1
|
Erkens R, Duse DA, Brum A, Chadt A, Becher S, Siragusa M, Quast C, Müssig J, Roden M, Cortese-Krott M, Ibáñez B, Lammert E, Fleming I, Jung C, Al-Hasani H, Heusch G, Kelm M. Inhibition of proline-rich tyrosine kinase 2 restores cardioprotection by remote ischaemic preconditioning in type 2 diabetes. Br J Pharmacol 2024; 181:4174-4194. [PMID: 38956895 DOI: 10.1111/bph.16483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/26/2024] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Remote ischaemic preconditioning (rIPC) for cardioprotection is severely impaired in diabetes, and therapeutic options to restore it are lacking. The vascular endothelium plays a key role in rIPC. Given that the activity of endothelial nitric oxide synthase (eNOS) is inhibited by proline-rich tyrosine kinase 2 (Pyk2), we hypothesized that pharmacological Pyk2 inhibition could restore eNOS activity and thus restore remote cardioprotection in diabetes. EXPERIMENTAL APPROACH New Zealand obese (NZO) mice that demonstrated key features of diabetes were studied. The consequence of Pyk2 inhibition on endothelial function, rIPC and infarct size after myocardial infarction were evaluated. The impact of plasma from mice and humans with or without diabetes was assessed in isolated buffer perfused murine hearts and aortic rings. KEY RESULTS Plasma from nondiabetic mice and humans, both subjected to rIPC, caused remote tissue protection. Similar to diabetic humans, NZO mice demonstrated endothelial dysfunction. NZO mice had reduced circulating nitrite levels, elevated arterial blood pressure and a larger infarct size after ischaemia and reperfusion than BL6 mice. Pyk2 increased the phosphorylation of eNOS at its inhibitory site (Tyr656), limiting its activity in diabetes. The cardioprotective effects of rIPC were abolished in diabetic NZO mice. Pharmacological Pyk2 inhibition restored endothelial function and rescued cardioprotective effects of rIPC. CONCLUSION AND IMPLICATIONS Endothelial function and remote tissue protection are impaired in diabetes. Pyk2 is a novel target for treating endothelial dysfunction and restoring cardioprotection through rIPC in diabetes.
Collapse
Affiliation(s)
- Ralf Erkens
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Dragos Andrei Duse
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Amanda Brum
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, Deutsches Diabetes Zentrum at Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
| | - Stefanie Becher
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Mauro Siragusa
- Center for Molecular Medicine, Institute for Vascular Signalling, Goethe University Frankfurt, Frankfurt, Germany
- German Centre for Cardiovascular Research, Partner site RhineMain, Frankfurt, Germany
| | - Christine Quast
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Johanna Müssig
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University and University Hospital Duesseldorf, Duesseldorf, Germany
- Institute for Clinical Diabetology, Deutsches Diabetes Zentrum at Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Miriam Cortese-Krott
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
- CARID Cardiovascular Research Institute Duesseldorf, Duesseldorf, Germany
| | - Borja Ibáñez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Eckhard Lammert
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
- Institute of Metabolic Physiology, Heinrich-Heine University, Duesseldorf, Germany
| | - Ingrid Fleming
- Center for Molecular Medicine, Institute for Vascular Signalling, Goethe University Frankfurt, Frankfurt, Germany
- German Centre for Cardiovascular Research, Partner site RhineMain, Frankfurt, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, Deutsches Diabetes Zentrum at Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University School of Medicine Essen, Essen, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
- CARID Cardiovascular Research Institute Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
2
|
LoBue A, Li Z, Heuser SK, Li J, Leo F, Vornholz L, Dunaway LS, Suvorava T, Isakson BE, Cortese-Krott MM. Generation and characterization of a conditional eNOS knock out mouse model for cell-specific reactivation of eNOS in gain-of-function studies. Nitric Oxide 2024; 153:106-113. [PMID: 39419166 DOI: 10.1016/j.niox.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) in the vessel wall regulates blood pressure and cardiovascular hemodynamics. In this study, we generated conditional eNOS knock out (KO) mice characterized by a duplicated/inverted exon 2 flanked with two pairs of loxP regions (eNOSinv/inv); a Cre-recombinase activity induces cell-specific reactivation of eNOS, as a result of a flipping of the inverted exon 2 (eNOSfl). This work aimed to test the efficiency of the Cre-mediated cell-specific recombination and the resulting eNOS expression/function. As proof of concept, we crossed eNOSinv/inv mice with DeleterCrepos (DelCrepos) mice, expressing Cre recombinase in all cells. We generated heterozygous eNOSfl/inv or homozygous eNOSfl/fl mice, and eNOSinv/inv littermate mice. We found that both eNOSfl/fl and eNOSfl/inv mice express eNOS and the overall expression level depends on the number of mutated alleles, while eNOSinv/inv mice did not show any eNOS expression. Vascular endothelial function was restored in eNOSfl/fl and eNOSfl/inv mice, as determined by ACh-dependent vasodilation of aortic rings. Cre-dependent reactivation of eNOS in eNOSfl/fl and eNOSfl/inv mice rescued eNOSinv/inv (phenotypically global eNOS KO) mice from hypertension. These findings demonstrate that eNOS expression is restored in eNOSfl/fl mice at comparable physiological levels of WT mice, and its functional activity is independent on the number of the reactivated alleles. Therefore, eNOSinv/inv mice are a useful model for studying the effects of conditional reactivation of eNOS and gene dosage effects in specific cells for gain-of-function studies.
Collapse
Affiliation(s)
- Anthea LoBue
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Zhixin Li
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Sophia K Heuser
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Junjie Li
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Francesca Leo
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Lukas Vornholz
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Tatsiana Suvorava
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden; CARID, Cardiovascular Research Institute Düsseldorf, Germany.
| |
Collapse
|
3
|
Richter K, Asci N, Singh VK, Yakoob SH, Meixner M, Zakrzewicz A, Liese J, Hecker A, Wilker S, Stumpf S, Schlüter KD, Rohde M, Gödecke A, Padberg W, Manzini I, Schmalzing G, Grau V. Activation of endothelial NO synthase and P2X7 receptor modification mediates the cholinergic control of ATP-induced interleukin-1β release by mononuclear phagocytes. Front Immunol 2023; 14:1140592. [PMID: 36969210 PMCID: PMC10034071 DOI: 10.3389/fimmu.2023.1140592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
ObjectiveThe pro-inflammatory cytokine interleukin-1β (IL-1β) plays a central role in host defense against infections. High systemic IL-1β levels, however, promote the pathogenesis of inflammatory disorders. Therefore, mechanisms controlling IL-1β release are of substantial clinical interest. Recently, we identified a cholinergic mechanism inhibiting the ATP-mediated IL-1β release by human monocytes via nicotinic acetylcholine receptor (nAChR) subunits α7, α9 and/or α10. We also discovered novel nAChR agonists that trigger this inhibitory function in monocytic cells without eliciting ionotropic functions at conventional nAChRs. Here, we investigate the ion flux-independent signaling pathway that links nAChR activation to the inhibition of the ATP-sensitive P2X7 receptor (P2X7R).MethodsDifferent human and murine mononuclear phagocytes were primed with lipopolysaccharide and stimulated with the P2X7R agonist BzATP in the presence or absence of nAChR agonists, endothelial NO synthase (eNOS) inhibitors, and NO donors. IL-1β was measured in cell culture supernatants. Patch-clamp and intracellular Ca2+ imaging experiments were performed on HEK cells overexpressing human P2X7R or P2X7R with point mutations at cysteine residues in the cytoplasmic C-terminal domain.ResultsThe inhibitory effect of nAChR agonists on the BzATP-induced IL-1β release was reversed in the presence of eNOS inhibitors (L-NIO, L-NAME) as well as in U937 cells after silencing of eNOS expression. In peripheral blood mononuclear leukocytes from eNOS gene-deficient mice, the inhibitory effect of nAChR agonists was absent, suggesting that nAChRs signal via eNOS to inhibit the BzATP-induced IL-1β release. Moreover, NO donors (SNAP, S-nitroso-N-acetyl-DL-penicillamine; SIN-1) inhibited the BzATP-induced IL-1β release by mononuclear phagocytes. The BzATP-induced ionotropic activity of the P2X7R was abolished in the presence of SIN-1 in both, Xenopus laevis oocytes and HEK cells over-expressing the human P2X7R. This inhibitory effect of SIN-1 was absent in HEK cells expressing P2X7R, in which C377 was mutated to alanine, indicating the importance of C377 for the regulation of the P2X7R function by protein modification.ConclusionWe provide first evidence that ion flux-independent, metabotropic signaling of monocytic nAChRs involves eNOS activation and P2X7R modification, resulting in an inhibition of ATP signaling and ATP-mediated IL-1β release. This signaling pathway might be an interesting target for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Katrin Richter
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
- *Correspondence: Katrin Richter,
| | - Nilay Asci
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Vijay K. Singh
- Department of Paediatric Haematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | | | - Marion Meixner
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Anna Zakrzewicz
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Juliane Liese
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Andreas Hecker
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Sigrid Wilker
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Sabine Stumpf
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | | | - Marius Rohde
- Department of Paediatric Haematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Axel Gödecke
- Institute of Cardiovascular Physiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Winfried Padberg
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Ivan Manzini
- Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Günther Schmalzing
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
| | - Veronika Grau
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Centre for Lung Research (DZL), Cardio Pulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
4
|
Nayeem MA, Geldenhuys WJ, Hanif A. Role of cytochrome P450-epoxygenase and soluble epoxide hydrolase in the regulation of vascular response. ADVANCES IN PHARMACOLOGY 2023; 97:37-131. [DOI: 10.1016/bs.apha.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
5
|
Nayeem MA, Hanif A, Geldenhuys WJ, Agba S. Crosstalk between adenosine receptors and CYP450-derived oxylipins in the modulation of cardiovascular, including coronary reactive hyperemic response. Pharmacol Ther 2022; 240:108213. [PMID: 35597366 DOI: 10.1016/j.pharmthera.2022.108213] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Adenosine is a ubiquitous endogenous nucleoside or autacoid that affects the cardiovascular system through the activation of four G-protein coupled receptors: adenosine A1 receptor (A1AR), adenosine A2A receptor (A2AAR), adenosine A2B receptor (A2BAR), and adenosine A3 receptor (A3AR). With the rapid generation of this nucleoside from cellular metabolism and the widespread distribution of its four G-protein coupled receptors in almost all organs and tissues of the body, this autacoid induces multiple physiological as well as pathological effects, not only regulating the cardiovascular system but also the central nervous system, peripheral vascular system, and immune system. Mounting evidence shows the role of CYP450-enzymes in cardiovascular physiology and pathology, and the genetic polymorphisms in CYP450s can increase susceptibility to cardiovascular diseases (CVDs). One of the most important physiological roles of CYP450-epoxygenases (CYP450-2C & CYP2J2) is the metabolism of arachidonic acid (AA) and linoleic acid (LA) into epoxyeicosatrienoic acids (EETs) and epoxyoctadecaenoic acid (EpOMEs) which generally involve in vasodilation. Like an increase in coronary reactive hyperemia (CRH), an increase in anti-inflammation, and cardioprotective effects. Moreover, the genetic polymorphisms in CYP450-epoxygenases will change the beneficial cardiovascular effects of metabolites or oxylipins into detrimental effects. The soluble epoxide hydrolase (sEH) is another crucial enzyme ubiquitously expressed in all living organisms and almost all organs and tissues. However, in contrast to CYP450-epoxygenases, sEH converts EETs into dihydroxyeicosatrienoic acid (DHETs), EpOMEs into dihydroxyoctadecaenoic acid (DiHOMEs), and others and reverses the beneficial effects of epoxy-fatty acids leading to vasoconstriction, reducing CRH, increase in pro-inflammation, increase in pro-thrombotic and become less cardioprotective. Therefore, polymorphisms in the sEH gene (Ephx2) cause the enzyme to become overactive, making it more vulnerable to CVDs, including hypertension. Besides the sEH, ω-hydroxylases (CYP450-4A11 & CYP450-4F2) derived metabolites from AA, ω terminal-hydroxyeicosatetraenoic acids (19-, 20-HETE), lipoxygenase-derived mid-chain hydroxyeicosatetraenoic acids (5-, 11-, 12-, 15-HETEs), and the cyclooxygenase-derived prostanoids (prostaglandins: PGD2, PGF2α; thromboxane: Txs, oxylipins) are involved in vasoconstriction, hypertension, reduction in CRH, pro-inflammation and cardiac toxicity. Interestingly, the interactions of adenosine receptors (A2AAR, A1AR) with CYP450-epoxygenases, ω-hydroxylases, sEH, and their derived metabolites or oxygenated polyunsaturated fatty acids (PUFAs or oxylipins) is shown in the regulation of the cardiovascular functions. In addition, much evidence demonstrates polymorphisms in CYP450-epoxygenases, ω-hydroxylases, and sEH genes (Ephx2) and adenosine receptor genes (ADORA1 & ADORA2) in the human population with the susceptibility to CVDs, including hypertension. CVDs are the number one cause of death globally, coronary artery disease (CAD) was the leading cause of death in the US in 2019, and hypertension is one of the most potent causes of CVDs. This review summarizes the articles related to the crosstalk between adenosine receptors and CYP450-derived oxylipins in vascular, including the CRH response in regular salt-diet fed and high salt-diet fed mice with the correlation of heart perfusate/plasma oxylipins. By using A2AAR-/-, A1AR-/-, eNOS-/-, sEH-/- or Ephx2-/-, vascular sEH-overexpressed (Tie2-sEH Tr), vascular CYP2J2-overexpressed (Tie2-CYP2J2 Tr), and wild-type (WT) mice. This review article also summarizes the role of pro-and anti-inflammatory oxylipins in cardiovascular function/dysfunction in mice and humans. Therefore, more studies are needed better to understand the crosstalk between the adenosine receptors and eicosanoids to develop diagnostic and therapeutic tools by using plasma oxylipins profiles in CVDs, including hypertensive cases in the future.
