1
|
Nelissen E, Schepers M, Ponsaerts L, Foulquier S, Bronckaers A, Vanmierlo T, Sandner P, Prickaerts J. Soluble guanylyl cyclase: A novel target for the treatment of vascular cognitive impairment? Pharmacol Res 2023; 197:106970. [PMID: 37884069 DOI: 10.1016/j.phrs.2023.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Vascular cognitive impairment (VCI) describes neurodegenerative disorders characterized by a vascular component. Pathologically, it involves decreased cerebral blood flow (CBF), white matter lesions, endothelial dysfunction, and blood-brain barrier (BBB) impairments. Molecularly, oxidative stress and inflammation are two of the major underlying mechanisms. Nitric oxide (NO) physiologically stimulates soluble guanylate cyclase (sGC) to induce cGMP production. However, under pathological conditions, NO seems to be at the basis of oxidative stress and inflammation, leading to a decrease in sGC activity and expression. The native form of sGC needs a ferrous heme group bound in order to be sensitive to NO (Fe(II)sGC). Oxidation of sGC leads to the conversion of ferrous to ferric heme (Fe(III)sGC) and even heme-loss (apo-sGC). Both Fe(III)sGC and apo-sGC are insensitive to NO, and the enzyme is therefore inactive. sGC activity can be enhanced either by targeting the NO-sensitive native sGC (Fe(II)sGC), or the inactive, oxidized sGC (Fe(III)sGC) and the heme-free apo-sGC. For this purpose, sGC stimulators acting on Fe(II)sGC and sGC activators acting on Fe(III)sGC/apo-sGC have been developed. These sGC agonists have shown their efficacy in cardiovascular diseases by restoring the physiological and protective functions of the NO-sGC-cGMP pathway, including the reduction of oxidative stress and inflammation, and improvement of vascular functioning. Yet, only very little research has been performed within the cerebrovascular system and VCI pathology when focusing on sGC modulation and its potential protective mechanisms on vascular and neural function. Therefore, within this review, the potential of sGC as a target for treating VCI is highlighted.
Collapse
Affiliation(s)
- Ellis Nelissen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands; Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Laura Ponsaerts
- Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium; Department of Cardio & Organ Systems (COS), Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience (MHeNS), School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Annelies Bronckaers
- Department of Cardio & Organ Systems (COS), Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands; Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113 Wuppertal, Germany; Hannover Medical School, 30625 Hannover, Germany
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
2
|
Fu J, Nisbett LM, Guo Y, Boon EM. NosP Detection of Heme Modulates Burkholderia thailandensis Biofilm Formation. Biochemistry 2023; 62:2426-2441. [PMID: 37498555 PMCID: PMC10478957 DOI: 10.1021/acs.biochem.3c00187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Aggregated bacteria embedded within self-secreted extracellular polymeric substances, or biofilms, are resistant to antibiotics and cause chronic infections. As such, they are a significant public health threat. Heme is an abundant iron source for pathogenic bacteria during infection; many bacteria have systems to detect heme assimilated from host cells, which is correlated with the transition between acute and chronic infection states. Here, we investigate the heme-sensing function of a newly discovered multifactorial sensory hemoprotein called NosP and its role in biofilm regulation in the soil-dwelling bacterium Burkholderia thailandensis, the close surrogate of Bio-Safety-Level-3 pathogen Burkholderia pseudomallei. The NosP family protein has previously been shown to exhibit both nitric oxide (NO)- and heme-sensing functions and to regulate biofilms through NosP-associated histidine kinases and two-component systems. Our in vitro studies suggest that BtNosP exhibits heme-binding kinetics and thermodynamics consistent with a labile heme-responsive protein and that the holo-form of BtNosP acts as an inhibitor of its associated histidine kinase BtNahK. Furthermore, our in vivo studies suggest that increasing the concentration of extracellular heme decreases B. thailandensis biofilm formation, and deletion of nosP and nahK abolishes this phenotype, consistent with a model that BtNosP detects heme and exerts an inhibitory effect on BtNahK to decrease the biofilm.
Collapse
Affiliation(s)
- Jiayuan Fu
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Lisa-Marie Nisbett
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Yulong Guo
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Elizabeth M Boon
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
3
|
Wittenborn EC, Thomas WC, Houghton KA, Wirachman ES, Wu Y, Marletta MA. Role of the Coiled-Coil Domain in Allosteric Activity Regulation in Soluble Guanylate Cyclase. Biochemistry 2023; 62:1568-1576. [PMID: 37129924 PMCID: PMC10686098 DOI: 10.1021/acs.biochem.3c00052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Soluble guanylate cyclase (sGC) is the primary nitric oxide (NO) receptor in higher eukaryotes, including humans. NO-dependent signaling via sGC is associated with important physiological effects in the vascular, pulmonary, and neurological systems, and sGC itself is an established drug target for the treatment of pulmonary hypertension due to its central role in vasodilation. Despite isolation in the late 1970s, high-resolution structural information on full-length sGC remained elusive until recent cryo-electron microscopy structures were determined of the protein in both the basal unactivated state and the NO-activated state. These structures revealed large-scale conformational changes upon activation that appear to be centered on rearrangements within the coiled-coil (CC) domains in the enzyme. Here, a structure-guided approach was used to engineer constitutively unactivated and constitutively activated sGC variants through mutagenesis of the CC domains. These results demonstrate that the activation-induced conformational change in the CC domains is necessary and sufficient for determining the level of sGC activity.
