1
|
Pino MTL, Rocca MV, Acosta LH, Cabilla JP. Challenging the Norm: The Unrecognized Impact of Soluble Guanylyl Cyclase Subunits in Cancer. Int J Mol Sci 2024; 25:10053. [PMID: 39337539 PMCID: PMC11432225 DOI: 10.3390/ijms251810053] [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: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Since the discovery of nitric oxide (NO), a long journey has led us to the present, during which much knowledge has been gained about its pathway members and their roles in physiological and various pathophysiological conditions. Soluble guanylyl cyclase (sGC), the main NO receptor composed of the sGCα1 and sGCβ1 subunits, has been one of the central figures in this narrative. However, the sGCα1 and sGCβ1 subunits remained obscured by the focus on sGC's enzymatic activity for many years. In this review, we restore the significance of the sGCα1 and sGCβ1 subunits by compiling and analyzing available but previously overlooked information regarding their roles beyond enzymatic activity. We delve into the basics of sGC expression regulation, from its transcriptional regulation to its interaction with proteins, placing particular emphasis on evidence thus far demonstrating the actions of each sGC subunit in different tumor models. Exploring the roles of sGC subunits in cancer offers a valuable opportunity to enhance our understanding of tumor biology and discover new therapeutic avenues.
Collapse
Affiliation(s)
- María Teresa L Pino
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| | - María Victoria Rocca
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| | - Lucas H Acosta
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| | - Jimena P Cabilla
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| |
Collapse
|
2
|
Liang W, Xie BK, Ding PW, Wang M, Yuan J, Cheng X, Liao YH, Yu M. Sacubitril/Valsartan Alleviates Experimental Autoimmune Myocarditis by Inhibiting Th17 Cell Differentiation Independently of the NLRP3 Inflammasome Pathway. Front Pharmacol 2021; 12:727838. [PMID: 34603042 PMCID: PMC8479108 DOI: 10.3389/fphar.2021.727838] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
Sacubitril/valsartan (Sac/Val) is a recently approved drug that is commonly used for treatment of heart failure. Several studies indicated that Sac/Val also regulated the secretion of inflammatory factors. However, the effect and mechanism of this drug modulation of inflammatory immune responses are uncertain. In this study, an experimental autoimmune myocarditis (EAM) mouse model was established by injection of α-myosin-heavy chain peptides. The effect of oral Sac/Val on EAM was evaluated by histological staining of heart tissues, measurements of cardiac troponin T and inflammatory markers (IL-6 and hsCRP). The effects of Sac/Val on NLRP3 inflammasome activation and Th1/Th17 cell differentiation were also determined. To further explore the signaling pathways, the expressions of cardiac soluble guanylyl cyclase (sGC) and NF-κB p65 were investigated. The results showed that Sac/Val downregulated the inflammatory response and attenuated the severity of EAM, but did not influence NLRP3 inflammasomes activation. Moreover, Sac/Val treatment inhibited cardiac Th17 cell differentiation, and this might be associated with sGC/NF-κB p65 signaling pathway. These findings indicate the potential use of Sac/Val for treatment of myocarditis.
Collapse
Affiliation(s)
- Wei Liang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bai-Kang Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Wu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Distinct Pharmacological Properties of Gaseous CO and CO-Releasing Molecule in Human Platelets. Int J Mol Sci 2021; 22:ijms22073584. [PMID: 33808315 PMCID: PMC8037872 DOI: 10.3390/ijms22073584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 01/26/2023] Open
Abstract
Carbon monoxide (CO)—gaseous or released by CO-RMs—both possess antiplatelet properties; however, it remains uncertain whether the mechanisms involved are the same. Here, we characterise the involvement of soluble guanylate cyclase (sGC) in the effects of CO—delivered by gaseous CO–saturated buffer (COG) and generated by CORM-A1—on platelet aggregation and energy metabolism, as well as on vasodilatation in aorta, using light transmission aggregometry, Seahorse XFe technique, and wire myography, respectively. ODQ completely prevented the inhibitory effect of COG on platelet aggregation, but did not modify antiplatelet effect of CORM-A1. In turn, COG did not affect, whereas CORM-A1 substantially inhibited energy metabolism in platelets. Even though activation of sGC by BAY 41-2272 or BAY 58-2667 inhibited significantly platelet aggregation, their effects on energy metabolism in platelets were absent or weak and could not contribute to antiplatelet effects of sGC activation. In contrast, vasodilatation of murine aortic rings, induced either by COG or CORM-A1, was dependent on sGC. We conclude that the source (COG vs. CORM-A1) and kinetics (rapid vs. slow) of CO delivery represent key determinants of the mechanism of antiplatelet action of CO, involving either impairment of energy metabolism or activation of sGG.