Collapse
Affiliation(s)
- Mohammed A Nayeem
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA.
| | - Ahmad Hanif
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Werner J Geldenhuys
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Stephanie Agba
- Graduate student, Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
6
|
Suvorava T, Metry S, Pick S, Kojda G. Alterations in endothelial nitric oxide synthase activity and their relevance to blood pressure. Biochem Pharmacol 2022; 205:115256. [DOI: 10.1016/j.bcp.2022.115256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022]
|
7
|
Cortese-Krott MM, Suvorava T, Leo F, Heuser SK, LoBue A, Li J, Becher S, Schneckmann R, Srivrastava T, Erkens R, Wolff G, Schmitt JP, Grandoch M, Lundberg JO, Pernow J, Isakson BE, Weitzberg E, Kelm M. Red blood cell eNOS is cardioprotective in acute myocardial infarction. Redox Biol 2022; 54:102370. [PMID: 35759945 PMCID: PMC9241051 DOI: 10.1016/j.redox.2022.102370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022] Open
Abstract
Red blood cells (RBCs) were shown to transport and release nitric oxide (NO) bioactivity and carry an endothelial NO synthase (eNOS). However, the pathophysiological significance of RBC eNOS for cardioprotection in vivo is unknown. Here we aimed to analyze the role of RBC eNOS in the regulation of coronary blood flow, cardiac performance, and acute myocardial infarction (AMI) in vivo. To specifically distinguish the role of RBC eNOS from the endothelial cell (EC) eNOS, we generated RBC- and EC-specific knock-out (KO) and knock-in (KI) mice by Cre-induced inactivation or reactivation of eNOS. We found that RBC eNOS KO mice had fully preserved coronary dilatory responses and LV function. Instead, EC eNOS KO mice had a decreased coronary flow response in isolated perfused hearts and an increased LV developed pressure in response to elevated arterial pressure, while stroke volume was preserved. Interestingly, RBC eNOS KO showed a significantly increased infarct size and aggravated LV dysfunction with decreased stroke volume and cardiac output. This is consistent with reduced NO bioavailability and oxygen delivery capacity in RBC eNOS KOs. Crucially, RBC eNOS KI mice had decreased infarct size and preserved LV function after AMI. In contrast, EC eNOS KO and EC eNOS KI had no differences in infarct size or LV dysfunction after AMI, as compared to the controls. These data demonstrate that EC eNOS controls coronary vasodilator function, but does not directly affect infarct size, while RBC eNOS limits infarct size in AMI. Therefore, RBC eNOS signaling may represent a novel target for interventions in ischemia/reperfusion after myocardial infarction.
Collapse
Affiliation(s)
- Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
| | - Tatsiana Suvorava
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Francesca Leo
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sophia K Heuser
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anthea LoBue
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Junjie Li
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stefanie Becher
- Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rebekka Schneckmann
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Tanu Srivrastava
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Ralf Erkens
- Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Georg Wolff
- Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Joachim P Schmitt
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Maria Grandoch
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - John Pernow
- Department of Cardiology, Karolinska Institute, Stockholm, Sweden
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Malte Kelm
- Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
8
|
Hocher B, Lu YP, Reichetzeder C, Zhang X, Tsuprykov O, Rahnenführer J, Xie L, Li J, Hu L, Krämer BK, Hasan AA. Paternal eNOS deficiency in mice affects glucose homeostasis and liver glycogen in male offspring without inheritance of eNOS deficiency itself. Diabetologia 2022; 65:1222-1236. [PMID: 35488925 PMCID: PMC9174141 DOI: 10.1007/s00125-022-05700-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 02/07/2022] [Indexed: 11/03/2022]
Abstract
AIMS/HYPOTHESIS It was shown that maternal endothelial nitric oxide synthase (eNOS) deficiency causes fatty liver disease and numerically lower fasting glucose in female wild-type offspring, suggesting that parental genetic variants may influence the offspring's phenotype via epigenetic modifications in the offspring despite the absence of a primary genetic defect. The aim of the current study was to analyse whether paternal eNOS deficiency may cause the same phenotype as seen with maternal eNOS deficiency. METHODS Heterozygous (+/-) male eNOS (Nos3) knockout mice or wild-type male mice were bred with female wild-type mice. The phenotype of wild-type offspring of heterozygous male eNOS knockout mice was compared with offspring from wild-type parents. RESULTS Global sperm DNA methylation decreased and sperm microRNA pattern altered substantially. Fasting glucose and liver glycogen storage were increased when analysing wild-type male and female offspring of +/- eNOS fathers. Wild-type male but not female offspring of +/- eNOS fathers had increased fasting insulin and increased insulin after glucose load. Analysing candidate genes for liver fat and carbohydrate metabolism revealed that the expression of genes encoding glucocorticoid receptor (Gr; also known as Nr3c1) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Pgc1a; also known as Ppargc1a) was increased while DNA methylation of Gr exon 1A and Pgc1a promoter was decreased in the liver of male wild-type offspring of +/- eNOS fathers. The endocrine pancreas in wild-type offspring was not affected. CONCLUSIONS/INTERPRETATION Our study suggests that paternal genetic defects such as eNOS deficiency may alter the epigenome of the sperm without transmission of the paternal genetic defect itself. In later life wild-type male offspring of +/- eNOS fathers developed increased fasting insulin and increased insulin after glucose load. These effects are associated with increased Gr and Pgc1a gene expression due to altered methylation of these genes.
Collapse
Affiliation(s)
- Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.
- Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany.
| | - Yong-Ping Lu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | | | - Xiaoli Zhang
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Oleg Tsuprykov
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Jan Rahnenführer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Li Xie
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jian Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Liang Hu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
9
|
Bartoli F, Debant M, Chuntharpursat-Bon E, Evans EL, Musialowski KE, Parsonage G, Morley LC, Futers TS, Sukumar P, Bowen TS, Kearney MT, Lichtenstein L, Roberts LD, Beech DJ. Endothelial Piezo1 sustains muscle capillary density and contributes to physical activity. J Clin Invest 2022; 132:141775. [PMID: 35025768 PMCID: PMC8884896 DOI: 10.1172/jci141775] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
Piezo1 forms mechanically activated nonselective cation channels that contribute to endothelial response to fluid flow. Here we reveal an important role in the control of capillary density. Conditional endothelial cell-specific deletion of Piezo1 in adult mice depressed physical performance. Muscle microvascular endothelial cell apoptosis and capillary rarefaction were evident and sufficient to account for the effect on performance. There was selective upregulation of thrombospondin-2 (TSP2), an inducer of endothelial cell apoptosis, with no effect on TSP1, a related important player in muscle physiology. TSP2 was poorly expressed in muscle endothelial cells but robustly expressed in muscle pericytes, in which nitric oxide (NO) repressed the Tsp2 gene without an effect on Tsp1. In endothelial cells, Piezo1 was required for normal expression of endothelial NO synthase. The data suggest an endothelial cell-pericyte partnership of muscle in which endothelial Piezo1 senses blood flow to sustain capillary density and thereby maintain physical capability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - T. Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | | | |
Collapse
|
10
|
Liao FF, Lin G, Chen X, Chen L, Zheng W, Raghow R, Zhou FM, Shih AY, Tan XL. Endothelial Nitric Oxide Synthase-Deficient Mice: A Model of Spontaneous Cerebral Small-Vessel Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1932-1945. [PMID: 33711310 PMCID: PMC8647425 DOI: 10.1016/j.ajpath.2021.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Age-related cerebral small-vessel disease (CSVD) is a major cause of stroke and dementia. Despite a widespread acceptance of small-vessel arteriopathy, lacunar infarction, diffuse white matter injury, and cognitive impairment as four cardinal features of CSVD, a unifying pathologic mechanism of CSVD remains elusive. Herein, we introduce partial endothelial nitric oxide synthase (eNOS)-deficient mice as a model of age-dependent, spontaneous CSVD. These mice developed cerebral hypoperfusion and blood-brain barrier leakage at a young age, which progressively worsened with advanced age. Their brains exhibited elevated oxidative stress, astrogliosis, cerebral amyloid angiopathy, microbleeds, microinfarction, and white matter pathology. Partial eNOS-deficient mice developed gait disturbances at middle age, and hippocampus-dependent memory deficits at older ages. These mice also showed enhanced expression of bone morphogenetic protein 4 (BMP4) in brain pericytes before myelin loss and white matter pathology. Because BMP4 signaling not only promotes astrogliogenesis but also blocks oligodendrocyte differentiation, we posit that paracrine actions of BMP4, localized within the neurovascular unit, promote white matter disorganization and neurodegeneration. These observations point to BMP4 signaling pathway in the aging brain vasculature as a potential therapeutic target. Finally, because studies in partial eNOS-deficient mice corroborated recent clinical evidence that blood-brain barrier disruption is a primary cause of white matter pathology, the mechanism of impaired nitric oxide signaling-mediated CSVD warrants further investigation.
Collapse
Affiliation(s)
- Francesca-Fang Liao
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee.
| | - Geng Lin
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, China
| | - Xingyong Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wei Zheng
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, China
| | - Rajendra Raghow
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Fu-Ming Zhou
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Xing-Lin Tan
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Neurology, Nanhai Hospital of Southern Medical University, Foshan, China
| |
Collapse
|
11
|
Batista RIM, Nogueira RC, Ferreira GC, Oliveira-Paula GH, Damacena-Angelis C, Pinheiro LC, Tanus-Santos JE. Antiseptic mouthwash inhibits antihypertensive and vascular protective effects of L-arginine. Eur J Pharmacol 2021; 907:174314. [PMID: 34245745 DOI: 10.1016/j.ejphar.2021.174314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022]
Abstract
L-arginine supplementation increases nitric oxide (NO) formation and bioavailability in hypertension. We tested the possibility that many effects of L-arginine are mediated by increased formation of NO and enhanced nitrite, nitrate and nitrosylated species concentrations, thus stimulating the enterosalivary cycle of nitrate. Those effects could be prevented by antiseptic mouthwash. We examined how the derangement of the enterosalivary cycle of nitrate affects the improvement of endothelial dysfunction (assessed with isolated aortic ring preparation), the antihypertensive (assessed by tail-cuff blood pressure measurement) and the antioxidant effects (assessed with the fluorescent dye DHE) of L-arginine in two-kidney, one-clip hypertension model in rats by using chlorhexidine to decrease the number of oral bacteria and to decrease nitrate reductase activity assessed from the tongue (by ozone-based chemiluminiscence assay). Nitrite, nitrate and nitrosylated species concentrations were assessed (ozone-based chemiluminiscence). Chlorhexidine mouthwash reduced the number of oral bacteria and tended to decrease the nitrate reductase activity from the tongue. Antiseptic mouthwash blunted the improvement of the endothelial dysfunction and the antihypertensive effects of L-arginine, impaired L-arginine-induced increases in plasma nitrite and nitrosylated species concentrations, and blunted L-arginine-induced increases in aortic nitrate concentrations and vascular antioxidant effects. Our results show for the first time that the vascular and antihypertensive effects of L-arginine are prevented by antiseptic mouthwash. These findings show an important new mechanism that should be taken into consideration to explain how the use of antibacterial mouth rinse may affect arterial blood pressure and the risk of developing cardiovascular and other diseases.
Collapse
Affiliation(s)
- Rose I M Batista
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Renato C Nogueira
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Graziele C Ferreira
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Gustavo H Oliveira-Paula
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Célio Damacena-Angelis
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Brazil.
| |
Collapse
|
12
|
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: 96] [Impact Index Per Article: 32.0] [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
|
13
|
NOX5-induced uncoupling of endothelial NO synthase is a causal mechanism and theragnostic target of an age-related hypertension endotype. PLoS Biol 2020; 18:e3000885. [PMID: 33170835 PMCID: PMC7654809 DOI: 10.1371/journal.pbio.3000885] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hypertension is the most important cause of death and disability in the elderly. In 9 out of 10 cases, the molecular cause, however, is unknown. One mechanistic hypothesis involves impaired endothelium-dependent vasodilation through reactive oxygen species (ROS) formation. Indeed, ROS forming NADPH oxidase (Nox) genes associate with hypertension, yet target validation has been negative. We re-investigate this association by molecular network analysis and identify NOX5, not present in rodents, as a sole neighbor to human vasodilatory endothelial nitric oxide (NO) signaling. In hypertensive patients, endothelial microparticles indeed contained higher levels of NOX5—but not NOX1, NOX2, or NOX4—with a bimodal distribution correlating with disease severity. Mechanistically, mice expressing human Nox5 in endothelial cells developed—upon aging—severe systolic hypertension and impaired endothelium-dependent vasodilation due to uncoupled NO synthase (NOS). We conclude that NOX5-induced uncoupling of endothelial NOS is a causal mechanism and theragnostic target of an age-related hypertension endotype. Nox5 knock-in (KI) mice represent the first mechanism-based animal model of hypertension. The causes of hypertension are not understood; treatments are symptomatic and prevent only few of the associated risks. This study applies network medicine to identify a subgroup of patients with NADPH oxidase 5-induced uncoupling of nitric oxide synthase as the cause of age-related hypertension, enabling a first-in-class mechanism-based treatment of hypertension.
