls the activity of MP on eNOSpThr497. Phosphatase inhibition suppresses both NO production and transendothelial resistance (TER) of ECs. In contrast, epigallocatechin-3-gallate (EGCG) signals ECs via the 67 kDa laminin-receptor (67LR) resulting in protein kinase A dependent activation of protein phosphatase-2A (PP2A). PP2A dephosphorylates MYPT1pThr696 and thereby stimulates MP activity inducing dephosphorylation of eNOSpThr497 and the 20 kDa myosin II light chains. Thus an interplay of MP and PP2A is involved in the physiological regulation of EC functions implying that an EGCG dependent activation of these phosphatases leads to enhanced NO production and EC barrier improvement.
Collapse
|
36
|
Saran U, Mani KP, Balaguru UM, Swaminathan A, Nagarajan S, Dharmarajan AM, Chatterjee S. sFRP4 signalling of apoptosis and angiostasis uses nitric oxide-cGMP-permeability axis of endothelium. Nitric Oxide 2017; 66:30-42. [PMID: 28267592 DOI: 10.1016/j.niox.2017.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 02/11/2017] [Accepted: 02/13/2017] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) plays a critical role in endothelial functions such as cellular migration, vascular permeability and angiogenesis. Angiogenesis, the formation of new blood vessels from "pre-existing" ones is a carefully regulated process and essential during reproduction, development and wound healing. Previously our lab group reported that Secreted Frizzled-Related Protein 4 (sFRP4) could inhibit angiogenesis in both in vitro and in vivo conditions. sFRP4 belongs to a family of secreted glycoproteins that function as antagonists of the canonical Wnt signalling pathway. Although the pro-apoptotic role of sFRP4 is well discussed in literature, little is known in regards to its anti-angiogenic property. The objective of this study was to elucidate sFRP4 implications in NO biology of the endothelium. Results demonstrate that sFRP4 causes endothelial dysfunction by suppressing NO-cGMP signaling and elevating corresponding ROS levels. The imbalance between NO and ROS levels results in apoptosis and subsequent leakiness of endothelium as confirmed in vivo (Texas red/Annxin - CAM assay) and in vitro (Monolayer permeability assay) conditions. Furthermore utilizing peptides synthesized from the CRD domain of sFRP4, our results showed that while these peptides were able to cause endothelial dysfunctions, they did not cause apoptosis of the endothelial cells. Thereby confirming that sFRP4 can mediate its anti-angiogenic effect independent of its pro-apoptotic property. In conclusion, the current study reports that sFRP4-mediated anti-angiogenesis occurs as a result of impaired NO-cGMP signaling which in turn allow for elevation of redox levels and promotion of apoptosis of endothelial cells.
Collapse
Affiliation(s)
- Uttara Saran
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai 600044, India
| | - Krishna Priya Mani
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai 600044, India
| | | | - Akila Swaminathan
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai 600044, India
| | - Shunmugam Nagarajan
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai 600044, India
| | - Arun M Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai 600044, India; Department of Biotechnology, Anna University, Chennai 600044, India.