Collapse
|
4
|
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: 9.7] [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
|
5
|
Ma H, Fang C, Liu L, Wang Q, Aniwashi J, Sulaiman Y, Abudilaheman K, Liu W. Identification of novel genes associated with litter size of indigenous sheep population in Xinjiang, China using specific-locus amplified fragment sequencing technology. PeerJ 2019; 7:e8079. [PMID: 31788357 PMCID: PMC6883954 DOI: 10.7717/peerj.8079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/21/2019] [Indexed: 12/20/2022] Open
Abstract
Background There are abundant sheep breed resources in the Xinjiang region of China attributing to its diverse ecological system, which include several high-litter size sheep populations. Previous studies have confirmed that the major high prolificacy gene cannot be used to detect high litter size. Our research team found a resource group in Pishan County, southern Xinjiang. It showed high fertility with an average litter size of two to four in one birth, excellent breast development, and a high survival rate of lambs. In the present study, we used this resource as an ideal sample for studying the genetic mechanisms of high prolificacy in sheep. Methods Indigenous sheep populations from Xinjiang, with different litter sizes, were selected for the research, and specific-locus amplified fragment sequencing (SLAF-seq) technology was used to comprehensively screen single nucleotide polymorphisms (SNPs) from the whole genome that may cause differences in litter size. Novel genes associated with litter size of sheep were detected using genome-wide association studies (GWAS), providing new clues revealing the regulation mechanism of sheep fecundity. Candidate genes related to ovulation and litter size were selected for verification using Kompetitive Allele Specific polymerase chain reaction (KASP) cluster analysis. Results We identified 685,300 SNPs using the SLAF-seq technique for subsequent genome-wide analysis. Subsequently, 155 SNPs were detected at the genome-wide level. Fourteen genes related to sheep reproduction were notated: COIL, SLK, FSHR, Plxna3, Ddx24, CXCL12, Pla2g7, ATP5F1A, KERA, GUCY1A1, LOC101107541, LOC101107119, LOC101107809, and BRAF. Based on literature reports, 30 loci of seven genes and candidate genes (CXCL12, FSHR, SLK, GUCY1A1, COIL, LOC101107541, and LOC101107119) related to ovulation and litter size were selected for verification using KASP cluster analysis. Among them, nine loci of three genes were successfully genotyped. Three loci of FSHR (GenBank ID: 443299, g. 75320741G>A site), GUCY1A1 (GenBank ID: 101110000, g. 43266624C>T site), and COIL (GenBank ID: 101123134, g. 7321466C>G site) were found to be significantly or extremely significantly associated with litter size. These three loci are expected to be used as molecular markers to determine differences in litter size in sheep.
Collapse
Affiliation(s)
- Haiyu Ma
- College of Animal Science, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| | - Chao Fang
- Department of Veterinary Managment of Animal Resources, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Lingling Liu
- College of Animal Science, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| | - Qiong Wang
- College of Animal Science, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| | - Jueken Aniwashi
- College of Animal Science, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| | - Yiming Sulaiman
- College of Animal Science, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| | | | - Wujun Liu
- College of Animal Science, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| |
Collapse
|
6
|
Xu H, Yue C, Chen L. Post-Transcriptional Regulation of Soluble Guanylate Cyclase that Governs Neuropathic Pain in Alzheimer’s Disease1. J Alzheimers Dis 2019; 71:1331-1338. [PMID: 31524174 DOI: 10.3233/jad-190743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Heng Xu
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei province, Wuhan, China
| | - Chengjin Yue
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei province, Wuhan, China
| | - Lin Chen
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei province, Wuhan, China
| |
Collapse
|
7
|
Mohammadoo-Khorasani M, Karami Tehrani F, Atri M. Soluble guanylate cyclase isoenzymes: The expression of α1, α2, β1, and β2 subunits in the benign and malignant breast tumors. J Cell Physiol 2019; 235:1358-1365. [PMID: 31270804 DOI: 10.1002/jcp.29054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/13/2019] [Indexed: 12/18/2022]
Abstract
Soluble guanylate cyclase (sGC) encompasses α and β subunits. This study examined the expression of α1, α2, β1, and β2 subunits in the malignant and benign breast tumors using the Western blot analysis. Both benign and malignant tumors showed a significantly higher expression of the α1 subunit in comparison with normal tissues (p < 0.0001). In contrast, the expression of α2 and β2 sGC were significantly lower in these tumors than normal tissues (p < .0015 and p < .001, p < .007 and p < .0001, respectively). The expression level of α1 sGC was significantly correlated with ER + PR+ (p < .0001). A significant correlation was also detected for sGC-α1 and -α2 expression with c-erbB2-negative status (p < .01). However, the expression level of sGC was not associated with tumor stage, tumor grade, or other clinicopathological features. In conclusion, as the expression of α1 sGC is upregulated and α2 and β2 sGC are downregulated in malignant breast tumors. Variations in the expression of sGC isoenzymes may be suggested as an indicator to confirm the enzyme antitumor activity.