Collapse
|
14
|
de Freitas MAG, Amaral NO, Álvares ADCM, de Oliveira SA, Mehdad A, Honda DE, Bessa ASM, Ramada MHS, Naves LM, Pontes CNR, Castro CH, Pedrino GR, de Freitas SM. Blood pressure-lowering effects of a Bowman-Birk inhibitor and its derived peptides in normotensive and hypertensive rats. Sci Rep 2020; 10:11680. [PMID: 32669617 PMCID: PMC7363796 DOI: 10.1038/s41598-020-66624-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Bioactive plant peptides have received considerable interest as potential antihypertensive agents with potentially fewer side effects than antihypertensive drugs. Here, the blood pressure-lowering effects of the Bowman-Birk protease inhibitor, BTCI, and its derived peptides, PepChy and PepTry, were investigated using normotensive (Wistar-WR) and spontaneously hypertensive rats (SHR). BTCI inhibited the proteases trypsin and chymotrypsin, respectively, at 6 µM and 40 µM, a 10-fold greater inhibition than observed with PepTry (60 µM) and PepChy (400 µM). These molecules also inhibited angiotensin converting enzyme (ACE) with IC50 values of 54.6 ± 2.9; 24.7 ± 1.1; and 24.4 ± 1.1 µM, respectively, occluding its catalytic site, as indicated by molecular docking simulation, mainly for PepChy and PepTry. Gavage administration of BTCI and the peptides promoted a decrease of systolic and diastolic blood pressure and an increase of renal and aortic vascular conductance. These effects were more expressive in SHR than in WR. Additionally, BTCI, PepChy and PepTry promoted coronary vasodilation and negative inotropic effects in isolated perfused hearts. The nitric oxide synthase inhibitor blunted the BTCI and PepChy, with no cardiac effects on PepTry. The findings of this study indicate a therapeutic potential of BTCI and its related peptides in the treatment of hypertension.
Collapse
Affiliation(s)
- Maria Alzira Garcia de Freitas
- Biology Institute, Department of Cell Biology, Laboratory of Biophysics, University of Brasília (UnB), Quadra 604, Asa Norte, Bloco J 1° andar, Brasília, DF, 70910-900, Brazil
| | - Nathalia Oda Amaral
- Center of Neuroscience and Cardiovascular Physiology; Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - Alice da Cunha Morales Álvares
- Biology Institute, Department of Cell Biology, Laboratory of Biophysics, University of Brasília (UnB), Quadra 604, Asa Norte, Bloco J 1° andar, Brasília, DF, 70910-900, Brazil
| | - Sandriele Aires de Oliveira
- Biology Institute, Department of Cell Biology, Laboratory of Biophysics, University of Brasília (UnB), Quadra 604, Asa Norte, Bloco J 1° andar, Brasília, DF, 70910-900, Brazil
| | - Azadeh Mehdad
- Biology Institute, Department of Cell Biology, Laboratory of Biophysics, University of Brasília (UnB), Quadra 604, Asa Norte, Bloco J 1° andar, Brasília, DF, 70910-900, Brazil
| | - Diego Elias Honda
- Biology Institute, Department of Cell Biology, Laboratory of Biophysics, University of Brasília (UnB), Quadra 604, Asa Norte, Bloco J 1° andar, Brasília, DF, 70910-900, Brazil
| | - Amanda Sá Martins Bessa
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology; Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - Marcelo Henrique Soller Ramada
- Graduate Program in Genomic Science and Biotechnology, and Graduate Program in Gerontology, Catholic University of Brasília, Brasília, DF, 70790-160, Brazil
| | - Lara Marques Naves
- Center of Neuroscience and Cardiovascular Physiology; Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - Carolina Nobre Ribeiro Pontes
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology; Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - Carlos Henrique Castro
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology; Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - Gustavo Rodrigues Pedrino
- Center of Neuroscience and Cardiovascular Physiology; Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Sonia Maria de Freitas
- Biology Institute, Department of Cell Biology, Laboratory of Biophysics, University of Brasília (UnB), Quadra 604, Asa Norte, Bloco J 1° andar, Brasília, DF, 70910-900, Brazil.
| |
Collapse
|
15
|
Wischmann P, Kuhn V, Suvorava T, Muessig JM, Fischer JW, Isakson BE, Haberkorn SM, Flögel U, Schrader J, Jung C, Cortese-Krott MM, Heusch G, Kelm M. Anaemia is associated with severe RBC dysfunction and a reduced circulating NO pool: vascular and cardiac eNOS are crucial for the adaptation to anaemia. Basic Res Cardiol 2020; 115:43. [PMID: 32533377 PMCID: PMC7293199 DOI: 10.1007/s00395-020-0799-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023]
Abstract
Anaemia is frequently present in patients with acute myocardial infarction (AMI) and contributes to an adverse prognosis. We hypothesised that, besides reduced oxygen carrying capacity, anaemia is associated with (1) red blood cell (RBC) dysfunction and a reduced circulating nitric oxide (NO) pool, (2) compensatory enhancement of vascular and cardiac endothelial nitric oxide synthase (eNOS) activity, and (3) contribution of both, RBC dysfunction and reduced circulatory NO pool to left ventricular (LV) dysfunction and fatal outcome in AMI. In mouse models of subacute and chronic anaemia from repeated mild blood loss the circulating NO pool, RBC, cardiac and vascular function were analysed at baseline and in reperfused AMI. In anaemia, RBC function resulted in profound changes in membrane properties, enhanced turnover, haemolysis, dysregulation of intra-erythrocytotic redox state, and RBC-eNOS. RBC from anaemic mice and from anaemic patients with acute coronary syndrome impaired the recovery of contractile function of isolated mouse hearts following ischaemia/reperfusion. In anaemia, the circulating NO pool was reduced. The cardiac and vascular adaptation to anaemia was characterised by increased arterial eNOS expression and activity and an eNOS-dependent increase of end-diastolic left ventricular volume. Endothelial dysfunction induced through genetic or pharmacologic reduction of eNOS-activity abrogated the anaemia-induced cardio-circulatory compensation. Superimposed AMI was associated with decreased survival. In summary, moderate blood loss anaemia is associated with severe RBC dysfunction and reduced circulating NO pool. Vascular and cardiac eNOS are crucial for the cardio-circulatory adaptation to anaemia. RBC dysfunction together with eNOS dysfunction may contribute to adverse outcomes in AMI.
Collapse
Affiliation(s)
- Patricia Wischmann
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Viktoria Kuhn
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Johanna M Muessig
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jens W Fischer
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Department of Pharmacology and Clinical Pharmacology, Heinrich-Heine University, Düsseldorf, Germany
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sebastian M Haberkorn
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Ulrich Flögel
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Jürgen Schrader
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany. .,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
16
|
Assessment of tissue perfusion and vascular function in mice by scanning laser Doppler perfusion imaging. Biochem Pharmacol 2020; 176:113893. [DOI: 10.1016/j.bcp.2020.113893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/27/2020] [Indexed: 01/20/2023]
|
17
|
Topchieva LV, Balan OV, Korneva VA, Malysheva IE, Pankrasheva KA. The Nitric Oxide Metabolite Level and NOS2 and NOS3 Gene Transcripts in Patients with Essential Arterial Hypertension. BIOL BULL+ 2020. [DOI: 10.1134/s1062359020010161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Perniss A, Liu S, Boonen B, Keshavarz M, Ruppert AL, Timm T, Pfeil U, Soultanova A, Kusumakshi S, Delventhal L, Aydin Ö, Pyrski M, Deckmann K, Hain T, Schmidt N, Ewers C, Günther A, Lochnit G, Chubanov V, Gudermann T, Oberwinkler J, Klein J, Mikoshiba K, Leinders-Zufall T, Offermanns S, Schütz B, Boehm U, Zufall F, Bufe B, Kummer W. Chemosensory Cell-Derived Acetylcholine Drives Tracheal Mucociliary Clearance in Response to Virulence-Associated Formyl Peptides. Immunity 2020; 52:683-699.e11. [DOI: 10.1016/j.immuni.2020.03.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 12/25/2019] [Accepted: 03/13/2020] [Indexed: 12/24/2022]
|
19
|
Requirement of β1 integrin for endothelium-dependent vasodilation and collateral formation in hindlimb ischemia. Sci Rep 2019; 9:16931. [PMID: 31729436 PMCID: PMC6858366 DOI: 10.1038/s41598-019-53137-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 10/25/2019] [Indexed: 01/07/2023] Open
Abstract
An acute increase in blood flow triggers flow-mediated dilation (FMD), which is mainly mediated by endothelial nitric oxide synthase (eNOS). A long-term increase in blood flow chronically enlarges the arterial lumen, a process called arteriogenesis. In several common human diseases, these processes are disrupted for as yet unknown reasons. Here, we asked whether β1 integrin, a mechanosensory protein in endothelial cells, is required for FMD and arteriogenesis in the ischemic hindlimb. Permanent ligation of the femoral artery in C57BL/6 J mice enlarged pre-existing collateral arteries and increased numbers of arterioles in the thigh. In the lower leg, the numbers of capillaries increased. Notably, injection of β1 integrin-blocking antibody or tamoxifen-induced endothelial cell-specific deletion of the gene for β1 integrin (Itgb1) inhibited both arteriogenesis and angiogenesis. Using high frequency ultrasound, we demonstrated that β1 integrin-blocking antibody or endothelial cell-specific depletion of β1 integrin attenuated FMD of the femoral artery, and blocking of β1 integrin function did not further decrease FMD in eNOS-deficient mice. Our data suggest that endothelial β1 integrin is required for both acute and chronic widening of the arterial lumen in response to hindlimb ischemia, potentially via functional interaction with eNOS.
Collapse
|
20
|
Liu L, Mao L, Wu X, Wu T, Liu W, Yang Y, Zhang T, Xu Y. BRG1 regulates endothelial-derived IL-33 to promote ischemia-reperfusion induced renal injury and fibrosis in mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2551-2561. [DOI: 10.1016/j.bbadis.2019.06.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/30/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
|
21
|
Gericke A, Wolff I, Musayeva A, Zadeh JK, Manicam C, Pfeiffer N, Li H, Xia N. Retinal arteriole reactivity in mice lacking the endothelial nitric oxide synthase (eNOS) gene. Exp Eye Res 2019; 181:150-156. [PMID: 30716330 DOI: 10.1016/j.exer.2019.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/17/2019] [Accepted: 01/31/2019] [Indexed: 12/20/2022]
Abstract
Dysfunctional vascular endothelial nitric oxide synthase (eNOS) has been proposed to play a main pathophysiological role in various ocular diseases. The aim of the present study was to test the hypothesis that the chronic lack of eNOS impairs endothelium-dependent vasodilation in retinal arterioles. The relevance of eNOS for mediating vascular responses was studied in retinal vascular preparations from eNOS-deficient mice (eNOS-/-) and wild-type controls in vitro. Changes in luminal diameter in response to vasoactive agents were measured by videomicroscopy. The thromboxane mimetic, U46619, induced similar concentration-dependent constriction of retinal arterioles in eNOS-/- and wild-type mice. Responses to the endothelium-independent vasodilator, nitroprusside, did not differ between both mouse genotypes, either. In contrast, responses to the endothelium-dependent vasodilator, acetylcholine, were blunted in eNOS-/- mice. Non-isoform-selective blockade of either nitric oxide synthase (NOS) or cyclooxygenase (COX) alone did not affect responses to acetylcholine. However, combined blockade of both enzyme families markedly attenuated cholinergic vasodilation. Also, combined blockade of COX and neuronal NOS (nNOS) blunted acetylcholine-induced vasodilation, while combined COX and inducible NOS (iNOS) inhibition had no effect. Simultaneous NOS and COX-1 blockade did not affect cholinergic vasodilation, whereas combined NOS and COX-2 inhibition markedly reduced vasodilation to acetylcholine. These findings are the first to demonstrate that the chronic lack of eNOS is associated with moderate endothelial dysfunction in retinal arterioles. However, eNOS-deficiency is partially compensated by nNOS and COX-2 metabolites, which are reciprocally regulated.
Collapse
Affiliation(s)
- Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Ismael Wolff
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Aytan Musayeva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Jenia Kouchek Zadeh
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| |
Collapse
|
22
|
Schuler D, Sansone R, Nicolaus C, Kelm M, Heiss C. Repetitive remote occlusion (RRO) stimulates eNOS-dependent blood flow and collateral expansion in hindlimb ischemia. Free Radic Biol Med 2018; 129:520-531. [PMID: 30336250 DOI: 10.1016/j.freeradbiomed.2018.10.399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/13/2018] [Accepted: 10/01/2018] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Collateral expansion is an important compensatory mechanism to alleviate tissue ischemia after arterial occlusion. We investigated the efficacy and mechanisms of temporary remote hindlimb occlusion to stimulate contralateral blood flow and collateral expansion after hindlimb ischemia in mice and evaluated translation to peripheral artery disease in humans. METHODS AND RESULTS We induced unilateral hindlimb ischemia via femoral artery excision in mice. We studied central hemodynamics, blood flow, and perfusion of the ischemic hindlimb during single and repetitive remote occlusion (RRO) of the contralateral non-ischemic hindlimb with a pressurized cuff. Similar experiments were performed in patients with unilateral peripheral artery disease (PAD). Contralateral occlusion of the non-ischemic hindlimb led to an acute increase in blood flow to the ischemic hindlimb without affecting central blood pressure and cardiac output. The increase in blood flow was sustained even after deflation of the pressure cuff. RRO over 12 days (8/day, each 5 min) led to significantly increased arterial inflow, lumen expansion of collateral arteries, and increased perfusion of the chronically ischemic hindlimb as compared to control. In NOS3-/- and after inhibition of NOS (L-NAME), and NO (ODQ), the acute and chronic effects of contralateral occlusion were abrogated and stimulation of guanylyl cyclase with cinaciguate exhibited a similar response as RRO and was not additive. Pilot studies in PAD patients demonstrated that contralateral occlusion increased arterial inflow to ischemic limbs and improved walking distance. CONCLUSIONS Repetitive remote contralateral occlusion stimulates arterial inflow, perfusion, and functional collateral expansion in chronic hindlimb ischemia via an eNOS-dependent mechanism underscoring the potential of remote occlusion as a novel treatment option in peripheral artery disease.