| |
Collapse
|
37
|
Jiang Q, Liu Z, Zhou Z, Wang L, Wang L, Yue F, Wang J, Wang H, Song L. Transcriptional activation and translocation of ancient NOS during immune response. FASEB J 2016; 30:3527-3540. [DOI: 10.1096/fj.201500193rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 06/28/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Qiufen Jiang
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
- University of Chinese Academy of Sciences Beijing China
| | - Zhaoqun Liu
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
- University of Chinese Academy of Sciences Beijing China
| | - Zhi Zhou
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
| | - Lingling Wang
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
- Key Laboratory of Mariculture and Stock Enhancement in North China's SeaMinistry of AgricultureDalian Ocean University Dalian China
| | - Leilei Wang
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
- University of Chinese Academy of Sciences Beijing China
| | - Feng Yue
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
- University of Chinese Academy of Sciences Beijing China
| | - Jingjing Wang
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
- University of Chinese Academy of Sciences Beijing China
| | - Hao Wang
- Key Laboratory of Experimental Marine BiologyInstitute of OceanologyChinese Academy of Sciences Qingdao China
| | - Linsheng Song
- Key Laboratory of Mariculture and Stock Enhancement in North China's SeaMinistry of AgricultureDalian Ocean University Dalian China
| |
Collapse
|
38
|
Biwer LA, Taddeo EP, Kenwood BM, Hoehn KL, Straub AC, Isakson BE. Two functionally distinct pools of eNOS in endothelium are facilitated by myoendothelial junction lipid composition. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:671-9. [PMID: 27106139 PMCID: PMC4869716 DOI: 10.1016/j.bbalip.2016.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 03/10/2016] [Accepted: 04/16/2016] [Indexed: 12/21/2022]
Abstract
In resistance arteries, endothelial cells (EC) make contact with smooth muscle cells (SMC), forming myoendothelial junctions (MEJ). Endothelial nitric oxide synthase (eNOS) is present in the luminal side of the EC (apical EC) and the basal side of the EC (MEJ). To test if these eNOS pools acted in sync or separately, we co-cultured ECs and SMCs, then stimulated SMCs with phenylephrine (PE). Adrenergic activation causes inositol [1,4,5] triphosphate (IP3) to move from SMC to EC through gap junctions at the MEJ. PE increases MEJ eNOS phosphorylation (eNOS-P) at S1177, but not in EC. Conversely, we used bradykinin (BK) to increase EC calcium; this increased EC eNOS-P but did not affect MEJ eNOS-P. Inhibiting gap junctions abrogated the MEJ eNOS-P after PE, but had no effect on BK eNOS-P. Differential lipid composition between apical EC and MEJ may account for the compartmentalized eNOS-P response. Indeed, DAG and phosphatidylserine are both enriched in MEJ. These lipids are cofactors for PKC activity, which was significantly increased at the MEJ after PE. Because PKC activity also relies on endoplasmic reticulum (ER) calcium release, we used thapsigargin and xestospongin C, BAPTA, and PKC inhibitors, which caused significant decreases in MEJ eNOS-P after PE. Functionally, BK inhibited leukocyte adhesion and PE caused an increase in SMC cGMP. We hypothesize that local lipid composition of the MEJ primes PKC and eNOS-P for stimulation by PE, allowing for compartmentalized function of eNOS in the blood vessel wall.
Collapse
Affiliation(s)
- Lauren A Biwer
- Department of Molecular Physiology and Biophysics, University of Virginia, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, USA
| | - Evan P Taddeo
- Department of Pharmacology, University of Virginia, USA
| | | | - Kyle L Hoehn
- Department of Pharmacology, University of Virginia, USA; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Australia
| | - Adam C Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, USA; Heart, Lung, Blood and Vascular Medicine Institute; University of Pittsburgh, USA
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, USA.
| |
Collapse
|
39
|
Cellular response of the blood-brain barrier to injury: Potential biomarkers and therapeutic targets for brain regeneration. Neurobiol Dis 2016; 91:262-73. [DOI: 10.1016/j.nbd.2016.03.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/09/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
|
40
|
Mowa CN, Jesmin S, Sakuma I, Usip S, Togashi H, Yoshioka M, Hattori Y, Papka R. Characterization of Vascular Endothelial Growth Factor (VEGF) in the Uterine Cervix over Pregnancy: Effects of Denervation and Implications for Cervical Ripening. J Histochem Cytochem 2016; 52:1665-74. [PMID: 15557221 DOI: 10.1369/jhc.4a6455.2004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bilateral neurectomy of the pelvic nerve (BLPN) that carries uterine cervix-related sensory nerves induces dystocia, and administration of its vasoactive neuropeptides induces changes in the cervical microvasculature, resembling those that occur in the ripening cervix. This study was designed to test the hypothesis that (a) the cervix of pregnant rats expresses vascular endothelial growth factor (VEGF) and components of the angiogenic signaling pathway [VEGF receptors (Flt-1, KDR), activity of protein kinase B, Akt (phosphorylated Akt), and endothelial nitric oxide synthase (eNOS)] and von Willebrand Factor (vWF) and that these molecules undergo changes with pregnancy, and (b) bilateral pelvic neurectomy (BLPN) alters levels of VEGF concentration in the cervix. Using RT-PCR and sequencing, two VEGF isoforms, 120 and 164, were identified in the rat cervix. VEGF, VEGF receptor-1 (Flt-1), eNOS, and vWF immunoreactivities (ir) were localized in the microvasculature of cervical stroma. Their protein levels increased during pregnancy but decreased to control levels by 2 days postpartum. VEGF receptor-2 (KDR)-ir was confined to the epithelium of the endocervix. BLPN downregulated levels of VEGF by a third. Therefore, the components of the angiogenic signaling pathway are expressed in the cervix and change over pregnancy. Furthermore, angiogenic and sensory neuronal factors may be important in regulating the dynamic microvasculature in the ripening cervix and may subsequently play a role in cervical ripening and the birth process.