Collapse
Affiliation(s)
- Elizabeth C. Wittenborn
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - William C. Thomas
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kimberly A. Houghton
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Erika S. Wirachman
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yang Wu
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael A. Marletta
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
4
|
Dai Y, Stuehr DJ. BAY58-2667 Activates Different Soluble Guanylyl Cyclase Species by Distinct Mechanisms that Indicate Its Principal Target in Cells is the Heme-Free Soluble Guanylyl Cyclase-Heat Shock Protein 90 Complex. Mol Pharmacol 2023; 103:286-296. [PMID: 36868790 PMCID: PMC10166446 DOI: 10.1124/molpharm.122.000624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/14/2022] [Accepted: 02/07/2023] [Indexed: 03/05/2023] Open
Abstract
Nitric oxide (NO)-unresponsive forms of soluble guanylyl cyclase (sGC) exist naturally and in disease can disable NO-sGC-cGMP signaling. Agonists like BAY58-2667 (BAY58) target these sGC forms, but their mechanisms of action in living cells are unclear. We studied rat lung fibroblast-6 cells and human airway smooth muscle cells that naturally express sGC and HEK293 cells that we transfected to express sGC and variants. Cells were cultured to build up different forms of sGC, and we used fluorescence and FRET-based measures to monitor BAY58-driven cGMP production and any protein partner exchange or heme loss events that may occur for each sGC species. We found that: (i) BAY58 activated cGMP production by the apo-sGCβ-Hsp90 species after a 5-8 minute delay that was associated with apo-sGCβ exchanging its Hsp90 partner with an sGCα subunit. (ii) In cells containing an artificially constructed heme-free sGC heterodimer, BAY58 initiated an immediate and three times faster cGMP production. However, this behavior was not observed in cells expressing native sGC under any condition. (iii) BAY58 activated cGMP production by ferric heme sGC only after a 30-minute delay, coincident with it initiating a delayed, slow ferric heme loss from sGCβ We conclude that the kinetics favor BAY58 activation of the apo-sGCβ-Hsp90 species over the ferric heme sGC species in living cells. The protein partner exchange events driven by BAY58 account for the initial delay in cGMP production and also limit the speed of subsequent cGMP production in the cells. Our findings clarify how agonists like BAY58 may activate sGC in health and disease. SIGNIFICANCE STATEMENT: A class of agonists can activate cyclic guanosine monophosphate (cGMP) synthesis by forms of soluble guanylyl cyclase (sGC) that do not respond to NO and accumulate in disease, but the mechanisms of action are unclear. This study clarifies what forms of sGC exist in living cells, which of these can be activated by the agonists, and the mechanisms and kinetics by which each form is activated. This information may help to hasten deployment of these agonists for pharmaceutical intervention and clinical therapy.
Collapse
Affiliation(s)
- Yue Dai
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
5
|
Guo J, Yu X, Liu Y, Lu L, Zhu D, Zhang Y, Li L, Zhang P, Gao Q, Lu X, Sun M. Prenatal hypothyroidism diminished exogenous NO-mediated diastolic effects in fetal rat thoracic aorta smooth muscle via increased oxidative stress. Reprod Toxicol 2022; 113:52-61. [PMID: 35970333 DOI: 10.1016/j.reprotox.2022.08.009] [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: 05/12/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Maternal hypothyroidism is an important problem of modern healthcare and is reported to increase the risk of cardiovascular diseases in the offspring later in life. However, it is unclear whether hypothyroidism during pregnancy causes vascular damage in the fetal period. We established the prenatal hypothyroidism rat model and collected the fetuses at the 21th day of gestation (GD21). Thyroid hormone concentrations in maternal and offspring blood serum were assessed by enzyme-linked immunosorbent assay (ELISA). The thoracic aortas of the fetuses were isolated for microvessel functional testing and histochemical stainings. qPCR and Western blot were performed to access mRNA and protein expression. We found that the concentrations of thyroid hormones in the serum of pregnant rats and fetuses were significantly suppressed at GD21. The responses of the fetal thoracic aortas to SNP were significantly attenuated in the PTU group. However, no statistical difference was found between the two groups when treated with either inhibitor (ODQ) or activator (BAY58-2667) of sGC. The production of O2-• in the arterial wall was significantly increased in hypothyroid fetuses. Moreover, the level of NADPH oxidase (NOX) was increased, while superoxide dismutase 2 (SOD2) was down-regulated in the PTU group, ultimately contributing to the increased production of superoxide. Additionally, decreased SNP-mediated vasodilation found in fetal vessels was improved by either NOX inhibitor (Apocynin) or SOD mimic (Tempol). These results indicate that increased oxidative stress is probably the cause of the diminished diastolic effect of exogenous NO in the thoracic artery of prenatal hypothyroidism exposed fetuses.
Collapse
Affiliation(s)
- Jun Guo
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Yanping Liu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Likui Lu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Dan Zhu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Yingying Zhang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Pengjie Zhang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Xiyuan Lu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China.
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China.
| |
Collapse
|
6
|
Tawa M, Okamura T. Factors influencing the soluble guanylate cyclase heme redox state in blood vessels. Vascul Pharmacol 2022; 145:107023. [PMID: 35718342 DOI: 10.1016/j.vph.2022.107023] [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: 03/11/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 11/15/2022]
Abstract
Soluble guanylate cyclase (sGC) plays an important role in maintaining vascular homeostasis, as an acceptor for the biological messenger nitric oxide (NO). However, only reduced sGC (with a ferrous heme) can be activated by NO; oxidized (ferric heme) and apo (absent heme) sGC cannot. In addition, the proportions of reduced, oxidized, and apo sGC change under pathological conditions. Although diseased blood vessels often show decreased NO bioavailability in the vascular wall, a shift of sGC heme redox balance in favor of the oxidized/apo forms can also occur. Therefore, sGC is of growing interest as a drug target for various cardiovascular diseases. Notably, the balance between NO-sensitive reduced sGC and NO-insensitive oxidized/apo sGC in the body is regulated in a reversible manner by various biological molecules and proteins. Many studies have attempted to identify endogenous factors and determinants that influence this redox state. For example, various reactive nitrogen and oxygen species are capable of inducing the oxidation of sGC heme. Conversely, a heme reductase and some antioxidants reduce the ferric heme in sGC to the ferrous state. This review summarizes the factors and mechanisms identified by these studies that operate to regulate the sGC heme redox state.