Collapse
Affiliation(s)
- Dominik Schuler
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Roberto Sansone
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Christopher Nicolaus
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany; CARID - Cardiovascular research Institute Duesseldorf, University Duesseldorf, Duesseldorf, Germany
| | - Christian Heiss
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany.
| |
Collapse
|
23
|
Nunes ADC, Alves PH, Mendes EP, Ianzer D, Castro CH. BJ-PRO-7A and BJ-PRO-10C induce vasodilatation and inotropic effects in normotensive and hypertensive rats: Role of nitric oxide and muscarinic receptors. Peptides 2018; 110:1-9. [PMID: 30355532 DOI: 10.1016/j.peptides.2018.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/12/2018] [Accepted: 10/17/2018] [Indexed: 02/05/2023]
Abstract
Bj-PRO-7a and Bj-PRO-10c belong to a family of proline-rich oligopeptides (PROs) identified in Bothrops jararaca (Bj) crude venom. Previous studies have shown an antihypertensive effect evoked by theses peptides. However, the mechanisms underlying the direct effects on vessels and heart remain to be unraveled. Thus, we investigated the effect of the Bj-PRO-7a and Bj-PRO-10c in the aorta and coronary arteries and in cardiac contractility in normotensive (Wistar) and hypertensive (SHR) rats. Pre-constricted aortic rings were exposed to increasing concentrations of Bj-PROs in presence or absence of muscarinic type 1 receptor antagonist (Pirenzepine), nonselective muscarinic receptor antagonist (Atropine), nitric oxide synthase inhibitor (L-NAME), guanylyl cyclase inhibitor (ODQ), adenylyl cyclase inhibitor (MDL), or argininosuccinate synthetase inhibitor (MDLA). The effects of Bj-PROs in the cardiac contractility and coronary vasomotricity were evaluated using Langendorff perfused heart preparation. The rat hearts were perfused with Bj-PRO-7a or Bj-PRO-10c in absence or presence of L-NAME, ODQ or MDL. Both Bj-PROs induced endothelium-dependent vasorelaxation in aortic rings from Wistars and SHRs. These effects were inhibited by L-NAME, ODQ or MDL. Atropine and Pirenzepine blocked the vasorelaxant effect of Bj-PRO-7a in aorta from both strains. MDLA inhibited the Bj-PRO-10c-induced vasorelaxation in aortic rings from SHR, but not Wistar. The Bj-PRO-7a induced coronary vasodilation only in SHR. L-NAME, ODQ and MDL inhibited this effect. Bj-PRO-10c induced coronary vasodilatation in both strains, which was blocked by L-NAME, ODQ and MDL. Bj-PRO-7a decreased the dP/dt max in Wistar hearts and the dP/dt min in Wistar and SHR hearts. These effects were abolished by L-NAME. Bj-PRO-10c decreased dP/dt max and dP/dt min in hearts from normotensive and hypertensive animals, which were abolished in the presence of L-NAME, MDL and ODQ. In summary, the Bj-PROs induced endothelium-dependent vasorelaxation in rat thoracic aorta, coronary vasodilation and negative inotropic effects through mechanisms mediated by activation of nitric oxide pathway.
Collapse
Affiliation(s)
- A D C Nunes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - P H Alves
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - E P Mendes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil
| | - D Ianzer
- Laboratory of Neural and Cardiovascular Physiology and Therapeutics, Department of Physiological Sciences, Federal University of Goiás, Goiânia, 74690-900, Brazil
| | - C H Castro
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| |
Collapse
|
24
|
Olchanheski LR, Sordi R, Oliveira JG, Alves GF, Mendes RT, Santos FA, Fernandes D. The role of potassium channels in the endothelial dysfunction induced by periodontitis. J Appl Oral Sci 2018; 26:e20180048. [PMID: 30304126 PMCID: PMC6172022 DOI: 10.1590/1678-7757-2018-0048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/18/2018] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Periodontitis is associated with endothelial dysfunction, which is clinically characterized by a reduction in endothelium-dependent relaxation. However, we have previously shown that impairment in endothelium-dependent relaxation is transient. Therefore, we evaluated which mediators are involved in endothelium-dependent relaxation recovery. MATERIAL AND METHODS Rats were subjected to ligature-induced experimental periodontitis. Twenty-one days after the procedure, the animals were prepared for blood pressure recording, and the responses to acetylcholine or sodium nitroprusside were obtained before and 30 minutes after injection of a nitric oxide synthase inhibitor (L-NAME), cyclooxygenase inhibitor (Indomethacin, SC-550 and NS- 398), or calcium-dependent potassium channel blockers (apamin plus TRAM- 34). The maxilla and mandible were removed for bone loss analysis. Blood and gingivae were obtained for C-reactive protein (CRP) and myeloperoxidase (MPO) measurement, respectively. RESULTS Experimental periodontitis induces bone loss and an increase in the gingival MPO and plasmatic CRP. Periodontitis also reduced endothelium-dependent vasodilation, a hallmark of endothelial dysfunction, 14 days after the procedure. However, the response was restored at day 21. We found that endothelium-dependent vasodilation at day 21 in ligature animals was mediated, at least in part, by the activation of endothelial calcium-activated potassium channels. CONCLUSIONS Periodontitis induces impairment in endothelial-dependent relaxation; this impairment recovers, even in the presence of periodontitis. The recovery is mediated by the activation of endothelial calcium-activated potassium channels in ligature animals. Although important for maintenance of vascular homeostasis, this effect could mask the lack of NO, which has other beneficial properties.
Collapse
Affiliation(s)
- Luiz Renato Olchanheski
- Universidade Estadual de Ponta Grossa, Departamento de Ciências Farmacêuticas, Ponta Grossa, Paraná, Brasil
| | - Regina Sordi
- Universidade Estadual de Ponta Grossa, Departamento de Ciências Farmacêuticas, Ponta Grossa, Paraná, Brasil.,Universidade Estadual de Ponta Grossa, Departamento de Biologia Molecular e Genética, Ponta Grossa, Paraná, Brasil
| | - Junior Garcia Oliveira
- Universidade Estadual de Ponta Grossa, Departamento de Ciências Farmacêuticas, Ponta Grossa, Paraná, Brasil
| | - Gustavo Ferreira Alves
- Universidade Federal de Santa Catarina, Departamento de Farmacologia, Florianópolis, Santa Catarina, Brasil
| | - Reila Taina Mendes
- Universidade Estadual de Ponta Grossa, Departamento de Odontologia, Ponta Grossa, Paraná, Brasil
| | - Fábio André Santos
- Universidade Estadual de Ponta Grossa, Departamento de Odontologia, Ponta Grossa, Paraná, Brasil
| | - Daniel Fernandes
- Universidade Federal de Santa Catarina, Departamento de Farmacologia, Florianópolis, Santa Catarina, Brasil
| |
Collapse
|
25
|
Han L, Liu J, Zhu L, Tan F, Qin Y, Huang H, Yu Y. Free fatty acid can induce cardiac dysfunction and alter insulin signaling pathways in the heart. Lipids Health Dis 2018; 17:185. [PMID: 30089498 PMCID: PMC6083561 DOI: 10.1186/s12944-018-0834-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/26/2018] [Indexed: 02/05/2023] Open
Abstract
Background Insulin resistance has been independently related to heart failure. However, the specific mechanisms of high FFA levels in the pathophysiology of heart failure in insulin-resistant states are remain largely unclear. This study investigated whether elevated circulating free fatty acids (FFA) levels result in impaired cardiac structure and function in vivo via insulin-related signaling pathways in myocardium. Methods Male Wistar rats were randomly divided into the intralipid group (20% intralipid plus heparin infusion) and the control group (glycerol infusion). Blood samples were collected before and after 6-, 12-, and 24-h infusions. Cardiac structure and function were measured using echocardiography. Maximum velocity of myocardial contraction (+dP/dt max) and diastole (−dP/dt max) were measured using a physiological polygraph in vivo. Heart tissues were collected for western blotting. Results Compared with the control group, plasma FFA, plasma glucose, and serum insulin levels increased significantly in the intralipid group. With increasing infusion time, cardiac function in the intralipid group decreased gradually compared with the control group. After a 24-h infusion, early (E’, cm/s) diastolic peak velocities and (−dP/dt max) decreased significantly. Protein expression of phosphatidylinositol 3-kinase (PI3K), the serine/threonine kinase Akt, and phosphorylated Akt in myocardium increased after a 6-h infusion and decreased significantly after a 24-h infusion in the intralipid group. Protein expression of glucose transporter type 4 (GLUT4), Adenosine 5′-monophosphate -activated protein kinase (AMPK), phosphorylated AMPK(p-AMPK), and endothelial nitric oxide synthase (eNOS) in myocardium gradually decreased in the intralipid group. Conclusions Elevated FFA levels may impair cardiac function and cardiac dysfunction might result from myocardial insulin resistance with significant changes to PI3K-Akt-GLUT4 and AMPK-eNOS signaling pathways with increasing FFA levels.
Collapse
Affiliation(s)
- Lina Han
- Department of endocrinology and metabolism, West China Hospital, Sichuan University, Guoxue lane 37, Chengdu City, Sichuan Province, 610041, People's Republic of China
| | - Jiali Liu
- Department of endocrinology and metabolism, West China Hospital, Sichuan University, Guoxue lane 37, Chengdu City, Sichuan Province, 610041, People's Republic of China
| | - Leilei Zhu
- Department of endocrinology and metabolism, West China Hospital, Sichuan University, Guoxue lane 37, Chengdu City, Sichuan Province, 610041, People's Republic of China
| | - Fang Tan
- Department of endocrinology and metabolism, West China Hospital, Sichuan University, Guoxue lane 37, Chengdu City, Sichuan Province, 610041, People's Republic of China
| | - Yupei Qin
- Department of Cardiovascular, West China Hospital, Sichuan University, Chengdu City, Sichuan Province, People's Republic of China
| | - He Huang
- Department of Cardiovascular, West China Hospital, Sichuan University, Chengdu City, Sichuan Province, People's Republic of China
| | - Yerong Yu
- Department of endocrinology and metabolism, West China Hospital, Sichuan University, Guoxue lane 37, Chengdu City, Sichuan Province, 610041, People's Republic of China.
| |
Collapse
|
26
|
Diederich L, Suvorava T, Sansone R, Keller TCS, Barbarino F, Sutton TR, Kramer CM, Lückstädt W, Isakson BE, Gohlke H, Feelisch M, Kelm M, Cortese-Krott MM. On the Effects of Reactive Oxygen Species and Nitric Oxide on Red Blood Cell Deformability. Front Physiol 2018; 9:332. [PMID: 29867516 PMCID: PMC5958211 DOI: 10.3389/fphys.2018.00332] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/16/2018] [Indexed: 01/08/2023] Open
Abstract
The main function of red blood cells (RBCs) is the transport of respiratory gases along the vascular tree. To fulfill their task, RBCs are able to elastically deform in response to mechanical forces and, pass through the narrow vessels of the microcirculation. Decreased RBC deformability was observed in pathological conditions linked to increased oxidative stress or decreased nitric oxide (NO) bioavailability, like hypertension. Treatments with oxidants and with NO were shown to affect RBC deformability ex vivo, but the mechanisms underpinning these effects are unknown. In this study we investigate whether changes in intracellular redox status/oxidative stress or nitrosation reactions induced by reactive oxygen species (ROS) or NO may affect RBC deformability. In a case-control study comparing RBCs from healthy and hypertensive participants, we found that RBC deformability was decreased, and levels of ROS were increased in RBCs from hypertensive patients as compared to RBCs from aged-matched healthy controls, while NO levels in RBCs were not significantly different. To study the effects of oxidants on RBC redox state and deformability, RBCs from healthy volunteers were treated with increasing concentrations of tert-butylhydroperoxide (t-BuOOH). We found that high concentrations of t-BuOOH (≥ 1 mM) significantly decreased the GSH/GSSG ratio in RBCs, decreased RBC deformability and increased blood bulk viscosity. Moreover, RBCs from Nrf2 knockout (KO) mice, a strain genetically deficient in a number of antioxidant/reducing enzymes, were more susceptible to t-BuOOH-induced impairment in RBC deformability as compared to wild type (WT) mice. To study the role of NO in RBC deformability we treated RBC suspensions from human volunteers with NO donors and nitrosothiols and analyzed deformability of RBCs from mice lacking the endothelial NO synthase (eNOS). We found that NO donors induced S-nitrosation of the cytoskeletal protein spectrin, but did not affect human RBC deformability or blood bulk viscosity; moreover, under unstressed conditions RBCs from eNOS KO mice showed fully preserved RBC deformability as compared to WT mice. Pre-treatment of human RBCs with nitrosothiols rescued t-BuOOH-mediated loss of RBC deformability. Taken together, these findings suggest that NO does not affect RBC deformability per se, but preserves RBC deformability in conditions of oxidative stress.