Collapse
Affiliation(s)
- C N Mowa
- Dept. of Neurobiology, Northeastern Ohio Universities College of Medicine, 4209 State Rt. 44, Rootstown, OH 44272, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Transcriptional and Posttranslational Regulation of eNOS in the Endothelium. ADVANCES IN PHARMACOLOGY 2016; 77:29-64. [PMID: 27451094 DOI: 10.1016/bs.apha.2016.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is a highly reactive free radical gas and these unique properties have been adapted for a surprising number of biological roles. In neurons, NO functions as a neurotransmitter; in immune cells, NO contributes to host defense; and in endothelial cells, NO is a major regulator of blood vessel homeostasis. In the vasculature, NO is synthesized on demand by a specific enzyme, endothelial nitric oxide synthase (eNOS) that is uniquely expressed in the endothelial cells that form the interface between the circulating blood and the various tissues of the body. NO regulates endothelial and blood vessel function via two distinct pathways, the activation of soluble guanylate cyclase and cGMP-dependent signaling and the S-nitrosylation of proteins with reactive thiols (S-nitrosylation). The chemical properties of NO also serve to reduce oxidation and regulate mitochondrial function. Reduced synthesis and/or compromised biological activity of NO precede the development of cardiovascular disease and this has generated a high level of interest in the mechanisms controlling the synthesis and fate of NO in the endothelium. The amount of NO produced results from the expression level of eNOS, which is regulated at the transcriptional and posttranscriptional levels as well as the acute posttranslational regulation of eNOS. The goal of this chapter is to highlight and integrate past and current knowledge of the mechanisms regulating eNOS expression in the endothelium and the posttranslational mechanisms regulating eNOS activity in both health and disease.
Collapse
|
42
|
Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. Pflugers Arch 2016; 468:1125-1137. [DOI: 10.1007/s00424-016-1839-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
|
43
|
Blum-Johnston C, Thorpe RB, Wee C, Romero M, Brunelle A, Blood Q, Wilson R, Blood AB, Francis M, Taylor MS, Longo LD, Pearce WJ, Wilson SM. Developmental acceleration of bradykinin-dependent relaxation by prenatal chronic hypoxia impedes normal development after birth. Am J Physiol Lung Cell Mol Physiol 2015; 310:L271-86. [PMID: 26637638 DOI: 10.1152/ajplung.00340.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/02/2015] [Indexed: 11/22/2022] Open
Abstract
Bradykinin-induced activation of the pulmonary endothelium triggers nitric oxide production and other signals that cause vasorelaxation, including stimulation of large-conductance Ca(2+)-activated K(+) (BKCa) channels in myocytes that hyperpolarize the plasma membrane and decrease intracellular Ca(2+). Intrauterine chronic hypoxia (CH) may reduce vasorelaxation in the fetal-to-newborn transition and contribute to pulmonary hypertension of the newborn. Thus we examined the effects of maturation and CH on the role of BKCa channels during bradykinin-induced vasorelaxation by examining endothelial Ca(2+) signals, wire myography, and Western immunoblots on pulmonary arteries isolated from near-term fetal (∼ 140 days gestation) and newborn, 10- to 20-day-old, sheep that lived in normoxia at 700 m or in CH at high altitude (3,801 m) for >100 days. CH enhanced bradykinin-induced relaxation of fetal vessels but decreased relaxation in newborns. Endothelial Ca(2+) responses decreased with maturation but increased with CH. Bradykinin-dependent relaxation was sensitive to 100 μM nitro-L-arginine methyl ester or 10 μM 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, supporting roles for endothelial nitric oxide synthase and soluble guanylate cyclase activation. Indomethacin blocked relaxation in CH vessels, suggesting upregulation of PLA2 pathways. BKCa channel inhibition with 1 mM tetraethylammonium reduced bradykinin-induced vasorelaxation in the normoxic newborn and fetal CH vessels. Maturation reduced whole cell BKCa channel α1-subunit expression but increased β1-subunit expression. These results suggest that CH amplifies the contribution of BKCa channels to bradykinin-induced vasorelaxation in fetal sheep but stunts further development of this vasodilatory pathway in newborns. This involves complex changes in multiple components of the bradykinin-signaling axes.