Collapse
Affiliation(s)
- Masashi Tawa
- Department of Pathological and Molecular Pharmacology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-1094, Japan.
| | - Tomio Okamura
- Emeritus Professor, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
7
|
Jüttner AA, Danser AHJ, Roks AJM. Pharmacological developments in antihypertensive treatment through nitric oxide-cGMP modulation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:57-94. [PMID: 35659377 DOI: 10.1016/bs.apha.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Treatment of hypertension until now has been directed at inhibition of vasoconstriction, of cardiac contractility and of blood volume regulation. Despite the arsenal of drugs available for this purpose, the control of target blood pressure is still a difficult goal to reach in outpatients. The nitric oxide-cyclic guanosine monophosphate signaling is one of the most important mediators of vasodilation. It might therefore be a potential and most welcome drug target for optimization of the treatment of hypertension. In this chapter we review the problems that can occur in this signaling system, the attempts that have been made to correct these problems, and those that are still under investigation. Recently developed, clinically safe medicines that are currently approved for other applications, such as myocardial infarction, await to be tested for essential systemic hypertension. We conclude that despite many years of research without translation, stimulation of nitric oxide-cyclic guanosine monophosphate is still a viable strategy in the prevention of the health risk posed by chronic hypertension.
Collapse
Affiliation(s)
- Annika A Jüttner
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, The Netherlands
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
Durgin BG, Wood KC, Hahn SA, McMahon B, Baust JJ, Straub AC. Smooth muscle cell CYB5R3 preserves cardiac and vascular function under chronic hypoxic stress. J Mol Cell Cardiol 2022; 162:72-80. [PMID: 34536439 PMCID: PMC8766905 DOI: 10.1016/j.yjmcc.2021.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
Chronic hypoxia is a major driver of cardiovascular complications, including heart failure. The nitric oxide (NO) - soluble guanylyl cyclase (sGC) - cyclic guanosine monophosphate (cGMP) pathway is integral to vascular tone maintenance. Specifically, NO binds its receptor sGC within vascular smooth muscle cells (SMC) in its reduced heme (Fe2+) form to increase intracellular cGMP production, activate protein kinase G (PKG) signaling, and induce vessel relaxation. Under chronic hypoxia, oxidative stress drives oxidation of sGC heme (Fe2+→Fe3+), rendering it NO-insensitive. We previously showed that cytochrome b5 reductase 3 (CYB5R3) in SMC is a sGC reductase important for maintaining NO-dependent vasodilation and conferring resilience to systemic hypertension and sickle cell disease-associated pulmonary hypertension. To test whether CYB5R3 may be protective in the context of chronic hypoxia, we subjected SMC-specific CYB5R3 knockout mice (SMC CYB5R3 KO) to 3 weeks hypoxia and assessed vascular and cardiac function using echocardiography, pressure volume loops and wire myography. Hypoxic stress caused 1) biventricular hypertrophy in both WT and SMC CYB5R3 KO, but to a larger degree in KO mice, 2) blunted vasodilation to NO-dependent activation of sGC in coronary and pulmonary arteries of KO mice, and 3) decreased, albeit still normal, cardiac function in KO mice. Overall, these data indicate that SMC CYB5R3 deficiency potentiates bilateral ventricular hypertrophy and blunts NO-dependent vasodilation under chronic hypoxia conditions. This implicates that SMC CYB5R3 KO mice post 3-week hypoxia have early stages of cardiac remodeling and functional changes that could foretell significantly impaired cardiac function with longer exposure to hypoxia.
Collapse
Affiliation(s)
- Brittany G Durgin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Scott A Hahn
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Brenda McMahon
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jeffrey J Baust
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Microvascular Research, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
9
|
Liu R, Kang Y, Chen L. Activation mechanism of human soluble guanylate cyclase by stimulators and activators. Nat Commun 2021; 12:5492. [PMID: 34535643 PMCID: PMC8448884 DOI: 10.1038/s41467-021-25617-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/18/2021] [Indexed: 01/14/2023] Open
Abstract
Soluble guanylate cyclase (sGC) is the receptor for nitric oxide (NO) in human. It is an important validated drug target for cardiovascular diseases. sGC can be pharmacologically activated by stimulators and activators. However, the detailed structural mechanisms, through which sGC is recognized and positively modulated by these drugs at high spacial resolution, are poorly understood. Here, we present cryo-electron microscopy structures of human sGC in complex with NO and sGC stimulators, YC-1 and riociguat, and also in complex with the activator cinaciguat. These structures uncover the molecular details of how stimulators interact with residues from both β H-NOX and CC domains, to stabilize sGC in the extended active conformation. In contrast, cinaciguat occupies the haem pocket in the β H-NOX domain and sGC shows both inactive and active conformations. These structures suggest a converged mechanism of sGC activation by pharmacological compounds. Soluble guanylate cyclase (sGC) is a validated drug target for cardiovascular diseases. Here, the authors report structures of human sGC in complex with NO and sGC stimulators or activator, providing insight into the mechanism of sGC activation by pharmacological compounds.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Yunlu Kang
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China. .,Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China.