Collapse
Affiliation(s)
- Milad Mohammadoo-Khorasani
- Cancer Research Laboratory, Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Karami Tehrani
- Cancer Research Laboratory, Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Morteza Atri
- Department of Surgery, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Khalid RR, Siddiqi AR, Mylonas E, Maryam A, Kokkinidis M. Dynamic Characterization of the Human Heme Nitric Oxide/Oxygen (HNOX) Domain under the Influence of Diatomic Gaseous Ligands. Int J Mol Sci 2019; 20:E698. [PMID: 30736292 PMCID: PMC6387030 DOI: 10.3390/ijms20030698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 01/25/2023] Open
Abstract
Soluble guanylate cyclase (sGC) regulates numerous physiological processes. The β subunit Heme Nitric Oxide/Oxygen (HNOX) domain makes this protein sensitive to small gaseous ligands. The structural basis of the activation mechanism of sGC under the influence of ligands (NO, O₂, CO) is poorly understood. We examine the effect of different ligands on the human sGC HNOX domain. HNOX systems with gaseous ligands were generated and explored using Molecular Dynamics (MD). The distance between heme Fe2+ and histidine in the NO-ligated HNOX (NO-HNOX) system is larger compared to the O₂, CO systems. NO-HNOX rapidly adopts the conformation of the five-group metal coordination system. Loops α, β, γ and helix-f exhibit increased mobility and different hydrogen bond networks in NO-HNOX compared to the other systems. The removal of His from the Fe coordination sphere in NO-HNOX is assisted by interaction of the imidazole ring with the surrounding residues which in turn leads to the release of signaling helix-f and activation of the sGC enzyme. Insights into the conformational dynamics of a human sGC HNOX domain, especially for regions which are functionally critical for signal transduction, are valuable in the understanding of cardiovascular diseases.
Collapse
Affiliation(s)
- Rana Rehan Khalid
- Department of Biosciences, COMSATS University, Islamabad 45550, Pakistan.
- Department of Biology, University of Crete, 70013 Heraklion, Greece.
| | - Abdul Rauf Siddiqi
- Department of Biosciences, COMSATS University, Islamabad 45550, Pakistan.
| | - Efstratios Mylonas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), 70013 Heraklion, Greece.
| | - Arooma Maryam
- Department of Biosciences, COMSATS University, Islamabad 45550, Pakistan.
| | - Michael Kokkinidis
- Department of Biology, University of Crete, 70013 Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), 70013 Heraklion, Greece.
| |
Collapse
|
9
|
Bertollotto GM, de Oliveira MG, Alexandre EC, Calmasini FB, Passos GR, Antunes E, Mónica FZ. Inhibition of Multidrug Resistance Proteins by MK 571 Enhances Bladder, Prostate, and Urethra Relaxation through cAMP or cGMP Accumulation. J Pharmacol Exp Ther 2018; 367:138-146. [PMID: 30108158 DOI: 10.1124/jpet.118.250076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/03/2018] [Indexed: 12/28/2022] Open
Abstract
The biologic effect of cAMP and cGMP is terminated by phosphodiesterases and multidrug resistance proteins MRP4 and MRP5, which pump cyclic nucleotides out of the cell. Therefore, this study aimed to characterize the role of MRP inhibitor, MK 571 (3-[[[3-[(1E)-2-(7-chloro-2-quinolinyl)ethenyl]phenyl][[3-(dimethylamino)-3-oxopropyl]thio]methyl]thio]propanoic acid), in the bladder, prostate, and urethra of male mice by means of functional assays, protein expression, and cyclic nucleotide quantification. The cumulative addition of MK 571 (1-30 µM) produced only small relaxation responses (approximately 25%) in all studied tissues. In the bladder, isoprenaline/fenoterol and forskolin concentration-dependently relaxed and MK 571 (20 µM) increased the maximal response values by 37% and 24%, respectively. When MK 571 was coincubated with fenoterol or forskolin, intracellular levels of cAMP and protein expression of phospho-vasodilator-stimulated phosphoprotein (p-VASP) Ser157 were significantly greater compared with bladders stimulated with fenoterol or forskolin alone. In the prostate and urethra, sodium nitroprusside concentration-dependently relaxed and MK 571 (20 µM) significantly increased relaxation responses by 70% and 56%, respectively, accompanied by greater intracellular levels of cGMP and protein expression of p-VASP Ser239 in the prostate. Tadalafil and BAY 41-2272 (5-cyclopropyl-2-[1-[(2-fluorophenyl)methyl]-1H-pyrazolo[3,4-b]pyridin-3-yl]-4-pyrimidinamine) also relaxed the prostate and urethra, respectively, and MK 571 markedly enhanced this response. The stable analog of cGMP (8-Br-cGMP) induced concentration-dependent relaxation responses in the prostate and urethra, and MK 571 significantly increased the relaxation response. In conclusion, to our knowledge, this is the first study to show that efflux transporters are physiologically active in the bladder, prostate, and urethra to control intracellular levels of cAMP or cGMP.