Collapse
Affiliation(s)
- Lukas Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Roberto Sansone
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - T C Stevenson Keller
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Frederik Barbarino
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Thomas R Sutton
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Wiebke Lückstädt
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Holger Gohlke
- Faculty of Mathematics and Natural Sciences, Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
27
|
Nrf2 Deficiency Unmasks the Significance of Nitric Oxide Synthase Activity for Cardioprotection. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8309698. [PMID: 29854098 PMCID: PMC5952436 DOI: 10.1155/2018/8309698] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/17/2018] [Accepted: 02/27/2018] [Indexed: 12/14/2022]
Abstract
The transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key master switch that controls the expression of antioxidant and cytoprotective enzymes, including enzymes catalyzing glutathione de novo synthesis. In this study, we aimed to analyze whether Nrf2 deficiency influences antioxidative capacity, redox state, NO metabolites, and outcome of myocardial ischemia reperfusion (I/R) injury. In Nrf2 knockout (Nrf2 KO) mice, we found elevated eNOS expression and preserved NO metabolite concentrations in the aorta and heart as compared to wild types (WT). Unexpectedly, Nrf2 KO mice have a smaller infarct size following myocardial ischemia/reperfusion injury than WT mice and show fully preserved left ventricular systolic function. Inhibition of NO synthesis at onset of ischemia and during early reperfusion increased myocardial damage and systolic dysfunction in Nrf2 KO mice, but not in WT mice. Consistent with this, infarct size and diastolic function were unaffected in eNOS knockout (eNOS KO) mice after ischemia/reperfusion. Taken together, these data suggest that eNOS upregulation under conditions of decreased antioxidant capacity might play an important role in cardioprotection against I/R. Due to the redundancy in cytoprotective mechanisms, this fundamental antioxidant property of eNOS is not evident upon acute NOS inhibition in WT mice or in eNOS KO mice until Nrf2-related signaling is abrogated.
Collapse
|
28
|
Orphan receptor GPR37L1 contributes to the sexual dimorphism of central cardiovascular control. Biol Sex Differ 2018; 9:14. [PMID: 29625592 PMCID: PMC5889568 DOI: 10.1186/s13293-018-0173-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/27/2018] [Indexed: 11/29/2022] Open
Abstract
Background Over 100 mammalian G protein-coupled receptors are yet to be matched with endogenous ligands; these so-called orphans are prospective drug targets for the treatment of disease. GPR37L1 is one such orphan, abundant in the brain and detectable as mRNA in the heart and kidney. GPR37L1 ablation was reported to cause hypertension and left ventricular hypertrophy, and thus, we sought to further define the role of GPR37L1 in blood pressure homeostasis. Methods We investigated the cardiovascular effects of GPR37L1 using wild-type (GPR37L1wt/wt) and null (GPR37L1KO/KO) mice established on a C57BL/6J background, both under baseline conditions and during AngII infusion. We profiled GPR37L1 tissue expression, examining the endogenous receptor by immunoblotting and a β-galactosidase reporter mouse by immunohistochemistry. Results GPR37L1 protein was abundant in the brain but not detectable in the heart and kidney. We measured blood pressure in GPR37L1wt/wt and GPR37L1KO/KO mice and found that deletion of GPR37L1 causes a female-specific increase in systolic, diastolic, and mean arterial pressures. When challenged with short-term AngII infusion, only male GPR37L1KO/KO mice developed exacerbated left ventricular hypertrophy and evidence of heart failure, while the female GPR37L1KO/KO mice were protected from cardiac fibrosis. Conclusions Despite its absence in the heart and kidney, GPR37L1 regulates baseline blood pressure in female mice and is crucial for cardiovascular compensatory responses in males. The expression of GPR37L1 in the brain, yet absence from peripheral cardiovascular tissues, suggests this orphan receptor is a hitherto unknown contributor to central cardiovascular control. Electronic supplementary material The online version of this article (10.1186/s13293-018-0173-y) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Wolhuter K, Whitwell HJ, Switzer CH, Burgoyne JR, Timms JF, Eaton P. Evidence against Stable Protein S-Nitrosylation as a Widespread Mechanism of Post-translational Regulation. Mol Cell 2018; 69:438-450.e5. [PMID: 29358077 PMCID: PMC5807093 DOI: 10.1016/j.molcel.2017.12.019] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/01/2017] [Accepted: 12/20/2017] [Indexed: 11/27/2022]
Abstract
S-nitrosation, commonly referred to as S-nitrosylation, is widely regarded as a ubiquitous, stable post-translational modification that directly regulates many proteins. Such a widespread role would appear to be incompatible with the inherent lability of the S-nitroso bond, especially its propensity to rapidly react with thiols to generate disulfide bonds. As anticipated, we observed robust and widespread protein S-nitrosation after exposing cells to nitrosocysteine or lipopolysaccharide. Proteins detected using the ascorbate-dependent biotin switch method are typically interpreted to be directly regulated by S-nitrosation. However, these S-nitrosated proteins are shown to predominantly comprise transient intermediates leading to disulfide bond formation. These disulfides are likely to be the dominant end effectors resulting from elevations in nitrosating cellular nitric oxide species. We propose that S-nitrosation primarily serves as a transient intermediate leading to disulfide formation. Overall, we conclude that the current widely held perception that stable S-nitrosation directly regulates the function of many proteins is significantly incorrect. Protein S-nitrosation is commonly regarded as a stable, regulatory modification However, S-nitrosothiols are labile and rapidly react with thiols to form disulfides Here disulfides are shown to be the dominant end effectors of nitrosative signaling Protein S-nitrosation as a regulatory end effector may occur, but this may be rare
Collapse
Affiliation(s)
- Kathryn Wolhuter
- King's College London, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre of Research Excellence the Rayne Institute, St. Thomas' Hospital, London SE1 7EH, UK
| | - Harry J Whitwell
- Institute for Women's Health, University College London, Gower Street, London WC1E 6BT, UK
| | - Christopher H Switzer
- King's College London, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre of Research Excellence the Rayne Institute, St. Thomas' Hospital, London SE1 7EH, UK
| | - Joseph R Burgoyne
- King's College London, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre of Research Excellence the Rayne Institute, St. Thomas' Hospital, London SE1 7EH, UK
| | - John F Timms
- Institute for Women's Health, University College London, Gower Street, London WC1E 6BT, UK
| | - Philip Eaton
- King's College London, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre of Research Excellence the Rayne Institute, St. Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
30
|
Ebner A, Kuerbis N, Brandt A, Zatschler B, Weinert S, Poitz DM, Ebner B, Augstein A, Wunderlich C, El-Armouche A, Strasser RH. Endothelial Nitric Oxide Synthase-Induced Hypertrophy and Vascular Dysfunction Contribute to the Left Ventricular Dysfunction in Caveolin-1−/− Mice. Can J Cardiol 2017; 33:1716-1724. [DOI: 10.1016/j.cjca.2017.09.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 02/02/2023] Open
|
31
|
Ertuna E, Loot AE, Fleming I, Yetik-Anacak G. The role of eNOS on the compensatory regulation of vascular tonus by H 2 S in mouse carotid arteries. Nitric Oxide 2017; 69:45-50. [DOI: 10.1016/j.niox.2017.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/06/2017] [Accepted: 04/12/2017] [Indexed: 11/30/2022]
|
32
|
Northcott JM, Czubryt MP, Wigle JT. Vascular senescence and ageing: a role for the MEOX proteins in promoting endothelial dysfunction. Can J Physiol Pharmacol 2017; 95:1067-1077. [PMID: 28727928 DOI: 10.1139/cjpp-2017-0149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the vascular system, ageing is accompanied by the accrual of senescent cells and is associated with an increased risk of vascular disease. Endothelial cell (EC) dysfunction is a hallmark of vascular disease and is characterized by decreased angiogenic potential, reduced nitric oxide bioavailability, impaired vasodilation, increased production of ROS, and enhanced inflammation. In ECs, the major producer of nitric oxide is the endothelial nitric oxide synthase (eNOS) enzyme that is encoded by the NOS3 gene. NOS3/eNOS function is tightly regulated at both the transcriptional and post-transcriptional levels to maintain normal vascular function. A key transcriptional regulator of eNOS expression is p53, which has been shown to play a central role in mediating cellular senescence and thereby vascular dysfunction. Herein, we show that, in ECs, the MEOX homeodomain transcription factors decrease the expression of genes involved in angiogenesis, repress eNOS expression at the mRNA and protein levels, and increase the expression of p53. These findings support a role for the MEOX proteins in promoting endothelial dysfunction.
Collapse
Affiliation(s)
- Josette M Northcott
- a Institute of Cardiovascular Sciences, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael P Czubryt
- a Institute of Cardiovascular Sciences, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,c Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jeffrey T Wigle
- a Institute of Cardiovascular Sciences, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
33
|
Merz T, Stenzel T, Nußbaum B, Wepler M, Szabo C, Wang R, Radermacher P, McCook O. Cardiovascular disease and resuscitated septic shock lead to the downregulation of the H 2S-producing enzyme cystathionine-γ-lyase in the porcine coronary artery. Intensive Care Med Exp 2017; 5:17. [PMID: 28321823 PMCID: PMC5359268 DOI: 10.1186/s40635-017-0131-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/13/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Downregulation of the hydrogen sulfide (H2S)-producing enzymes cystathionine-γ-lyase (CSE), cystathionine-β-synthase (CBS), and/or 3-mercaptopyruvate sulfurtransferase (3-MST) is associated with chronic cardiovascular pathologies. Nevertheless, equivocal data are available on both the expression and function of these enzymes in coronary arteries (CA). We recently reported that atherosclerotic pigs subjected to sepsis developed impaired cardiac function, which coincided with decreased myocardial CSE expression and increased nitrotyrosine formation. To define the endogenous source(s) of H2S in the CA, we studied the expression of CBS, CSE, or 3-MST in the CA of pigs subjected to septic shock with/without pre-existing cardiovascular co-morbidity. METHODS Anesthetized and instrumented FBM "familial hypercholesterolemia Bretoncelles Meishan" pigs with high-fat diet-induced hypercholesterolemia and atherosclerosis were subjected to polymicrobial septic shock, or sham procedure, and subsequent intensive care therapy for 24 h. Young German domestic pigs were used as naïve controls. CSE, CBS, 3-MST, HO-1, eNOS, and nitrotyrosine expression was quantified by immunohistochemistry of formalin-fixed paraffin sections. RESULTS FBM pigs, in the absence of septic shock, showed decreased CSE expression in the media. This decrease became more pronounced after sepsis. The expression pattern of HO-1 resembled the pattern of CSE expression. CBS protein was not detected in the media of any of the CA examined but was localized to the adventitia and only in the atheromatous plaques containing foam cells of the CA, in regions that also displayed abundant nitrotyrosine formation. The CBS expression in the adventitia was not associated with nitrotyrosine formation. 3-MST expression was not found in any of the CA samples. CONCLUSIONS We hypothesize that (i) the reduced CSE expression in FBM pigs may contribute to their cardiovascular disease phenotype and moreover (ii) the further decrease in CA CSE expression in sepsis may contribute to the sepsis-associated cardiac dysfunction.
Collapse
Affiliation(s)
- Tamara Merz
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany
| | - Tatjana Stenzel
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany
| | - Benedikt Nußbaum
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany.,Universitätsklinik Ulm, Klinik für Anästhesiologie, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Martin Wepler
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany.,Universitätsklinik Ulm, Klinik für Anästhesiologie, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, 601 Harborside Drive, Galveston, TX, 77555, USA
| | - Rui Wang
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Peter Radermacher
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany
| | - Oscar McCook
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany. .,Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.
| |
Collapse
|
34
|
Thieme M, Sivritas SH, Mergia E, Potthoff SA, Yang G, Hering L, Grave K, Hoch H, Rump LC, Stegbauer J. Phosphodiesterase 5 inhibition ameliorates angiotensin II-dependent hypertension and renal vascular dysfunction. Am J Physiol Renal Physiol 2017; 312:F474-F481. [PMID: 28052870 DOI: 10.1152/ajprenal.00376.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/23/2016] [Accepted: 01/03/2017] [Indexed: 01/24/2023] Open
Abstract
Changes in renal hemodynamics have a major impact on blood pressure (BP). Angiotensin (Ang) II has been shown to induce vascular dysfunction by interacting with phosphodiesterase (PDE)1 and PDE5. The predominant PDE isoform responsible for renal vascular dysfunction in hypertension is unknown. Here, we measured the effects of PDE5 (sildenafil) or PDE1 (vinpocetine) inhibition on renal blood flow (RBF), BP, and renal vascular function in normotensive and hypertensive mice. During acute short-term Ang II infusion, sildenafil decreased BP and increased RBF in C57BL/6 (WT) mice. In contrast, vinpocetine showed no effect on RBF and BP. Additionally, renal cGMP levels were significantly increased after acute sildenafil but not after vinpocetine infusion, indicating a predominant role of PDE5 in renal vasculature. Furthermore, chronic Ang II infusion (500 ng·kg-1·min-1) increased BP and led to impaired NO-dependent vasodilation in kidneys of WT mice. Additional treatment with sildenafil (100 mg·kg-1·day-1) attenuated Ang II-dependent hypertension and improved NO-mediated vasodilation. During chronic Ang II infusion, urinary nitrite excretion, a marker for renal NO generation, was increased in WT mice, whereas renal cGMP generation was decreased and restored after sildenafil treatment, suggesting a preserved cGMP signaling after PDE5 inhibition. To investigate the dependency of PDE5 effects on NO/cGMP signaling, we next analyzed eNOS-KO mice, a mouse model characterized by low vascular NO/cGMP levels. In eNOS-KO mice, chronic Ang II infusion increased BP but did not impair NO-mediated vasodilation. Moreover, sildenafil did not influence BP or vascular function in eNOS-KO mice. These results highlight PDE5 as a key regulator of renal hemodynamics in hypertension.