| |
Collapse
|
10
|
Maturation, inactivation, and recovery mechanisms of soluble guanylyl cyclase. J Biol Chem 2021; 296:100336. [PMID: 33508317 PMCID: PMC7949132 DOI: 10.1016/j.jbc.2021.100336] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 11/22/2022] Open
Abstract
Soluble guanylate cyclase (sGC) is a heme-containing heterodimeric enzyme that generates many molecules of cGMP in response to its ligand nitric oxide (NO); sGC thereby acts as an amplifier in NO-driven biological signaling cascades. Because sGC helps regulate the cardiovascular, neuronal, and gastrointestinal systems through its cGMP production, boosting sGC activity and preventing or reversing sGC inactivation are important therapeutic and pharmacologic goals. Work over the last two decades is uncovering the processes by which sGC matures to become functional, how sGC is inactivated, and how sGC is rescued from damage. A diverse group of small molecules and proteins have been implicated in these processes, including NO itself, reactive oxygen species, cellular heme, cell chaperone Hsp90, and various redox enzymes as well as pharmacologic sGC agonists. This review highlights their participation and provides an update on the processes that enable sGC maturation, drive its inactivation, or assist in its recovery in various settings within the cell, in hopes of reaching a better understanding of how sGC function is regulated in health and disease.
Collapse
|
11
|
Brescia M, Chao YC, Koschinski A, Tomek J, Zaccolo M. Multi-Compartment, Early Disruption of cGMP and cAMP Signalling in Cardiac Myocytes from the mdx Model of Duchenne Muscular Dystrophy. Int J Mol Sci 2020; 21:ijms21197056. [PMID: 32992747 PMCID: PMC7582831 DOI: 10.3390/ijms21197056] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most frequent and severe form of muscular dystrophy. The disease presents with progressive body-wide muscle deterioration and, with recent advances in respiratory care, cardiac involvement is an important cause of morbidity and mortality. DMD is caused by mutations in the dystrophin gene resulting in the absence of dystrophin and, consequently, disturbance of other proteins that form the dystrophin-associated protein complex (DAPC), including neuronal nitric oxide synthase (nNOS). The molecular mechanisms that link the absence of dystrophin with the alteration of cardiac function remain poorly understood but disruption of NO-cGMP signalling, mishandling of calcium and mitochondrial disturbances have been hypothesized to play a role. cGMP and cAMP are second messengers that are key in the regulation of cardiac myocyte function and disruption of cyclic nucleotide signalling leads to cardiomyopathy. cGMP and cAMP signals are compartmentalised and local regulation relies on the activity of phosphodiesterases (PDEs). Here, using genetically encoded FRET reporters targeted to distinct subcellular compartments of neonatal cardiac myocytes from the DMD mouse model mdx, we investigate whether lack of dystrophin disrupts local cyclic nucleotide signalling, thus potentially providing an early trigger for the development of cardiomyopathy. Our data show a significant alteration of both basal and stimulated cyclic nucleotide levels in all compartments investigated, as well as a complex reorganization of local PDE activities.
Collapse
|
12
|
Heckler I, Hossain S, Boon EM. Heme inhibits the activity of a c-di-GMP phosphodiesterase in Vibrio cholerae. Biochem Biophys Res Commun 2020; 529:1112-1116. [PMID: 32819573 PMCID: PMC8608024 DOI: 10.1016/j.bbrc.2020.06.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 06/11/2023]
Abstract
Heme, a complex of iron and protoporphyrin IX, plays an essential role in numerous biological processes including oxygen transport, oxygen storage, and electron transfer. The role of heme as a prosthetic group in bacterial hemoprotein gas sensors, which utilize heme as a cofactor for the binding of diatomic gas molecules, has been well studied. Less well known is the role of protein sensors of heme. In this report, we characterize the heme binding properties of a phosphodiesterase, CdpA, from Vibrio cholerae. We demonstrate that the N-terminal domain of CdpA is a NosP domain capable of heme binding, which consequently inhibits the c-di-GMP hydrolysis activity of the C-terminal phosphodiesterase domain. Further evidence for CdpA as a heme responsive sensor is supported by a relatively fast rate of heme dissociation. This study provides insight into an emerging class of heme-responsive sensor proteins.
Collapse
Affiliation(s)
- Ilana Heckler
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sajjad Hossain
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Elizabeth M Boon
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
13
|
Tyrosine 135 of the β1 subunit as binding site of BAY-543: Importance of the Y-x-S-x-R motif for binding and activation by sGC activator drugs. Eur J Pharmacol 2020; 881:173203. [DOI: 10.1016/j.ejphar.2020.173203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 11/22/2022]
|
14
|
Dai Y, Sweeny EA, Schlanger S, Ghosh A, Stuehr DJ. GAPDH delivers heme to soluble guanylyl cyclase. J Biol Chem 2020; 295:8145-8154. [PMID: 32358060 DOI: 10.1074/jbc.ra120.013802] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Soluble guanylyl cyclase (sGC) is a key component of NO-cGMP signaling in mammals. Although heme must bind in the sGC β1 subunit (sGCβ) for sGC to function, how heme is delivered to sGCβ remains unknown. Given that GAPDH displays properties of a heme chaperone for inducible NO synthase, here we investigated whether heme delivery to apo-sGCβ involves GAPDH. We utilized an sGCβ reporter construct, tetra-Cys sGCβ, whose heme insertion can be followed by fluorescence quenching in live cells, assessed how lowering cell GAPDH expression impacts heme delivery, and examined whether expressing WT GAPDH or a GAPDH variant defective in heme binding recovers heme delivery. We also studied interaction between GAPDH and sGCβ in cells and their complex formation and potential heme transfer using purified proteins. We found that heme delivery to apo-sGCβ correlates with cellular GAPDH expression levels and depends on the ability of GAPDH to bind intracellular heme, that apo-sGCβ associates with GAPDH in cells and dissociates when heme binds sGCβ, and that the purified GAPDH-heme complex binds to apo-sGCβ and transfers its heme to sGCβ. On the basis of these results, we propose a model where GAPDH obtains mitochondrial heme and then forms a complex with apo-sGCβ to accomplish heme delivery to sGCβ. Our findings illuminate a critical step in sGC maturation and uncover an additional mechanism that regulates its activity in health and disease.