Collapse
Affiliation(s)
- Carla Blum-Johnston
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
| | - Richard B Thorpe
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Chelsea Wee
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Monica Romero
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, California
| | - Alexander Brunelle
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Quintin Blood
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Rachael Wilson
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California;
| | - Arlin B Blood
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California; and
| | - Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Birmingham, Alabama
| | - Mark S Taylor
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Birmingham, Alabama
| | - Lawrence D Longo
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - William J Pearce
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean M Wilson
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
44
|
Trane AE, Hiob MA, Uy T, Pavlov D, Bernatchez P. Caveolin-1 scaffolding domain residue phenylalanine 92 modulates Akt signaling. Eur J Pharmacol 2015; 766:46-55. [DOI: 10.1016/j.ejphar.2015.09.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/18/2015] [Accepted: 09/21/2015] [Indexed: 02/02/2023]
|
45
|
Peng H, Zhuang Y, Chen Y, Rizzo AN, Chen W. The Characteristics and Regulatory Mechanisms of Superoxide Generation from eNOS Reductase Domain. PLoS One 2015; 10:e0140365. [PMID: 26465144 PMCID: PMC4605588 DOI: 10.1371/journal.pone.0140365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/24/2015] [Indexed: 11/21/2022] Open
Abstract
In addition to superoxide (O2.-) generation from nitric oxide synthase (NOS) oxygenase domain, a new O2.- generation site has been identified in the reductase domain of inducible NOS (iNOS) and neuronal NOS (nNOS). Cysteine S-glutathionylation in eNOS reductase domain also induces O2.- generation from eNOS reductase domain. However, the characteristics and regulatory mechanism of the O2.- generation from NOS reductase domain remain unclear. We cloned and purified the wild type bovine eNOS (WT eNOS), a mutant of Serine 1179 replaced with aspartic acid eNOS (S1179D eNOS), which mimics the negative charge caused by phosphorylationand truncated eNOS reductase domain (eNOS RD). Both WT eNOS and S1179D eNOS generated significant amount of O2.- in the absence of BH4 and L-arginine. The capacity of O2.- generation from S1179D eNOS was significantly higher than that of WT eNOS (1.74:1). O2.- generation from both WT eNOS and S1179D eNOS were not completely inhibited by 100nM tetrahydrobiopterin(BH4). This BH4 un-inhibited O2.- generation from eNOS was blocked by 10mM flavoprotein inhibitor, diphenyleneiodonium (DPI). Purified eNOS reductase domain protein confirmed that this BH4 un-inhibited O2.- generation originates at the FMN or FAD/NADPH binding site of eNOS reductase domain. DEPMPO-OOH adduct EPR signals and NADPH consumptions analyses showed that O2.- generation from eNOS reductase domain was regulated by Serine 1179 phosphorylation and DPI, but not by L-arginine, BH4 or calmodulin (CaM). In addition to the heme center of eNOS oxygenase domain, we confirmed another O2.- generation site in the eNOS reductase domain and characterized its regulatory properties.