Collapse
Affiliation(s)
- Manuel Thieme
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Sema H Sivritas
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Evanthia Mergia
- Department of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Sebastian A Potthoff
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Guang Yang
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Lydia Hering
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Katharina Grave
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Henning Hoch
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Lars C Rump
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; and
| |
Collapse
|
35
|
Effects of chronic nitric oxide synthase inhibition on V'O 2max and exercise capacity in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2016; 390:235-244. [PMID: 27915453 DOI: 10.1007/s00210-016-1318-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 11/14/2016] [Indexed: 10/20/2022]
Abstract
Acute inhibition of NOS by L-NAME (Nω-nitro-L-arginine methyl ester) is known to decrease maximal oxygen consumption (V'O2max) and impair maximal exercise capacity, whereas the effects of chronic L-NAME treatment on V'O2max and exercise performance have not been studied so far. In this study, we analysed the effect of L-NAME treatment, (LN2 and LN12, respectively) on V'O2max and exercise capacity (in maximal incremental running and prolonged sub-maximal incremental running tests), systemic NO bioavailability (plasma nitrite (NO2-) and nitrate (NO3-)) and prostacyclin (PGI2) production in C57BL6/J mice. Mice treated with L-NAME for 2 weeks (LN2) displayed higher V'O2max and better running capacity than age-matched control mice. In LN2 mice, NO bioavailability was preserved, as evidenced by maintained NO2- plasma concentration. PGI2 production was activated (increased 6-keto-PGF1α plasma concentration) and the number of circulating erythrocytes (RBC) and haemoglobin concentration were increased. In mice treated with L-NAME for 12 weeks (LN12), NO bioavailability was decreased (lower NO2- plasma concentration), and 6-keto-PGF1α plasma concentration and RBC number were not elevated compared to age-matched control mice. However, LN12 mice still performed better during the maximal incremental running test despite having lower V'O2max. Interestingly, the LN12 mice showed poorer running capacity during the prolonged sub-maximal incremental running test. To conclude, short-term (2 weeks) but not long-term (12 weeks) treatment with L-NAME activated robust compensatory mechanisms involving preservation of NO2- plasma concentration, overproduction of PGI2 and increased number of RBCs, which might explain the fully preserved exercise capacity despite the inhibition of NOS.
Collapse
|
36
|
Nitric oxide up-regulates endothelial expression of angiotensin II type 2 receptors. Biochem Pharmacol 2016; 112:24-36. [PMID: 27235748 DOI: 10.1016/j.bcp.2016.05.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/24/2016] [Indexed: 11/21/2022]
Abstract
Increasing vascular NO levels following up-regulation of endothelial nitric oxide synthase (eNOS) is considered beneficial in cardiovascular disease. Whether such beneficial effects exerted by increased NO-levels include the vascular renin-angiotensin system remains elucidated. Exposure of endothelial cells originated from porcine aorta, mouse brain and human umbilical veins to different NO-donors showed that expression of the angiotensin-II-type-2-receptor (AT2) mRNA and protein is up-regulated by activation of soluble guanylyl cyclase, protein kinase G and p38 mitogen-activated protein kinase without changing AT2 mRNA stability. In mice, endothelial-specific overexpression of eNOS stimulated, while chronic treatment with the NOS-blocker l-nitroarginine inhibited AT2 expression. The NO-induced AT2 up-regulation was associated with a profound inhibition of angiotensin-converting enzyme (ACE)-activity. In endothelial cells this reduction of ACE-activity was reversed by either the AT2 antagonist PD 123119 or by inhibition of transcription with actinomycin D. Furthermore, in C57Bl/6 mice an acute i.v. bolus of l-nitroarginine did not change AT2-expression and ACE-activity suggesting that inhibition of ACE-activity by endogenous NO is crucially dependent on AT2 protein level. Likewise, three weeks of either voluntary or forced exercise training increased AT2 expression and reduced ACE-activity in C57Bl/6 but not in mice lacking eNOS suggesting significance of this signaling interaction for vascular physiology. Finally, aortic AT2 expression is about 5 times greater in female as compared to male C57Bl/6 and at the same time aortic ACE activity is reduced in females by more than 50%. Together these findings imply that endothelial NO regulates AT2 expression and that AT2 may regulate ACE-activity.
Collapse
|
37
|
Hocher B, Haumann H, Rahnenführer J, Reichetzeder C, Kalk P, Pfab T, Tsuprykov O, Winter S, Hofmann U, Li J, Püschel GP, Lang F, Schuppan D, Schwab M, Schaeffeler E. Maternal eNOS deficiency determines a fatty liver phenotype of the offspring in a sex dependent manner. Epigenetics 2016; 11:539-52. [PMID: 27175980 PMCID: PMC4939931 DOI: 10.1080/15592294.2016.1184800] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Maternal environmental factors can impact on the phenotype of the offspring via the induction of epigenetic adaptive mechanisms. The advanced fetal programming hypothesis proposes that maternal genetic variants may influence the offspring's phenotype indirectly via epigenetic modification, despite the absence of a primary genetic defect. To test this hypothesis, heterozygous female eNOS knockout mice and wild type mice were bred with male wild type mice. We then assessed the impact of maternal eNOS deficiency on the liver phenotype of wild type offspring. Birth weight of male wild type offspring born to female heterozygous eNOS knockout mice was reduced compared to offspring of wild type mice. Moreover, the offspring displayed a sex specific liver phenotype, with an increased liver weight, due to steatosis. This was accompanied by sex specific differences in expression and DNA methylation of distinct genes. Liver global DNA methylation was significantly enhanced in both male and female offspring. Also, hepatic parameters of carbohydrate metabolism were reduced in male and female offspring. In addition, male mice displayed reductions in various amino acids in the liver. Maternal genetic alterations, such as partial deletion of the eNOS gene, can affect liver metabolism of wild type offspring without transmission of the intrinsic defect. This occurs in a sex specific way, with more detrimental effects in females. This finding demonstrates that a maternal genetic defect can epigenetically alter the phenotype of the offspring, without inheritance of the defect itself. Importantly, these acquired epigenetic phenotypic changes can persist into adulthood.
Collapse
Affiliation(s)
- Berthold Hocher
- a Department of Basic Medicine , Medical College of Hunan Normal University , Changsha , China.,b Institute of Nutritional Science, University of Potsdam , Nuthetal , Germany.,c IFLb, Laboratoriumsmedizin Berlin GmbH , Berlin , Germany
| | - Hannah Haumann
- d Department of Internal Medicine IV/Nephrology (UKBF) , Charité Campus Benjamin Franklin , Berlin , Germany.,e Center for Cardiovascular Research, Charité Campus Mitte , Berlin , Germany
| | - Jan Rahnenführer
- b Institute of Nutritional Science, University of Potsdam , Nuthetal , Germany
| | | | - Philipp Kalk
- d Department of Internal Medicine IV/Nephrology (UKBF) , Charité Campus Benjamin Franklin , Berlin , Germany.,e Center for Cardiovascular Research, Charité Campus Mitte , Berlin , Germany
| | - Thiemo Pfab
- d Department of Internal Medicine IV/Nephrology (UKBF) , Charité Campus Benjamin Franklin , Berlin , Germany.,f Diaverum Deutschland , Potsdam , Germany
| | - Oleg Tsuprykov
- b Institute of Nutritional Science, University of Potsdam , Nuthetal , Germany.,e Center for Cardiovascular Research, Charité Campus Mitte , Berlin , Germany
| | - Stefan Winter
- g Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology , Stuttgart , Germany.,h University of Tübingen , Tübingen , Germany
| | - Ute Hofmann
- g Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology , Stuttgart , Germany.,h University of Tübingen , Tübingen , Germany
| | - Jian Li
- a Department of Basic Medicine , Medical College of Hunan Normal University , Changsha , China
| | - Gerhard P Püschel
- b Institute of Nutritional Science, University of Potsdam , Nuthetal , Germany
| | - Florian Lang
- i Institute of Physiology, University of Tübingen , Tübingen , Germany
| | - Detlef Schuppan
- j Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany.,k Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| | - Matthias Schwab
- g Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology , Stuttgart , Germany.,l Department of Clinical Pharmacology , University Hospital Tübingen , Tübingen , Germany.,m Department of Pharmacy and Biochemistry , University Tübingen , Tübingen , Germany
| | - Elke Schaeffeler
- g Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology , Stuttgart , Germany.,h University of Tübingen , Tübingen , Germany
| |
Collapse
|
38
|
Campi P, Herrera BS, de Jesus FN, Napolitano M, Teixeira SA, Maia-Dantas A, Spolidorio LC, Akamine EH, Mayer MPA, de Carvalho MHC, Costa SKP, Muscara MN. Endothelial dysfunction in rats with ligature-induced periodontitis: Participation of nitric oxide and cycloxygenase-2-derived products. Arch Oral Biol 2015; 63:66-74. [PMID: 26691575 DOI: 10.1016/j.archoralbio.2015.11.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/06/2015] [Accepted: 11/29/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Considering the evident relationship between periodontitis and cardiovascular diseases in humans, we aimed to study the in vitro vascular reactivity of aorta rings prepared from rats with ligature-induced periodontitis. METHODS Seven days after the induction of unilateral periodontitis, the animals were euthanised; rings were prepared from the descending abdominal aortas and mounted in tissue baths for the in vitro measurement of the isometric force responses to norepinephrine (NE) and acetylcholine (ACh), as well as in the presence of inhibitors of nitric oxide synthase (NOS) and cycloxygenase (COX) isoenzymes. Aortic COX and NOS gene expressions were analysed by RT-PCR, as well as protein COX-2 expression by Western blot. RESULTS Periodontitis resulted in significant alveolar bone loss and did not affect arterial pressure. However, both NE-induced contraction and ACh-induced relaxation were significantly decreased and related to the presence of endothelium. Diminished eNOS and augmented COX-2 and iNOS expressions were found in the aortas from rats with periodontitis, and the pharmacological inhibition of COX-2 or iNOS improved the observed vasomotor deficiencies. CONCLUSIONS We can thus conclude that periodontitis induces significant endothelial dysfunction in rat aorta which is characterized by decreased eNOS expression and mediated by upregulated iNOS and COX-2 products.
Collapse
Affiliation(s)
- Paula Campi
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Bruno Schneider Herrera
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Department of Physiology and Pathology, Araraquara School of Dentistry, Sao Paulo State University, Araraquara, SP, Brazil
| | - Flavia Neto de Jesus
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mauro Napolitano
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Simone Aparecida Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Aline Maia-Dantas
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luis Carlos Spolidorio
- Department of Physiology and Pathology, Araraquara School of Dentistry, Sao Paulo State University, Araraquara, SP, Brazil
| | - Eliana Hiromi Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Soraia Katia Pereira Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marcelo Nicolas Muscara
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
39
|
Erkens R, Kramer CM, Lückstädt W, Panknin C, Krause L, Weidenbach M, Dirzka J, Krenz T, Mergia E, Suvorava T, Kelm M, Cortese-Krott MM. Left ventricular diastolic dysfunction in Nrf2 knock out mice is associated with cardiac hypertrophy, decreased expression of SERCA2a, and preserved endothelial function. Free Radic Biol Med 2015; 89:906-17. [PMID: 26475037 DOI: 10.1016/j.freeradbiomed.2015.10.409] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/09/2015] [Indexed: 11/20/2022]
Abstract
Increased production of reactive oxygen species and failure of the antioxidant defense system are considered to play a central role in the pathogenesis of cardiovascular disease. The transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key master switch controlling the expression of antioxidant and protective enzymes, and was proposed to participate in protection of vascular and cardiac function. This study was undertaken to analyze cardiac and vascular phenotype of mice lacking Nrf2. We found that Nrf2 knock out (Nrf2 KO) mice have a left ventricular (LV) diastolic dysfunction, characterized by prolonged E wave deceleration time, relaxation time and total diastolic time, increased E/A ratio and myocardial performance index, as assessed by echocardiography. LV dysfunction in Nrf2 KO mice was associated with cardiac hypertrophy, and a downregulation of the sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2a) in the myocardium. Accordingly, cardiac relaxation was impaired, as demonstrated by decreased responses to β-adrenergic stimulation by isoproterenol ex vivo, and to the cardiac glycoside ouabain in vivo. Surprisingly, we found that vascular endothelial function and endothelial nitric oxide synthase (eNOS)-mediated vascular responses were fully preserved, blood pressure was decreased, and eNOS was upregulated in the aorta and the heart of Nrf2 KO mice. Taken together, these results show that LV dysfunction in Nrf2 KO mice is mainly associated with cardiac hypertrophy and downregulation of SERCA2a, and is independent from changes in coronary vascular function or systemic hemodynamics, which are preserved by a compensatory upregulation of eNOS. These data provide new insights into how Nrf2 expression/function impacts the cardiovascular system.