Collapse
Affiliation(s)
- Yue Dai
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Elizabeth A Sweeny
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Simon Schlanger
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
15
|
Potoka KP, Wood KC, Baust JJ, Bueno M, Hahn SA, Vanderpool RR, Bachman T, Mallampalli GM, Osei-Hwedieh DO, Schrott V, Sun B, Bullock GC, Becker-Pelster EM, Wittwer M, Stampfuss J, Mathar I, Stasch JP, Truebel H, Sandner P, Mora AL, Straub AC, Gladwin MT. Nitric Oxide-Independent Soluble Guanylate Cyclase Activation Improves Vascular Function and Cardiac Remodeling in Sickle Cell Disease. Am J Respir Cell Mol Biol 2019; 58:636-647. [PMID: 29268036 DOI: 10.1165/rcmb.2017-0292oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sickle cell disease (SCD) is associated with intravascular hemolysis and oxidative inhibition of nitric oxide (NO) signaling. BAY 54-6544 is a small-molecule activator of oxidized soluble guanylate cyclase (sGC), which, unlike endogenous NO and the sGC stimulator, BAY 41-8543, preferentially binds and activates heme-free, NO-insensitive sGC to restore enzymatic cGMP production. We tested orally delivered sGC activator, BAY 54-6544 (17 mg/kg/d), sGC stimulator, BAY 41-8543, sildenafil, and placebo for 4-12 weeks in the Berkeley transgenic mouse model of SCD (BERK-SCD) and their hemizygous (Hemi) littermate controls (BERK-Hemi). Right ventricular (RV) maximum systolic pressure (RVmaxSP) was measured using micro right-heart catheterization. RV hypertrophy (RVH) was determined using Fulton's index and RV corrected weight (ratio of RV to tibia). Pulmonary artery vasoreactivity was tested for endothelium-dependent and -independent vessel relaxation. Right-heart catheterization revealed higher RVmaxSP and RVH in BERK-SCD versus BERK-Hemi, which worsened with age. Treatment with the sGC activator more effectively lowered RVmaxSP and RVH, with 90-day treatment delivering superior results, when compared with other treatments and placebo groups. In myography experiments, acetylcholine-induced (endothelium-dependent) and sodium-nitroprusside-induced (endothelium-independent NO donor) relaxation of the pulmonary artery harvested from placebo-treated BERK-SCD was impaired relative to BERK-Hemi but improved after therapy with sGC activator. By contrast, no significant effect for sGC stimulator or sildenafil was observed in BERK-SCD. These findings suggest that sGC is oxidized in the pulmonary arteries of transgenic SCD mice, leading to blunted responses to NO, and that the sGC activator, BAY 54-6544, may represent a novel therapy for SCD-associated pulmonary arterial hypertension and cardiac remodeling.
Collapse
Affiliation(s)
- Karin P Potoka
- 1 Division of Newborn Medicine, Department of Pediatrics.,2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Katherine C Wood
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,3 University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jeffrey J Baust
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Marta Bueno
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,4 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Scott A Hahn
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | | | - Tim Bachman
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Grace M Mallampalli
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,3 University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Valerie Schrott
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Bin Sun
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Grant C Bullock
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | | | | | | | | | | | - Hubert Truebel
- 5 Bayer AG, Wuppertal, Germany.,6 University of Witten/Herdecke, Witten, Germany
| | - Peter Sandner
- 5 Bayer AG, Wuppertal, Germany.,8 Hannover Medical School, Hannover, Germany; and
| | - Ana L Mora
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,4 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam C Straub
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,9 Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,4 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
A Randomized, Controlled Phase I/II Study to Evaluate the Safety and Efficacy of MGV354 for Ocular Hypertension or Glaucoma. Am J Ophthalmol 2018; 192:113-123. [PMID: 29802818 DOI: 10.1016/j.ajo.2018.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/10/2018] [Accepted: 05/16/2018] [Indexed: 11/22/2022]
Abstract
PURPOSE To assess the clinical safety, tolerability, and efficacy of topically administered MGV354, a soluble guanylate cyclase (sGC) activator, in patients with ocular hypertension (OH) or glaucoma. DESIGN Double-masked, randomized, and vehicle-controlled study. METHODS Parts 1 and 2 evaluated safety and tolerability to identify the maximum tolerated dose (MTD) of once-daily MGV354 in 32 healthy volunteers (Part 1) and 16 patients with OH or glaucoma (Part 2) at a single clinical site. Part 3 was a multisite trial that evaluated intraocular pressure (IOP)-lowering efficacy of the MTD administered nightly for 1 week in 50 patients with minimum IOP of 24 mm Hg at 8 AM, with a main outcome measure of mean diurnal IOP at day 8 compared to baseline (ClinicalTrials.govNCT02743780). RESULTS There was no difference in favor of MGV354 for IOP lowering; change from baseline to day 8 in mean diurnal IOP was -0.6 mm Hg for MGV354-treated patients and -1.1 mm Hg for vehicle-treated patients in Part 3, with a confidence interval of -0.7 to 1.7. The most common adverse events reported after MGV354 administration were conjunctival and ocular hyperemia. CONCLUSIONS Overall, MGV354 0.1% demonstrated no statistically significant effect compared to vehicle in lowering IOP based on the study's main outcome measure. MGV354 produced ocular hyperemia consistent with its pharmacology.