Collapse
Affiliation(s)
- Gabriela Maria Bertollotto
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | - Eduardo Costa Alexandre
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Fabiano Beraldi Calmasini
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Gabriela Reolon Passos
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Fabiola Zakia Mónica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
10
|
Peters S, Paolillo M, Mergia E, Koesling D, Kennel L, Schmidtko A, Russwurm M, Feil R. cGMP Imaging in Brain Slices Reveals Brain Region-Specific Activity of NO-Sensitive Guanylyl Cyclases (NO-GCs) and NO-GC Stimulators. Int J Mol Sci 2018; 19:ijms19082313. [PMID: 30087260 PMCID: PMC6122017 DOI: 10.3390/ijms19082313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/01/2018] [Accepted: 08/04/2018] [Indexed: 11/23/2022] Open
Abstract
Impaired NO-cGMP signaling has been linked to several neurological disorders. NO-sensitive guanylyl cyclase (NO-GC), of which two isoforms—NO-GC1 and NO-GC2—are known, represents a promising drug target to increase cGMP in the brain. Drug-like small molecules have been discovered that work synergistically with NO to stimulate NO-GC activity. However, the effects of NO-GC stimulators in the brain are not well understood. In the present study, we used Förster/fluorescence resonance energy transfer (FRET)-based real-time imaging of cGMP in acute brain slices and primary neurons of cGMP sensor mice to comparatively assess the activity of two structurally different NO-GC stimulators, IWP-051 and BAY 41-2272, in the cerebellum, striatum and hippocampus. BAY 41-2272 potentiated an elevation of cGMP induced by the NO donor DEA/NO in all tested brain regions. Interestingly, IWP-051 potentiated DEA/NO-induced cGMP increases in the cerebellum and striatum, but not in the hippocampal CA1 area or primary hippocampal neurons. The brain-region-selective activity of IWP-051 suggested that it might act in a NO-GC isoform-selective manner. Results of mRNA in situ hybridization indicated that the cerebellum and striatum express NO-GC1 and NO-GC2, while the hippocampal CA1 area expresses mainly NO-GC2. IWP-051-potentiated DEA/NO-induced cGMP signals in the striatum of NO-GC2 knockout mice but was ineffective in the striatum of NO-GC1 knockout mice. These results indicate that IWP-051 preferentially stimulates NO-GC1 signaling in brain slices. Interestingly, no evidence for an isoform-specific effect of IWP-051 was observed when the cGMP-forming activity of whole brain homogenates was measured. This apparent discrepancy suggests that the method and conditions of cGMP measurement can influence results with NO-GC stimulators. Nevertheless, it is clear that NO-GC stimulators enhance cGMP signaling in the brain and should be further developed for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Stefanie Peters
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany.
| | - Michael Paolillo
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany.
| | - Evanthia Mergia
- Institut für Pharmakologie und Toxikologie, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| | - Doris Koesling
- Institut für Pharmakologie und Toxikologie, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| | - Lea Kennel
- Pharmakologisches Institut für Naturwissenschaftler, University of Frankfurt, 60438 Frankfurt am Main, Germany.
| | - Achim Schmidtko
- Pharmakologisches Institut für Naturwissenschaftler, University of Frankfurt, 60438 Frankfurt am Main, Germany.
| | - Michael Russwurm
- Institut für Pharmakologie und Toxikologie, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
11
|
Real-Time Imaging Reveals Augmentation of Glutamate-Induced Ca 2+ Transients by the NO-cGMP Pathway in Cerebellar Granule Neurons. Int J Mol Sci 2018; 19:ijms19082185. [PMID: 30049956 PMCID: PMC6121606 DOI: 10.3390/ijms19082185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 01/11/2023] Open
Abstract
Dysfunctions of NO-cGMP signaling have been implicated in various neurological disorders. We have studied the potential crosstalk of cGMP and Ca2+ signaling in cerebellar granule neurons (CGNs) by simultaneous real-time imaging of these second messengers in living cells. The NO donor DEA/NO evoked cGMP signals in the granule cell layer of acute cerebellar slices from transgenic mice expressing a cGMP sensor protein. cGMP and Ca2+ dynamics were visualized in individual CGNs in primary cultures prepared from 7-day-old cGMP sensor mice. DEA/NO increased the intracellular cGMP concentration and augmented glutamate-induced Ca2+ transients. These effects of DEA/NO were absent in CGNs isolated from knockout mice lacking NO-sensitive guanylyl cyclase. Furthermore, application of the cGMP analogues 8-Br-cGMP and 8-pCPT-cGMP, which activate cGMP effector proteins such as cyclic nucleotide-gated cation channels and cGMP-dependent protein kinases (cGKs), also potentiated glutamate-induced Ca2+ transients. Western blot analysis failed to detect cGK type I or II in our primary CGNs. The addition of phosphodiesterase (PDE) inhibitors during cGMP imaging showed that CGNs degrade cGMP mainly via Zaprinast-sensitive PDEs, most likely PDE5 and/or PDE10, but not via PDE1, 2, or 3. In sum, these data delineate a cGK-independent NO-cGMP signaling cascade that increases glutamate-induced Ca2+ signaling in CGNs. This cGMP–Ca2+ crosstalk likely affects neurotransmitter-stimulated functions of CGNs.