Collapse
Affiliation(s)
- Ralf Erkens
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Wiebke Lückstädt
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Christina Panknin
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Lisann Krause
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Mathias Weidenbach
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Jennifer Dirzka
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Thomas Krenz
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Evanthia Mergia
- Institute for Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
40
|
Lendeckel U, Wolke C, Bernstein HG, Keilhoff G. Effects of nitric oxide synthase deficiency on a disintegrin and metalloproteinase domain-containing protein 12 expression in mouse brain samples. Mol Med Rep 2015; 12:2253-62. [PMID: 25892053 DOI: 10.3892/mmr.2015.3643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/03/2015] [Indexed: 11/06/2022] Open
Abstract
A disintegrin and metalloproteinase domain-containing protein 12 (ADAM12) belongs to the ADAM family of transmembrane proteins. Via proteolysis, cell adhesion, cell-cell fusion, cell-matrix interaction and membrane protein shedding, ADAM proteins are involved in normal brain development, and also in cancer genesis and progression, and in inflammation. Therefore, neurobiological research focusing on this protein is increasing. Nitric oxide (NO), which is endogenously produced by NO synthases (NOS), is associated with glial tumors. However, knock-out of NOS produces only limited antitumor effects. The present study analyzed the expression of ADAM12 in the cortex and hippocampus of C57/BL6 wild-type mice, and endothelial NOS-, neuronal NOS-(nNOS) or inducible NOS (iNOS)-deficient (-/-) mice, at different stages of development. Expression of ADAM12 was quantified using immunoblot analysis of cortical and hippocampal tissue samples from fetal, neonatal (5 days postnatal), adult (12 weeks old) or >1 year old mice. Using reverse transcription-quantitative polymerase chain reaction, ADAM12 expression was analyzed in cultured N9, OLN93, C6 and PC12 cells, representing the four main cell types in the brain, following NOS inhibition. ADAM12 expression was low in all mouse genotypes and regions of the brain, and in fetal and neonatal mice, an increase in expression was observed with increasing age. The highest levels of expression were observed in the cortex of adult mice, iNOS(-/-) mice of >1 year and wild-type mice, and in the hippocampus of adult and iNOS(-/-) mice of >1 year. By contrast, ADAM12 expression was lowest in adult nNOS(-/-) mice. Inhibition of NOS using N(ω)-Nitro-L-arginine methyl ester hydrochloride, induced ADAM12 mRNA expression in N9 and PC12 cell lines. Inhibition of NOS using L-N(6)-(1-Iminoethyl)lysine dihydrochloride, induced ADAM12 mRNA expression in N9 and C6 cell lines. No change in ADAM12 expression was observed in OLN93 cells following NOS inhibition. ADAM12 expression in mouse hippocampus and cortex samples demonstrated considerable variation during development, with a marked increase observed in adult and >1 year old mice, compared with that in fetal and neonatal mice.
Collapse
Affiliation(s)
- Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Ernst‑Moritz‑Arndt University, Greifswald D‑17475, Germany
| | - Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Ernst‑Moritz‑Arndt University, Greifswald D‑17475, Germany
| | - Hans-Gert Bernstein
- Clinic of Psychiatry, Psychotherapy and Psychosomatic Medicine, Otto‑von‑Guericke University, Magdeburg D‑39120, Germany
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto‑von‑Guericke University, Magdeburg D‑39120, Germany
| |
Collapse
|
41
|
Gorressen S, Stern M, van de Sandt AM, Cortese-Krott MM, Ohlig J, Rassaf T, Gödecke A, Fischer JW, Heusch G, Merx MW, Kelm M. Circulating NOS3 modulates left ventricular remodeling following reperfused myocardial infarction. PLoS One 2015; 10:e0120961. [PMID: 25875863 PMCID: PMC4397096 DOI: 10.1371/journal.pone.0120961] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/27/2015] [Indexed: 12/21/2022] Open
Abstract
Purpose Nitric oxide (NO) is constitutively produced and released from the endothelium and several blood cell types by the isoform 3 of the NO synthase (NOS3). We have shown that NO protects against myocardial ischemia/reperfusion (I/R) injury and that depletion of circulating NOS3 increases within 24h of ischemia/reperfusion the size of myocardial infarction (MI) in chimeric mice devoid of circulating NOS3. In the current study we hypothesized that circulating NOS3 also affects remodeling of the left ventricle following reperfused MI. Methods To analyze the role of circulating NOS3 we transplanted bone marrow of NOS3−/− and wild type (WT) mice into WT mice, producing chimerae expressing NOS3 only in vascular endothelium (BC−/EC+) or in both, blood cells and vascular endothelium (BC+/EC+). Both groups underwent 60 min of coronary occlusion in a closed-chest model of reperfused MI. During the 3 weeks post MI, structural and functional LV remodeling was serially assessed (24h, 4d, 1w, 2w and 3w) by echocardiography. At 72 hours post MI, gene expression of several extracellular matrix (ECM) modifying molecules was determined by quantitative RT-PCR analysis. At 3 weeks post MI, hemodynamics were obtained by pressure catheter, scar size and collagen content were quantified post mortem by Gomori’s One-step trichrome staining. Results Three weeks post MI, LV end-systolic (53.2±5.9μl;***p≤0.001;n = 5) and end-diastolic volumes (82.7±5.6μl;*p<0.05;n = 5) were significantly increased in BC−/EC+, along with decreased LV developed pressure (67.5±1.8mmHg;n = 18;***p≤0.001) and increased scar size/left ventricle (19.5±1.5%;n = 13;**p≤0.01) compared to BC+/EC+ (ESV:35.6±2.2μl; EDV:69.1±2.6μl n = 8; LVDP:83.2±3.2mmHg;n = 24;scar size/LV13.8±0.7%;n = 16). Myocardial scar of BC−/EC+ was characterized by increased total collagen content (20.2±0.8%;n = 13;***p≤0.001) compared to BC+/EC+ (15.9±0.5;n = 16), and increased collagen type I and III subtypes. Conclusion Circulating NOS3 ameliorates maladaptive left ventricular remodeling following reperfused myocardial infarction.
Collapse
Affiliation(s)
- Simone Gorressen
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Manuel Stern
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Annette M. van de Sandt
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Miriam M. Cortese-Krott
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jan Ohlig
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tienush Rassaf
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Axel Gödecke
- Medical Faculty, Department of Cardiovascular Physiology, Heinrich-Heine-University, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
| | - Jens W. Fischer
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
- Medical Faculty, Institute of Pharmacology und Clinical Pharmacology, Heinrich Heine University, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Marc W. Merx
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Cardiology, Vascular Medicine and Intensive Care Medicine, Robert Koch Krankenhaus, Klinikum Region Hannover, Hannover, Germany
| | - Malte Kelm
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
42
|
Suvorava T, Stegbauer J, Thieme M, Pick S, Friedrich S, Rump LC, Hohlfeld T, Kojda G. Sustained hypertension despite endothelial-specific eNOS rescue in eNOS-deficient mice. Biochem Biophys Res Commun 2015; 458:576-583. [PMID: 25680465 DOI: 10.1016/j.bbrc.2015.01.152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/28/2015] [Indexed: 02/07/2023]
Abstract
The aim of the study was to evaluate the possible contribution of non-endothelial eNOS to the regulation of blood pressure (BP). To accomplish this, a double transgenic strain expressing eNOS exclusively in the vascular endothelium (eNOS-Tg/KO) has been generated by endothelial-specific targeting of bovine eNOS in eNOS-deficient mice (eNOS-KO). Expression of eNOS was evaluated in aorta, myocardium, kidney, brain stem and skeletal muscle. Organ bath studies revealed a complete normalization of aortic reactivity to acetylcholine, phenylephrine and the NO-donors in eNOS-Tg/KO. Function of eNOS in resistance arteries was demonstrated by acute i.v. infusion of acetylcholine and the NOS-inhibitor L-NAME. Acetylcholine decreased mean arterial pressure in all strains but eNOS-KO responded significantly less sensitive as compared eNOS-Tg/KO and C57BL/6. Likewise, acute i.v. L-NAME application elevated mean arterial pressure in C57BL/6 and eNOS-Tg/KO, but not in eNOS-KO. In striking contrast to these findings, mean, systolic and diastolic BP in eNOS-Tg/KO remained significantly elevated and was similar to values of eNOS-KO. Chronic oral treatment with L-NAME increased BP to the level of eNOS-KO only in C57BL/6, but had no effect on hypertension in eNOS-KO and eNOS-Tg/KO. Taken together, functional reconstitution of eNOS in the vasculature of eNOS-KO not even partially lowered BP. These data suggest that the activity of eNOS expressed in non-vascular tissue might play a role in physiologic BP regulation.
Collapse
Affiliation(s)
- Tatsiana Suvorava
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Manuel Thieme
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Stephanie Pick
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sebastian Friedrich
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Lars C Rump
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Thomas Hohlfeld
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Georg Kojda
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
43
|
Villela NR, dos Santos AOMT, de Miranda ML, Bouskela E. Fluid resuscitation therapy in endotoxemic hamsters improves survival and attenuates capillary perfusion deficits and inflammatory responses by a mechanism related to nitric oxide. J Transl Med 2014; 12:232. [PMID: 25151363 PMCID: PMC4158098 DOI: 10.1186/s12967-014-0232-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/15/2014] [Indexed: 12/21/2022] Open
Abstract
Background Relative hypovolemia is frequently found in early stages of severe sepsis and septic shock and prompt and aggressive fluid therapy has become standard of care improving tissue perfusion and patient outcome. This paper investigates the role of the nitric oxide pathway on beneficial microcirculatory effects of fluid resuscitation. Methods After skinfold chamber implantation procedures and endotoxemia induction by intravenous Escherichia coli lipopolysaccharide administration (2 mg.kg−1), male golden Syrian hamsters were fluid resuscitated and then sequentially treated with L-Nω-Nitroarginine and L-Arginine hydrochloride (LPS/FR/LNNA group). Intravital microscopy of skinfold chamber preparations allowed quantitative analysis of microvascular variables including venular leukocyte rolling and adhesion. Macro-hemodynamic, biochemical and hematological parameters as well as survival rate were also evaluated. Endotoxemic hamsters treated with fluid therapy alone (LPS/FR group) and non-treated animals (LPS group) served as controls. Results Fluid resuscitation was effective in reducing lipopolysaccharide-induced microcirculatory changes. After 3 hours of lipopolysaccharide administration, non-fluid resuscitated animals (LPS group) had the lowest functional capillary density (1% from baseline for LPS group vs. 19% for LPS/FR one; p <0.05). At the same time point, arteriolar mean internal diameter was significantly wider in LPS/FR group than in LPS one (100% vs. 50% from baseline). Fluid resuscitation also reduced leukocyte-endothelium interactions and sequestration (p <0.05 for LPS vs. LPS/FR group) and increased survival (median survival time: 2 and 5.5 days for LPS and LPS/FR groups, respectively; p <0.05). Nitric oxide synthase inhibition prevented these protective effects, while L-Arginine administration markedly restored many of them. Conclusion Our results suggest that the underlying mechanism of fluid therapy is the restoration of nitric oxide bioavailability, because inhibition of NOS prevented many of its beneficial effects. Nevertheless, further investigations are required in experimental models closer to conditions of human sepsis to confirm these results.
Collapse
Affiliation(s)
- Nivaldo Ribeiro Villela
- Department of Surgery, Division of Anesthesiology, Faculty of Medical Sciences, Rio de Janeiro State University, Boulevard 28 de Setembro, Rio de Janeiro, 77 - Vila Isabel, 20,551-030, RJ, Brazil.
| | | | | | | |
Collapse
|
44
|
Crosstalk between nitrite, myoglobin and reactive oxygen species to regulate vasodilation under hypoxia. PLoS One 2014; 9:e105951. [PMID: 25148388 PMCID: PMC4141839 DOI: 10.1371/journal.pone.0105951] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/28/2014] [Indexed: 11/19/2022] Open
Abstract
The systemic response to decreasing oxygen levels is hypoxic vasodilation. While this mechanism has been known for more than a century, the underlying cellular events have remained incompletely understood. Nitrite signaling is critically involved in vessel relaxation under hypoxia. This can be attributed to the presence of myoglobin in the vessel wall together with other potential nitrite reductases, which generate nitric oxide, one of the most potent vasodilatory signaling molecules. Questions remain relating to the precise concentration of nitrite and the exact dose-response relations between nitrite and myoglobin under hypoxia. It is furthermore unclear whether regulatory mechanisms exist which balance this interaction. Nitrite tissue levels were similar across all species investigated. We then investigated the exact fractional myoglobin desaturation in an ex vivo approach when gassing with 1% oxygen. Within a short time frame myoglobin desaturated to 58±12%. Given that myoglobin significantly contributes to nitrite reduction under hypoxia, dose-response experiments using physiological to pharmacological nitrite concentrations were conducted. Along all concentrations, abrogation of myoglobin in mice impaired vasodilation. As reactive oxygen species may counteract the vasodilatory response, we used superoxide dismutase and its mimic tempol as well as catalase and ebselen to reduce the levels of reactive oxygen species during hypoxic vasodilation. Incubation of tempol in conjunction with catalase alone and catalase/ebselen increased the vasodilatory response to nitrite. Our study shows that modest hypoxia leads to a significant nitrite-dependent vessel relaxation. This requires the presence of vascular myoglobin for both physiological and pharmacological nitrite levels. Reactive oxygen species, in turn, modulate this vasodilation response.