Collapse
Affiliation(s)
- Hu Peng
- Department of Emergency Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Yugang Zhuang
- Department of Emergency Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Yuanzhuo Chen
- Department of Emergency Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Alicia N. Rizzo
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Weiguo Chen
- Department of Emergency Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
46
|
Newby EA, Myers DA, Ducsay CA. Fetal endocrine and metabolic adaptations to hypoxia: the role of the hypothalamic-pituitary-adrenal axis. Am J Physiol Endocrinol Metab 2015; 309:E429-39. [PMID: 26173460 PMCID: PMC4556885 DOI: 10.1152/ajpendo.00126.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/10/2015] [Indexed: 11/22/2022]
Abstract
In utero, hypoxia is a significant yet common stress that perturbs homeostasis and can occur due to preeclampsia, preterm labor, maternal smoking, heart or lung disease, obesity, and high altitude. The fetus has the extraordinary capacity to respond to stress during development. This is mediated in part by the hypothalamic-pituitary-adrenal (HPA) axis and more recently explored changes in perirenal adipose tissue (PAT) in response to hypoxia. Obvious ethical considerations limit studies of the human fetus, and fetal studies in the rodent model are limited due to size considerations and major differences in developmental landmarks. The sheep is a common model that has been used extensively to study the effects of both acute and chronic hypoxia on fetal development. In response to high-altitude-induced, moderate long-term hypoxia (LTH), both the HPA axis and PAT adapt to preserve normal fetal growth and development while allowing for responses to acute stress. Although these adaptations appear beneficial during fetal development, they may become deleterious postnatally and into adulthood. The goal of this review is to examine the role of the HPA axis in the convergence of endocrine and metabolic adaptive responses to hypoxia in the fetus.
Collapse
Affiliation(s)
- Elizabeth A Newby
- Center for Perinatal Biology, Loma Linda University, Loma Linda, California; and
| | - Dean A Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Charles A Ducsay
- Center for Perinatal Biology, Loma Linda University, Loma Linda, California; and
| |
Collapse
|
47
|
Hamilton K, MacKenzie A. Gender specific generation of nitroxyl (HNO) from rat endothelium. Vascul Pharmacol 2015; 71:208-14. [DOI: 10.1016/j.vph.2015.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/16/2015] [Accepted: 03/09/2015] [Indexed: 10/23/2022]
|
48
|
Retamal MA, León-Paravic CG, Ezquer M, Ezquer F, Rio RD, Pupo A, Martínez AD, González C. Carbon monoxide: A new player in the redox regulation of connexin hemichannels. IUBMB Life 2015; 67:428-37. [DOI: 10.1002/iub.1388] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/05/2015] [Indexed: 01/23/2023]
Affiliation(s)
- Mauricio A. Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina; Clínica Alemana Universidad del Desarrollo; Santiago Chile
| | - Carmen G. León-Paravic
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina; Clínica Alemana Universidad del Desarrollo; Santiago Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina; Clínica Alemana Universidad del Desarrollo; Santiago Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina; Clínica Alemana Universidad del Desarrollo; Santiago Chile
| | - Rodrigo Del Rio
- Centro de Investigación Biomédica; Universidad Autónoma de Chile; Santiago Chile
| | - Amaury Pupo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias; Instituto de Neurociencia; Universidad de Valparaíso; Valparaíso Chile
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias; Instituto de Neurociencia; Universidad de Valparaíso; Valparaíso Chile
| | - Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias; Instituto de Neurociencia; Universidad de Valparaíso; Valparaíso Chile
| |
Collapse
|
49
|