Collapse
|
12
|
Li S, Lowell AN, Newmister SA, Yu F, Williams RM, Sherman DH. Decoding cyclase-dependent assembly of hapalindole and fischerindole alkaloids. Nat Chem Biol 2017; 13:467-469. [PMID: 28288107 PMCID: PMC5391265 DOI: 10.1038/nchembio.2327] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 12/22/2016] [Indexed: 11/25/2022]
Abstract
The formation of C-C bonds in an enantioselective fashion to create complex polycyclic scaffolds in the hapalindole- and fischerindole- type alkaloids from Stigonematales cyanobacteria represents a compelling and urgent challenge in adapting microbial biosynthesis as a catalytic platform in drug development. Here we determine the biochemical basis for tri- and tetracyclic core formation in these secondary metabolites, involving a new class of cyclases that catalyze a complex cyclization cascade.
Collapse
Affiliation(s)
- Shasha Li
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew N Lowell
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Sean A Newmister
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Fengan Yu
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert M Williams
- Department of Chemistry, Colorado State University, Fort Collins, Colorado, USA
- University of Colorado Cancer Center, Aurora, Colorado, USA
| | - David H Sherman
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology &Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Van Rymenant E, Van Camp J, Pauwels B, Boydens C, Vanden Daele L, Beerens K, Brouckaert P, Smagghe G, Kerimi A, Williamson G, Grootaert C, Van de Voorde J. Ferulic acid-4-O-sulfate rather than ferulic acid relaxes arteries and lowers blood pressure in mice. J Nutr Biochem 2017; 44:44-51. [PMID: 28391055 DOI: 10.1016/j.jnutbio.2017.02.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/23/2017] [Accepted: 02/17/2017] [Indexed: 02/03/2023]
Abstract
Consumption of foods rich in ferulic acid (FA) such as wholegrain cereals, or FA precursors such as chlorogenic acids in coffee, is inversely correlated with risk of cardiovascular disease and type 2 diabetes. As a result of digestion and phase II metabolism in the gut and liver, FA is converted predominantly into ferulic acid-4-O-sulfate (FA-sul), an abundant plasma metabolite. Although FA-sul is the main metabolite, very little has been reported regarding its bioactivities. We have compared the ex vivo vasorelaxing effect of FA and FA-sul (10-7-3.10-5M) on isolated mouse arteries mounted in tissue myographs. FA-sul, but not FA, elicited a concentration-dependent vasorelaxation of saphenous and femoral arteries and aortae. The FA-sul-mediated vasorelaxation was blunted by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a soluble guanylate cyclase (sGC) inhibitor. The role of sGC was confirmed in femoral arteries isolated from sGCα1(-/-) knockout mice. Furthermore, 4-aminopyridine, a specific inhibitor of voltage-dependent potassium channels, significantly decreased FA-sul-mediated effects. In anesthetized mice, intravenous injection of FA-sul decreased mean arterial pressure, whereas FA had no effect, confirming the results obtained ex vivo. FA-sul is probably one of the major metabolites accounting for the blood pressure-lowering effects associated with FA consumption.
Collapse
Affiliation(s)
- Evelien Van Rymenant
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Gent, Belgium.
| | - John Van Camp
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Gent, Belgium.
| | - Bart Pauwels
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, 9000 Gent, Belgium.
| | - Charlotte Boydens
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, 9000 Gent, Belgium.
| | - Laura Vanden Daele
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, 9000 Gent, Belgium.
| | - Katrijn Beerens
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Gent, Belgium
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University-VIB, Technologiepark 927, 9052 Gent, Belgium.
| | - Guy Smagghe
- Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Gent, Belgium.
| | - Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| | - Charlotte Grootaert
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Gent, Belgium.
| | - Johan Van de Voorde
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, 9000 Gent, Belgium.