Collapse
|
17
|
Horst BG, Marletta MA. Physiological activation and deactivation of soluble guanylate cyclase. Nitric Oxide 2018; 77:65-74. [PMID: 29704567 PMCID: PMC6919197 DOI: 10.1016/j.niox.2018.04.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 01/24/2023]
Abstract
Soluble guanylate cyclase (sGC) is responsible for transducing the gaseous signaling molecule nitric oxide (NO) into the ubiquitous secondary signaling messenger cyclic guanosine monophosphate in eukaryotic organisms. sGC is exquisitely tuned to respond to low levels of NO, allowing cells to respond to non-toxic levels of NO. In this review, the structure of sGC is discussed in the context of sGC activation and deactivation. The sequence of events in the activation pathway are described into a comprehensive model of in vivo sGC activation as elucidated both from studies with purified enzyme and those done in cells. This model is then used to discuss the deactivation of sGC, as well as the molecular mechanisms of pathophysiological deactivation.
Collapse
Affiliation(s)
- Benjamin G Horst
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - Michael A Marletta
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
18
|
Kollau A, Opelt M, Wölkart G, Gorren ACF, Russwurm M, Koesling D, Mayer B, Schrammel A. Irreversible Activation and Stabilization of Soluble Guanylate Cyclase by the Protoporphyrin IX Mimetic Cinaciguat. Mol Pharmacol 2018; 93:73-78. [PMID: 29138269 PMCID: PMC5916872 DOI: 10.1124/mol.117.109918] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/18/2017] [Indexed: 02/02/2023] Open
Abstract
Belonging to the class of so-called soluble guanylate cyclase (sGC) activators, cinaciguat and BAY 60-2770 are interesting therapeutic tools for the treatment of various cardiovascular pathologies. The drugs are supposed to preferentially stimulate oxidized or heme-depleted, but not native sGC. Since this concept has been challenged by studies demonstrating complete relaxation of nondiseased vessels, this study was designed to reinvestigate the mode of action in greater detail. To this purpose, the effect of cinaciguat was studied on vessel tone of porcine coronary arteries and rat thoracic aortas. Organ bath studies showed that the compound caused time- and concentration-dependent relaxation of precontracted vessels with a maximal effect observed at 90 minutes. The dilatory response was not affected by extensive washout of the drug. Cinaciguat-induced vasodilation was associated with a time- and concentration-dependent increase of cGMP levels. Experiments with purified sGC in the presence of Tween 20 showed that cinaciguat activates the heme-free enzyme in a concentration-dependent manner with an EC50 value of ∼0.2 µM and maximal cGMP formation at 10 µM. By contrast, the effect of cinaciguat on 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one-oxidized (ferric) sGC was moderate, reaching ∼10%-15% of maximal activity. Dilution experiments of cinaciguat/Tween 20-preincubated sGC revealed the irreversible character of the drug. Assuming a sensitive balance between heme-free, ferric, and nitric oxide-sensitive ferrous sGC in cells and tissues, we propose that cinaciguat by virtue of its irreversible mode of action is capable of shifting this equilibrium toward the heme-free apo-sGC species.
Collapse
Affiliation(s)
- Alexander Kollau
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Marissa Opelt
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Gerald Wölkart
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Antonius C F Gorren
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Michael Russwurm
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Doris Koesling
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Bernd Mayer
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| | - Astrid Schrammel
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria (A.K., M.O., G.W., A.C.F.G., B.M., A.S.); and Department of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany (M.R., D.K.)
| |
Collapse
|
19
|
Pan J, Zhang X, Yuan H, Xu Q, Zhang H, Zhou Y, Huang ZX, Tan X. The molecular mechanism of heme loss from oxidized soluble guanylate cyclase induced by conformational change. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:488-500. [PMID: 26876536 DOI: 10.1016/j.bbapap.2016.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/05/2016] [Accepted: 02/10/2016] [Indexed: 11/25/2022]
Abstract
Heme oxidation and loss of soluble guanylate cyclase (sGC) is thought to be an important contributor to the development of cardiovascular diseases. Nevertheless, it remains unknown why the heme loses readily in oxidized sGC. In the current study, the conformational change of sGC upon heme oxidation by ODQ was studied based on the fluorescence resonance energy transfer (FRET) between the heme and a fluorophore fluorescein arsenical helix binder (FlAsH-EDT2) labeled at different domains of sGC β1. This study provides an opportunity to monitor the domain movement of sGC relative to the heme. The results indicated that heme oxidation by ODQ in truncated sCC induced the heme-associated αF helix moving away from the heme, the Per/Arnt/Sim domain (PAS) domain moving closer to the heme, but led the helical domain going further from the heme. We proposed that the synergistic effect of these conformational changes of the discrete region upon heme oxidation forces the heme pocket open, and subsequent heme loss readily. Furthermore, the kinetic studies suggested that the heme oxidation was a fast process and the conformational change was a relatively slow process. The kinetics of heme loss from oxidized sGC was monitored by a new method based on the heme group de-quenching the fluorescence of FlAsH-EDT2.