Moustafa A, Habara Y. A novel role for carbon monoxide as a potent regulator of intracellular Ca2+and nitric oxide in rat pancreatic acinar cells. Am J Physiol Cell Physiol 2014; 307:C1039-49. [DOI: 10.1152/ajpcell.00252.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO) is known as an essential gaseous messenger that regulates a wide array of physiological and pathological processes, similar to nitric oxide (NO) and hydrogen sulfide. The aim of the present study was to elucidate the potential role of CO in Ca2+homeostasis and to explore the underlying mechanisms in pancreatic acinar cells. The exogenous application of a CO-releasing molecule dose-dependently increased intracellular Ca2+concentration ([Ca2+]i). A heme oxygenase (HO) inducer increased [Ca2+]iin a concentration-dependent manner, and the increase was diminished by an HO inhibitor. The CO-induced [Ca2+]iincrease persisted in the absence of extracellular Ca2+, indicating that Ca2+release is the initial source for the increase. The inhibition of G protein, phospholipase C (PLC), and inositol 1,4,5-trisphosphate (IP3) receptor diminished the CO-induced [Ca2+]iincrease. CO upregulated endothelial nitric oxide synthase (eNOS) expression and stimulated NO production, and NOS inhibitor, calmodulin inhibitor, or the absence of extracellular Ca2+eliminated the latter response. Blocking the phosphatidylinositol 3-kinase (PI3K)-Akt/protein kinase B (PKB) pathway abolished CO-induced NO production. Pretreatment with an NOS inhibitor, NO scavenger, or soluble guanylate cyclase inhibitor, did not affect the CO-induced [Ca2+]iincrease, indicating that NO, soluble guanylate cyclase, and cyclic guanosine 5′-monophosphate are not involved in the CO-induced [Ca2+]iincrease. CO inhibited the secretory responses to CCK-octapeptide or carbachol. We conclude that CO acts as a regulator not only for [Ca2+]ihomeostasis via a PLC-IP3-IP3receptor cascade but also for NO production via the calmodulin and PI3K-Akt/PKB pathway, and both CO and NO interact. Moreover, CO may provide potential therapy to ameliorate acute pancreatitis by inhibiting amylase secretion.
Collapse
Affiliation(s)
- Amira Moustafa
- Laboratory of Physiology, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; and
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Yoshiaki Habara
- Laboratory of Physiology, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; and
| |
Collapse
|
50
|
Aicart-Ramos C, Sánchez-Ruiloba L, Gómez-Parrizas M, Zaragoza C, Iglesias T, Rodríguez-Crespo I. Protein kinase D activity controls endothelial nitric oxide synthesis. J Cell Sci 2014; 127:3360-72. [PMID: 24928905 DOI: 10.1242/jcs.148601] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) regulates key functions of the endothelium, such as angiogenesis or vessel repair in processes involving endothelial nitric oxide synthase (eNOS) activation. One of the effector kinases that become activated in endothelial cells upon VEGF treatment is protein kinase D (PKD). Here, we show that PKD phosphorylates eNOS, leading to its activation and a concomitant increase in NO synthesis. Using mass spectrometry, we show that the purified active kinase specifically phosphorylates recombinant eNOS on Ser1179. Treatment of endothelial cells with VEGF or phorbol 12,13-dibutyrate (PDBu) activates PKD and increases eNOS Ser1179 phosphorylation. In addition, pharmacological inhibition of PKD and gene silencing of both PKD1 and PKD2 abrogate VEGF signaling, resulting in a clear diminished migration of endothelial cells in a wound healing assay. Finally, inhibition of PKD in mice results in an almost complete disappearance of the VEGF-induced vasodilatation, as monitored through determination of the diameter of the carotid artery. Hence, our data indicate that PKD is a new regulatory kinase of eNOS in endothelial cells whose activity orchestrates mammalian vascular tone.
Collapse
Affiliation(s)
- Clara Aicart-Ramos
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Lucía Sánchez-Ruiloba
- Instituto de Investigaciones Biomédicas "Alberto Sols". CSIC-UAM, C/Arturo Duperier, Madrid 28029, Spain CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Carlos Zaragoza
- Cardiovascular Research Unit University Francisco de Vitoria/Hospital Ramón y Cajal, Ctra Colmenar Viejo Km 9,100, Madrid 28034, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols". CSIC-UAM, C/Arturo Duperier, Madrid 28029, Spain CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ignacio Rodríguez-Crespo
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Madrid 28040, Spain
| |
Collapse
|