| |
Collapse
|
14
|
Probing the Molecular Mechanism of Human Soluble Guanylate Cyclase Activation by NO in vitro and in vivo. Sci Rep 2017; 7:43112. [PMID: 28230071 PMCID: PMC5322342 DOI: 10.1038/srep43112] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/19/2017] [Indexed: 11/12/2022] Open
Abstract
Soluble guanylate cyclase (sGC) is a heme-containing metalloprotein in NO-sGC-cGMP signaling. NO binds to the heme of sGC to catalyze the synthesis of the second messenger cGMP, which plays a critical role in several physiological processes. However, the molecular mechanism for sGC to mediate the NO signaling remains unclear. Here fluorophore FlAsH-EDT2 and fluorescent proteins were employed to study the NO-induced sGC activation. FlAsH-EDT2 labeling study revealed that NO binding to the H-NOX domain of sGC increased the distance between H-NOX and PAS domain and the separation between H-NOX and coiled-coil domain. The heme pocket conformation changed from “closed” to “open” upon NO binding. In addition, the NO-induced conformational change of sGC was firstly investigated in vivo through fluorescence lifetime imaging microscopy. The results both in vitro and in vivo indicated the conformational change of the catalytic domain of sGC from “open” to “closed” upon NO binding. NO binding to the heme of H-NOX domain caused breaking of Fe-N coordination bond, initiated the domain moving and conformational change, induced the allosteric effect of sGC to trigger the NO-signaling from H-NOX via PAS & coiled-coil to the catalytic domain, and ultimately stimulates the cyclase activity of sGC.
Collapse
|
15
|
Alexandropoulos II, Argyriou AI, Marousis KD, Topouzis S, Papapetropoulos A, Spyroulias GA. (1)H, (13)C, (15)N backbone and side-chain resonance assignment of Nostoc sp. C139A variant of the heme-nitric oxide/oxygen binding (H-NOX) domain. BIOMOLECULAR NMR ASSIGNMENTS 2016; 10:395-400. [PMID: 27614467 DOI: 10.1007/s12104-016-9707-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/01/2016] [Indexed: 06/06/2023]
Abstract
The H-NOX (Heme-nitric oxide/oxygen binding) domain is conserved across eukaryotes and bacteria. In human soluble guanylyl cyclase (sGC) the H-NOX domain functions as a sensor for the gaseous signaling agent nitric oxide (NO). sGC contains the heme-binding H-NOX domain at its N-terminus, which regulates the catalytic site contained within the C-terminal end of the enzyme catalyzing the conversion of GTP (guanosine 5'-triphosphate) to GMP (guanylyl monophosphate). Here, we present the backbone and side-chain assignments of the (1)H, (13)C and (15)N resonances of the 183-residue H-NOX domain from Nostoc sp. through solution NMR.
Collapse
Affiliation(s)
| | | | | | - Stavros Topouzis
- Department of Pharmacy, Laboratory of Molecular Pharmacology, University of Patras, 26504, Patras, Greece
| | - Andreas Papapetropoulos
- Department of Pharmacy, Laboratory of Molecular Pharmacology, University of Patras, 26504, Patras, Greece
- Faculty of Pharmacy, National and Kapodistrian University of Athens, 15 771, Athens, Greece
| | | |
Collapse
|
16
|
El-Sehemy A, Postovit LM, Fu Y. Nitric oxide signaling in human ovarian cancer: A potential therapeutic target. Nitric Oxide 2016; 54:30-7. [DOI: 10.1016/j.niox.2016.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 02/05/2016] [Accepted: 02/11/2016] [Indexed: 12/27/2022]
|
17
|
Feiteiro J, Verde I, Cairrão E. Cyclic guanosine monophosphate compartmentation in human vascular smooth muscle cells. Cell Signal 2015; 28:109-116. [PMID: 26689737 DOI: 10.1016/j.cellsig.2015.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 01/12/2023]
Abstract
AIMS The role of different vascular subtypes of phosphodiesterases (PDE) in cGMP compartmentalization was evaluated in human smooth muscle cells. METHODS AND RESULTS To understand how the cGMP conveys different information we infected smooth muscle cells with adenovirus containing mutants of the rat olfactory cyclic nucleotide-gated (CNG) channel-subunit and we recorded the associated cGMP-gated current (ICNG). The whole cell configuration of patch clamp technique was used to measure the ICNG and also the potassium current (IK) in human umbilical artery smooth muscle cells (HUASMC). ANP (0.1μM) induced a clear activation of basal ICNG, whereas SNP (100 μM) had a slight effect. The nonselective PDE inhibitor (IBMX; 100 μM), the PDE5 inhibitor (T0-156; 1 μM) and the PDE3 inhibitor (cilostamide; 10 μM), all had a tiny effects on the basal ICNG current. Concerning potassium channels, we observed that ANP and testosterone induced activation of IK and this activation is bigger than that elicited by SNP, cilostamide and T0-156. Cilostamide and T0-156 decreased the CNG stimulation induced by ANP and testosterone, suggesting that pGC pool is controlled by PDE3 and 5. Thus, the effects of SNP show the existence of two separated pools, one localized next to the plasma membrane and controlled by the PDE5 and PDE3, and a second pool localized in the cytosol of the cells that is regulated mainly by PDE3. CONCLUSIONS Our results show the existence of cGMP compartmentalization in human vascular smooth muscle cells and this phenomenon can open new perspectives concerning the examination of PDE families as therapeutic targets.