Collapse
Affiliation(s)
- Jie Pan
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China
| | - Xiaoxue Zhang
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China
| | - Hong Yuan
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China
| | - Qiming Xu
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Huijuan Zhang
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China
| | - Yajun Zhou
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China
| | - Zhong-Xian Huang
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China
| | - Xiangshi Tan
- Department of Chemistry & Shanghai Key laboratory of Chemical Biology for Protein Science, Fudan University, Shanghai 200433, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China.
| |
Collapse
|
20
|
Hoffmann LS, Kretschmer A, Lawrenz B, Hocher B, Stasch JP. Chronic Activation of Heme Free Guanylate Cyclase Leads to Renal Protection in Dahl Salt-Sensitive Rats. PLoS One 2015; 10:e0145048. [PMID: 26717150 PMCID: PMC4700984 DOI: 10.1371/journal.pone.0145048] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/29/2015] [Indexed: 12/31/2022] Open
Abstract
The nitric oxide (NO)/soluble guanylate cyclase (sGC)/cyclic guanosine monophasphate (cGMP)-signalling pathway is impaired under oxidative stress conditions due to oxidation and subsequent loss of the prosthetic sGC heme group as observed in particular in chronic renal failure. Thus, the pool of heme free sGC is increased under pathological conditions. sGC activators such as cinaciguat selectively activate the heme free form of sGC and target the disease associated enzyme. In this study, a therapeutic effect of long-term activation of heme free sGC by the sGC activator cinaciguat was investigated in an experimental model of salt-sensitive hypertension, a condition that is associated with increased oxidative stress, heme loss from sGC and development of chronic renal failure. For that purpose Dahl/ss rats, which develop severe hypertension upon high salt intake, were fed a high salt diet (8% NaCl) containing either placebo or cinaciguat for 21 weeks. Cinaciguat markedly improved survival and ameliorated the salt-induced increase in blood pressure upon treatment with cinaciguat compared to placebo. Renal function was significantly improved in the cinaciguat group compared to the placebo group as indicated by a significantly improved glomerular filtration rate and reduced urinary protein excretion. This was due to anti-fibrotic and anti-inflammatory effects of the cinaciguat treatment. Taken together, this is the first study showing that long-term activation of heme free sGC leads to renal protection in an experimental model of hypertension and chronic kidney disease. These results underline the promising potential of cinaciguat to treat renal diseases by targeting the disease associated heme free form of sGC.
Collapse
Affiliation(s)
- Linda S. Hoffmann
- Pharma Research Centre, Bayer HealthCare, Wuppertal, Germany
- * E-mail:
| | - Axel Kretschmer
- Pharma Research Centre, Bayer HealthCare, Wuppertal, Germany
| | - Bettina Lawrenz
- Pharma Research Centre, Bayer HealthCare, Wuppertal, Germany
| | - Berthold Hocher
- Instute of Nutritional Science, University of Potsdam, Potsdam, Germany, and IFLb Laboratoriumsmedizin Berlin GmbH, Berlin, Germany
| | - Johannes-Peter Stasch
- Pharma Research Centre, Bayer HealthCare, Wuppertal, Germany
- School of Pharmacy, Martin-Luther-University, Halle an der Saale, Germany
| |
Collapse
|
21
|
Raat NJ, Tabima DM, Specht PA, Tejero J, Champion HC, Kim-Shapiro DB, Baust J, Mik EG, Hildesheim M, Stasch JP, Becker EM, Truebel H, Gladwin MT. Direct sGC activation bypasses NO scavenging reactions of intravascular free oxy-hemoglobin and limits vasoconstriction. Antioxid Redox Signal 2013; 19:2232-43. [PMID: 23697678 PMCID: PMC3869448 DOI: 10.1089/ars.2013.5181] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 05/10/2013] [Accepted: 05/22/2013] [Indexed: 01/26/2023]
Abstract
AIMS Hemoglobin-based oxygen carriers (HBOC) provide a potential alternative to red blood cell (RBC) transfusion. Their clinical application has been limited by adverse effects, in large part thought to be mediated by the intravascular scavenging of the vasodilator nitric oxide (NO) by cell-free plasma oxy-hemoglobin. Free hemoglobin may also cause endothelial dysfunction and platelet activation in hemolytic diseases and after transfusion of aged stored RBCs. The new soluble guanylate cyclase (sGC) stimulator Bay 41-8543 and sGC activator Bay 60-2770 directly modulate sGC, independent of NO bioavailability, providing a potential therapeutic mechanism to bypass hemoglobin-mediated NO inactivation. RESULTS Infusions of human hemoglobin solutions and the HBOC Oxyglobin into rats produced a severe hypertensive response, even at low plasma heme concentrations approaching 10 μM. These reactions were only observed for ferrous oxy-hemoglobin and not analogs that do not rapidly scavenge NO. Infusions of L-NG-Nitroarginine methyl ester (L-NAME), a competitive NO synthase inhibitor, after hemoglobin infusion did not produce additive vasoconstriction, suggesting that vasoconstriction is related to scavenging of vascular NO. Open-chest hemodynamic studies confirmed that hypertension occurred secondary to direct effects on increasing vascular resistance, with limited negative cardiac inotropic effects. Intravascular hemoglobin reduced the vasodilatory potency of sodium nitroprusside (SNP) and sildenafil, but had no effect on vasodilatation by direct NO-independent activation of sGC by BAY 41-8543 and BAY 60-2770. INNOVATION AND CONCLUSION These data suggest that both sGC stimulators and sGC activators could be used to restore cyclic guanosine monophosphate-dependent vasodilation in conditions where cell-free plasma hemoglobin is sufficient to inhibit endogenous NO signaling.