Collapse
|
45
|
Vecoli C, Novelli M, Pippa A, Giacopelli D, Beffy P, Masiello P, L’Abbate A, Neglia D. Partial deletion of eNOS gene causes hyperinsulinemic state, unbalance of cardiac insulin signaling pathways and coronary dysfunction independently of high fat diet. PLoS One 2014; 9:e104156. [PMID: 25093405 PMCID: PMC4122412 DOI: 10.1371/journal.pone.0104156] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/10/2014] [Indexed: 11/23/2022] Open
Abstract
Abnormalities in eNOS gene, possibly interacting with high fat diet (HFD), affect peripheral vascular function and glucose metabolism. The relative role of eNOS gene, HFD and metabolic derangement on coronary function has not been fully elucidated. We test whether eNOS gene deficiency per se or in association with HFD modulates coronary function through mechanisms involving molecular pathways related to insulin signaling. Wild type (WT), eNOS−/− and eNOS+/− mice were studied. WT and eNOS+/− mice were fed with either standard or HF diet for 16 weeks and compared with standard diet fed eNOS−/−. Glucose and insulin tolerance tests were performed during the last week of diet. Coronary resistance (CR) was measured at baseline and during infusions of acetylcholine (Ach) or sodium-nitroprusside (SNP) to evaluate endothelium-dependent or independent vasodilation, in the Langendorff isolated hearts. Cardiac expression of Akt and ERK genes as evaluation of two major insulin-regulated signaling pathways involved in the control of vascular tone were assessed by western blot. HFD-fed mice developed an overt diabetic state. Conversely, chow-fed genetically modified mice (in particular eNOS−/−) showed a metabolic pattern characterized by normoglycemia and hyperinsulinemia with a limited degree of insulin resistance. CR was significantly higher in animals with eNOS gene deletions than in WT, independently of diet. Percent decrease in CR, during Ach infusion, was significantly lower in both eNOS−/− and eNOS+/− mice than in WT, independently of diet. SNP reduced CR in all groups except eNOS−/−. The cardiac ERK1-2/Akt ratio, increased in animals with eNOS gene deletions compared with WT, independently of diet. These results suggest that the eNOS genetic deficiency, associated or not with HFD, has a relevant effect on coronary vascular function, possibly mediated by increase in blood insulin levels and unbalance in insulin-dependent signaling in coronary vessels, consistent with a shift towards a vasoconstrictive pattern.
Collapse
Affiliation(s)
| | - Michela Novelli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Anna Pippa
- Institute of Life Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
| | | | | | - Pellegrino Masiello
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Antonio L’Abbate
- Istituto di Fisiologia Clinica-CNR, Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Danilo Neglia
- Istituto di Fisiologia Clinica-CNR, Pisa, Italy
- Fondazione Toscana G. Monasterio-CNR, Pisa, Italy
- * E-mail:
| |
Collapse
|
46
|
Bernstein HG, Bannier J, Meyer-Lotz G, Steiner J, Keilhoff G, Dobrowolny H, Walter M, Bogerts B. Distribution of immunoreactive glutamine synthetase in the adult human and mouse brain. Qualitative and quantitative observations with special emphasis on extra-astroglial protein localization. J Chem Neuroanat 2014; 61-62:33-50. [PMID: 25058171 DOI: 10.1016/j.jchemneu.2014.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 12/20/2022]
Abstract
Glutamine synthetase catalyzes the ATP-dependent condensation of ammonia and glutamate to form glutamine, thus playing a pivotal role in glutamate and glutamine homoeostasis. Despite a plethora of studies on this enzyme, knowledge about the regional and cellular distribution of this enzyme in human brain is still fragmentary. Therefore, we mapped fourteen post-mortem brains of psychically healthy individuals for the distribution of the glutamine synthetase immunoreactive protein. It was found that glutamine synthetase immunoreactivity is expressed in multiple gray and white matter astrocytes, but also in oligodendrocytes, ependymal cells and certain neurons. Since a possible extra-astrocytic expression of glutamine synthetase is highly controversial, we paid special attention to its appearance in oligodendrocytes and neurons. By double immunolabeling of mouse brain slices and cultured mouse brain cells for glutamine synthetase and cell-type-specific markers we provide evidence that besides astrocytes subpopulations of oligodendrocytes, microglial cells and neurons express glutamine synthetase. Moreover, we show that glutamine synthetase-immunopositive neurons are not randomly distributed throughout human and mouse brain, but represent a subpopulation of nitrergic (i.e. neuronal nitric oxide synthase expressing) neurons. Possible functional implications of an extra-astrocytic localization of glutamine synthetase are discussed.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany.
| | - Jana Bannier
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany
| | - Gabriela Meyer-Lotz
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany
| | - Martin Walter
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany; Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Bernhard Bogerts
- Department of Psychiatry, Otto-von-Guericke University, Faculty of Medicine, Magdeburg, Germany
| |
Collapse
|
47
|
Role of nitric oxide synthase isoforms for ophthalmic artery reactivity in mice. Exp Eye Res 2014; 127:1-8. [PMID: 25017185 DOI: 10.1016/j.exer.2014.06.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 06/12/2014] [Accepted: 06/19/2014] [Indexed: 11/23/2022]
Abstract
Nitric oxide synthases (NOS) are involved in regulation of ocular vascular tone and blood flow. While endothelial NOS (eNOS) has recently been shown to mediate endothelium-dependent vasodilation in mouse retinal arterioles, the contribution of individual NOS isoforms to vascular responses is unknown in the retrobulbar vasculature. Moreover, it is unknown whether the lack of a single NOS isoform affects neuron survival in the retina. Thus, the goal of the present study was to examine the hypothesis that the lack of individual nitric oxide synthase (NOS) isoforms affects the reactivity of mouse ophthalmic arteries and neuron density in the retinal ganglion cell (RGC) layer. Mice deficient in one of the three NOS isoforms (nNOS-/-, iNOS-/- and eNOS-/-) were compared to respective wild type controls. Intraocular pressure (IOP) was measured in conscious mice using rebound tonometry. To examine the role of each NOS isoform for mediating vascular responses, ophthalmic arteries were studied in vitro using video microscopy. Neuron density in the RGC layer was calculated from retinal wholemounts stained with cresyl blue. IOP was similar in all NOS-deficient genotypes and respective wild type controls. In ophthalmic arteries, phenylephrine, nitroprusside and acetylcholine evoked concentration-dependent responses that did not differ between individual NOS-deficient genotypes and their respective controls. In all genotypes except eNOS-/- mice, vasodilation to acetylcholine was markedly reduced after incubation with L-NAME, a non-isoform-selective inhibitor of NOS. In contrast, pharmacological inhibition of nNOS and iNOS had no effect on acetylcholine-induced vasodilation in any of the mouse genotypes. Neuron density in the RGC layer was similar in all NOS-deficient genotypes and respective controls. Our findings suggest that eNOS contributes to endothelium-dependent dilation of murine ophthalmic arteries. However, the chronic lack of eNOS is functionally compensated by NOS-independent vasodilator mechanisms. The lack of a single NOS isoform does not appear to affect IOP or neuron density in the RGC layer.
Collapse
|
48
|
Sachse G, Faulhaber J, Seniuk A, Ehmke H, Pongs O. Smooth muscle BK channel activity influences blood pressure independent of vascular tone in mice. J Physiol 2014; 592:2563-74. [PMID: 24687584 DOI: 10.1113/jphysiol.2014.272880] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The large conductance voltage- and Ca(2+)-activated K(+) (BK) channel is an important determinant of vascular tone and contributes to blood pressure regulation. Both activities depend on the ancillary BKβ1 subunit. To determine the significance of smooth muscle BK channel activity for blood pressure regulation, we investigated the potential link between changes in arterial tone and altered blood pressure in BKβ1 knockout (BKβ1(-/-)) mice from three different genetically defined strains. While vascular tone was consistently increased in all BKβ1(-/-) mice independent of genetic background, BKβ1(-/-) strains exhibited increased (strain A), unaltered (strain B) or decreased (strain C) mean arterial blood pressures compared to their corresponding BKβ1(+/+) controls. In agreement with previous data on aldosterone regulation by renal/adrenal BK channel function, BKβ1(-/-) strain A mice have increased plasma aldosterone and increased blood pressure. Consistently, blockade of mineralocorticoid receptors by spironolactone treatment reversibly restored the elevated blood pressure to the BKβ1(+/+) strain A level. In contrast, loss of BKβ1 did not affect plasma aldosterone in strain C mice. Smooth muscle-restricted restoration of BKβ1 expression increased blood pressure in BKβ1(-/-) strain C mice, implying that impaired smooth muscle BK channel activity lowers blood pressure in these animals. We conclude that BK channel activity directly affects vascular tone but influences blood pressure independent of this effect via different pathways.
Collapse
Affiliation(s)
- Gregor Sachse
- Institut für Neurale Signalverarbeitung, Zentrum für Molekulare Neurobiologie, Falkenried 94, D-20251, Hamburg, Germany Department of Physiology, Anatomy and Genetics, South Parks Road, Oxford, OX1 3QX, UK
| | - Jörg Faulhaber
- Institut für Zelluläre und Integrative Physiologie, UKE, Martinistr. 52, D-20246, Hamburg, Germany
| | - Anika Seniuk
- Institut für Zelluläre und Integrative Physiologie, UKE, Martinistr. 52, D-20246, Hamburg, Germany
| | - Heimo Ehmke
- Institut für Zelluläre und Integrative Physiologie, UKE, Martinistr. 52, D-20246, Hamburg, Germany
| | - Olaf Pongs
- Institut für Neurale Signalverarbeitung, Zentrum für Molekulare Neurobiologie, Falkenried 94, D-20251, Hamburg, Germany Institut für Physiologie (Geb 56), Universität des Saarlandes, 66421, Homburg/Saar, Germany
| |
Collapse
|
49
|
Duncker DJ, van Deel ED, de Waard MC, de Boer M, Merkus D, van der Velden J. Exercise training in adverse cardiac remodeling. Pflugers Arch 2014; 466:1079-91. [PMID: 24573174 DOI: 10.1007/s00424-014-1464-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 12/14/2022]
Abstract
Cardiac remodeling in response to a myocardial infarction or chronic pressure-overload is an independent risk factor for the development of heart failure. In contrast, cardiac remodeling produced by regular physical exercise is associated with a decreased risk for heart failure. There is evidence that exercise training has a beneficial effect on disease progression and survival in patients with cardiac remodeling and dysfunction, but concern has also been expressed that exercise training may aggravate pathological remodeling and dysfunction. Here we present studies from our laboratory into the effects of exercise training on pathological cardiac remodeling and dysfunction in mice. The results indicate that even in the presence of a large infarct, exercise training exerts beneficial effects on the heart. These effects were mimicked in part by endothelial nitric oxide synthase (eNOS) overexpression and abrogated by eNOS deficiency, demonstrating the importance of nitric oxide signaling in mediating the cardiac effects of exercise. Exercise prior to a myocardial infarction was also cardioprotective. In contrast, exercise tended to aggravate pathological cardiac remodeling and dysfunction in the setting of pressure-overload produced by an aortic stenosis. These observations emphasize the critical importance of the underlying pathological stimulus for cardiac hypertrophy and remodeling, in determining the effects of exercise training. Future studies are needed to define the influence of exercise type, intensity and duration in different models and severities of pathological cardiac remodeling. Together such studies will aid in optimizing the therapy of exercise training in the setting of cardiovascular disease.
Collapse
Affiliation(s)
- Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands,
| | | | | | | | | | | |
Collapse
|
50
|
Sun D, Cuevas AJ, Gotlinger K, Hwang SH, Hammock BD, Schwartzman ML, Huang A. Soluble epoxide hydrolase-dependent regulation of myogenic response and blood pressure. Am J Physiol Heart Circ Physiol 2014; 306:H1146-53. [PMID: 24561863 DOI: 10.1152/ajpheart.00920.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid via cytochrome P450 (CYP)/epoxygenases. EETs possess cardioprotective properties and are catalyzed by soluble epoxide hydrolase (sEH) to dihydroxyeicosatrienoic acids (DHETs) that lack vasoactive property. To date, the role of sEH in the regulation of myogenic response of resistant arteries, a key player in the control of blood pressure, remains unknown. To this end, experiments were conducted on sEH-knockout (KO) mice, wild-type (WT) mice, and endothelial nitric oxide synthase (eNOS)-KO mice treated with t-TUCB, a sEH inhibitor, for 4 wk. sEH-KO and t-TUCB-treated mice displayed significantly lower blood pressure, associated with significantly increased vascular EETs and ratio of EETs/DHETs. Pressure-diameter relationships were assessed in isolated and cannulated gracilis muscle arterioles. All arterioles constricted in response to increases in transmural pressure from 60 to 140 mmHg. The myogenic constriction was significantly reduced, expressed as an upward shift of pressure-diameter curve, in arterioles of sEH-KO and t-TUCB-treated eNOS-KO mice compared with their controls. Removal of the endothelium, or treatment of the vessels with PPOH, an inhibitor of EET synthase, restored the attenuated pressure-induced constriction to the levels similar to those observed in their controls but had no effects on control vessels. No difference was observed in the myogenic index, or in the vascular expression of eNOS, CYP2C29 (EET synthase), and CYP4A (20-HETE synthase) among these groups of mice. In conclusion, the increased EET bioavailability, as a function of deficiency/inhibition of sEH, potentiates vasodilator responses that counteract pressure-induced vasoconstriction to lower blood pressure.
Collapse
Affiliation(s)
- Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York
| | | | | | | | | | | | | |
Collapse
|