Collapse
Affiliation(s)
- Joana Feiteiro
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, Covilhã, Portugal
| | - Ignacio Verde
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, Covilhã, Portugal
| | - Elisa Cairrão
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
18
|
Smoleń S, Walaszek DJ, Karczewski M, Martin E, Gryko D. Towards NO-free Regulation of sGC: Design and Synthesis oftrans-AB-porphyrins. Isr J Chem 2015. [DOI: 10.1002/ijch.201500019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
19
|
Pauwels B, Boydens C, Brouckaert P, Van de Voorde J. Oximes induce erection and are resistant to oxidative stress. J Sex Med 2015; 12:906-15. [PMID: 25689429 DOI: 10.1111/jsm.12846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Because of their nitric oxide (NO)-donating capacities, oxime derivatives have shown to offer some therapeutic perspective for the treatment of erectile dysfunction (ED) as well as cardiovascular diseases. However, to date the in vivo effect of these oximes on erectile function remains unknown. In many disease states oxidative stress occurs, impairing NO-mediated relaxations. Hence the influence of oxidative stress on oxime-induced effects is also of interest. AIMS This study aimed to evaluate the in vivo effect of formaldoxime (FAL) and formamidoxime (FAM) on blood pressure and intracavernosal pressure (ICP); and to examine the role of soluble guanylyl cyclase (sGC) and the influence of oxidative stress on the FAL and FAM responses. METHODS Blood pressure and ICP were monitored in vivo after resp. intravenous or intracavernosal injection of FAL and FAM. Moreover isometric tension was measured in vitro on isolated mice corpora cavernosa (CC), thoracic aorta, and femoral artery in organ baths. The role of sGC was investigated using transgenic mice lacking the alpha 1 subunit of sGC. MAIN OUTCOME MEASURES Mean arterial pressure (MAP) and ICP were measured after FAL/FAM injection. In vitro relaxation of CC strips was evaluated in response to addition of FAL/FAM. RESULTS In vivo both FAL and FAM elicit a dose-dependent lowering of blood pressure (maximal ΔMAP: 33.66 ± 4.07 mm Hg [FAL] and 20.43 ± 2.06 mm Hg [FAM] ) as well as an increase of ICP (maximal increase of ICP/MAP: 70.29 ± 2.88% [FAL] and 52.91 ± 8.61% [FAM] ). The FAL/FAM effect is significantly lower in knockout vs. wild-type mice. Oxidative stress has an inhibitory effect on corporal NO-mediated relaxations induced by electrical field stimulation, acetylcholine, and sodium nitroprusside whereas the responses to 8-(4-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate sodium salt, FAL and FAM were not influenced. CONCLUSIONS Oximes induce erection which is mediated by sGC. The oxime-induced relaxations are resistant to oxidative stress, which increases their therapeutic potential for the treatment of ED.
Collapse
Affiliation(s)
- Bart Pauwels
- Department of Pharmacology, Ghent University, Ghent, Belgium
| | | | | | | |
Collapse
|
20
|
Rajagopal S, Nalli AD, Kumar DP, Bhattacharya S, Hu W, Mahavadi S, Grider JR, Murthy KS. Cytokine-induced S-nitrosylation of soluble guanylyl cyclase and expression of phosphodiesterase 1A contribute to dysfunction of longitudinal smooth muscle relaxation. J Pharmacol Exp Ther 2014; 352:509-18. [PMID: 25550199 DOI: 10.1124/jpet.114.221929] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The effect of proinflammatory cytokines on the expression and activity of soluble guanylyl cyclase (sGC) and cGMP-phosphodiesterases (PDEs) was determined in intestinal longitudinal smooth muscle. In control muscle cells, cGMP levels are regulated via activation of sGC and PDE5; the activity of the latter is regulated via feedback phosphorylation by cGMP-dependent protein kinase. In muscle cells isolated from muscle strips cultured with interleukin-1β (IL-1β) or tumor necrosis factor α (TNF-α) or obtained from the colon of TNBS (2,4,6-trinitrobenzene sulfonic acid)-treated mice, expression of inducible nitric oxide synthase (iNOS) was induced and sGC was S-nitrosylated, resulting in attenuation of nitric oxide (NO)-induced sGC activity and cGMP formation. The effect of cytokines on sGC S-nitrosylation and activity was blocked by the iNOS inhibitor 1400W [N-([3-(aminomethyl)phenyl]methyl)ethanimidamide dihydrochloride]. The effect of cytokines on cGMP levels measured in the absence of IBMX (3-isobutyl-1-methylxanthine), however, was partly reversed by 1400W or PDE1 inhibitor vinpocetine and completely reversed by a combination of 1400W and vinpocetine. Expression of PDE1A was induced and was accompanied by an increase in PDE1A activity in muscle cells isolated from muscle strips cultured with IL-1β or TNF-α or obtained from the colon of TNBS-treated mice; the effect of cytokines on PDE1 expression and activity was blocked by MG132 (benzyl N-[(2S)-4-methyl-1-[[(2S)-4-methyl-1-[[(2S)-4-methyl-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]carbamate), an inhibitor of nuclear factor κB activity. NO-induced muscle relaxation was inhibited in longitudinal muscle cells isolated from muscle strips cultured with IL-1β or TNF-α or obtained from the colon of TNBS-treated mice, and this inhibition was completely reversed by the combination of both 1400W and vinpocetine. Inhibition of smooth muscle relaxation during inflammation reflects the combined effects of decreased sGC activity via S-nitrosylation and increased cGMP hydrolysis via PDE1 expression.