Collapse
Affiliation(s)
- Nicolaas J.H. Raat
- Laboratory of Experimental Anesthesiology, Department of Anesthesiology, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D. Marcela Tabima
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patricia A.C. Specht
- Laboratory of Experimental Anesthesiology, Department of Anesthesiology, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jesús Tejero
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hunter C. Champion
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel B. Kim-Shapiro
- Department of Physics and the Translational Science Center, Wake Forest University, Winston-Salem, North Carolina
| | - Jeff Baust
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Egbert G. Mik
- Laboratory of Experimental Anesthesiology, Department of Anesthesiology, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mariana Hildesheim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Johannes-Peter Stasch
- Bayer Pharma AG, Wuppertal, Germany
- Institute of Pharmacy, Martin Luther University, Halle, Germany
| | - Eva-Maria Becker
- Bayer Pharma AG, Wuppertal, Germany
- Department of Human Medicine, University Witten/Herdecke, Witten, Germany
| | - Hubert Truebel
- Bayer Pharma AG, Wuppertal, Germany
- Department of Human Medicine, University Witten/Herdecke, Witten, Germany
| | - Mark T. Gladwin
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
22
|
Allerston CK, von Delft F, Gileadi O. Crystal structures of the catalytic domain of human soluble guanylate cyclase. PLoS One 2013; 8:e57644. [PMID: 23505436 PMCID: PMC3591389 DOI: 10.1371/journal.pone.0057644] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/22/2013] [Indexed: 01/05/2023] Open
Abstract
Soluble guanylate cyclase (sGC) catalyses the synthesis of cyclic GMP in response to nitric oxide. The enzyme is a heterodimer of homologous α and β subunits, each of which is composed of multiple domains. We present here crystal structures of a heterodimer of the catalytic domains of the α and β subunits, as well as an inactive homodimer of β subunits. This first structure of a metazoan, heteromeric cyclase provides several observations. First, the structures resemble known structures of adenylate cyclases and other guanylate cyclases in overall fold and in the arrangement of conserved active-site residues, which are contributed by both subunits at the interface. Second, the subunit interaction surface is promiscuous, allowing both homodimeric and heteromeric association; the preference of the full-length enzyme for heterodimer formation must derive from the combined contribution of other interaction interfaces. Third, the heterodimeric structure is in an inactive conformation, but can be superposed onto an active conformation of adenylate cyclase by a structural transition involving a 26° rigid-body rotation of the α subunit. In the modelled active conformation, most active site residues in the subunit interface are precisely aligned with those of adenylate cyclase. Finally, the modelled active conformation also reveals a cavity related to the active site by pseudo-symmetry. The pseudosymmetric site lacks key active site residues, but may bind allosteric regulators in a manner analogous to the binding of forskolin to adenylate cyclase. This indicates the possibility of developing a new class of small-molecule modulators of guanylate cyclase activity targeting the catalytic domain.
Collapse
Affiliation(s)
- Charles K. Allerston
- Structural Genomics Consortium, University of Oxford, Oxford, The United Kingdom
| | - Frank von Delft
- Structural Genomics Consortium, University of Oxford, Oxford, The United Kingdom
| | - Opher Gileadi
- Structural Genomics Consortium, University of Oxford, Oxford, The United Kingdom
- * E-mail:
| |
Collapse
|
23
|
|
24
|
Mendes-Silverio CB, Leiria LOS, Morganti RP, Anhê GF, Marcondes S, Mónica FZ, De Nucci G, Antunes E. Activation of haem-oxidized soluble guanylyl cyclase with BAY 60-2770 in human platelets lead to overstimulation of the cyclic GMP signaling pathway. PLoS One 2012; 7:e47223. [PMID: 23144808 PMCID: PMC3493568 DOI: 10.1371/journal.pone.0047223] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND AIMS Nitric oxide-independent soluble guanylyl cyclase (sGC) activators reactivate the haem-oxidized enzyme in vascular diseases. This study was undertaken to investigate the anti-platelet mechanisms of the haem-independent sGC activator BAY 60-2770 in human washed platelets. The hypothesis that sGC oxidation potentiates the anti-platelet activities of BAY 60-2770 has been tested. METHODS Human washed platelet aggregation and adhesion assays, as well as flow cytometry for α(IIb)β(3) integrin activation and Western blot for α1 and β1 sGC subunits were performed. Intracellular calcium levels were monitored in platelets loaded with a fluorogenic calcium-binding dye (FluoForte). RESULTS BAY 60-2770 (0.001-10 µM) produced significant inhibition of collagen (2 µg/ml)- and thrombin (0.1 U/ml)-induced platelet aggregation that was markedly potentiated by the sGC inhibitor ODQ (10 µM). In fibrinogen-coated plates, BAY 60-2770 significantly inhibited platelet adhesion, an effect potentiated by ODQ. BAY 60-2770 increased the cGMP levels and reduced the intracellular Ca(2+) levels, both of which were potentiated by ODQ. The cell-permeable cGMP analogue 8-Br-cGMP (100 µM) inhibited platelet aggregation and Ca(2+) levels in an ODQ-insensitive manner. The cAMP levels remained unchanged by BAY 60-2770. Collagen- and thrombin-induced α(IIb)β(3) activation was markedly inhibited by BAY 60-2770 that was further inhibited by ODQ. The effects of sodium nitroprusside (3 µM) were all prevented by ODQ. Incubation with ODQ (10 µM) significantly reduced the protein levels of α1 and β1 sGC subunits, which were prevented by BAY 60-2770. CONCLUSION The inhibitory effects of BAY 60-2770 on aggregation, adhesion, intracellular Ca(2+) levels and α(IIb)β(3) activation are all potentiated in haem-oxidizing conditions. BAY 60-2770 prevents ODQ-induced decrease in sGC protein levels. BAY 60-2770 could be of therapeutic interest in cardiovascular diseases associated with thrombotic complications.
Collapse
Affiliation(s)
- Camila B. Mendes-Silverio
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Luiz O. S. Leiria
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Rafael P. Morganti
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gabriel F. Anhê
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Sisi Marcondes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Fabíola Z. Mónica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- * E-mail:
| |
Collapse
|