Collapse
Affiliation(s)
- Senthilkumar Rajagopal
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Ancy D Nalli
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Divya P Kumar
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Sayak Bhattacharya
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Wenhui Hu
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Sunila Mahavadi
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - John R Grider
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Karnam S Murthy
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
21
|
Bibli SI, Yang G, Zhou Z, Wang R, Topouzis S, Papapetropoulos A. Role of cGMP in hydrogen sulfide signaling. Nitric Oxide 2014; 46:7-13. [PMID: 25553675 DOI: 10.1016/j.niox.2014.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/26/2014] [Accepted: 12/08/2014] [Indexed: 10/24/2022]
Abstract
The importance of hydrogen sulfide (H2S) in physiology and disease is being increasingly recognized in recent years. Unlike nitric oxide (NO) that signals mainly through soluble guanyl cyclase (sGC)/cGMP, H2S is more promiscuous, affecting multiple pathways. It interacts with ion channels, enzymes, transcription factors and receptors. It was originally reported that H2S does not alter the levels of cyclic nucleotides. More recent publications, however, have shown increases in intracellular cGMP following exposure of cells or tissues to exogenously administered or endogenously produced H2S. Herein, we discuss the evidence for the participation of cGMP in H2S signaling and reconcile the seemingly divergent results presented in the literature on the role of this cyclic nucleotide in the biological actions of H2S.
Collapse
Affiliation(s)
| | - Guangdong Yang
- School of Kinesiology, Cardiovascular and Metabolic Research Unit (CMRU), Lakehead University, Thunder Bay, Ontario, Canada
| | - Zongmin Zhou
- "G. P. Livanos" Laboratory, First Department of Critical Care and Pulmonary Services, Evangelismos Hospital, University of Athens, Athens, Greece
| | - Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
| | - Stavros Topouzis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece
| | - Andreas Papapetropoulos
- Faculty of Pharmacy, University of Athens, Athens, Greece; "G. P. Livanos" Laboratory, First Department of Critical Care and Pulmonary Services, Evangelismos Hospital, University of Athens, Athens, Greece.
| |
Collapse
|
22
|
Martin E, Golunski E, Laing ST, Estrera AL, Sharina IG. Alternative splicing impairs soluble guanylyl cyclase function in aortic aneurysm. Am J Physiol Heart Circ Physiol 2014; 307:H1565-75. [PMID: 25239802 DOI: 10.1152/ajpheart.00222.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) receptor soluble guanylyl cyclase (sGC) is a key regulator of several important vascular functions and is important for maintaining cardiovascular homeostasis and vascular plasticity. Diminished sGC expression and function contributes to pathogenesis of several cardiovascular diseases. However, the processes that control sGC expression in vascular tissue remain poorly understood. Previous work in animal and cell models revealed the complexity of alternative splicing of sGC genes and demonstrated its importance in modulation of sGC function. The aim of this study was to examine the role of alternative splicing of α1 and β1 sGC in healthy and diseased human vascular tissue. Our study found a variety of α1 and β1 sGC splice forms expressed in human aorta. Their composition and abundance were different between samples of aortic tissue removed during surgical repair of aortic aneurysm and samples of aortas without aneurysm. Aortas with aneurysm demonstrated decreased sGC activity, which correlated with increased expression of dysfunctional sGC splice variants. In addition, the expression of 55-kDa oxidation-resistant α1 isoform B sGC (α1-IsoB) was significantly lower in aortic samples with aneurysm. The α1-IsoB splice variant was demonstrated to support sGC activity in aortic lysates. Together, our results suggest that alternative splicing contributes to diminished sGC function in vascular dysfunction. Precise understanding of sGC splicing regulation could help to design new therapeutic interventions and to personalize sGC-targeting therapies in treatments of vascular disease.
Collapse
Affiliation(s)
- Emil Martin
- Department of Internal Medicine/Cardiology, The University of Texas Health Science Center in Houston Medical School, Houston, Texas; and
| | - Eva Golunski
- Department of Internal Medicine/Cardiology, The University of Texas Health Science Center in Houston Medical School, Houston, Texas; and
| | - Susan T Laing
- Department of Internal Medicine/Cardiology, The University of Texas Health Science Center in Houston Medical School, Houston, Texas; and
| | - Anthony L Estrera
- Department of Cardiothoracic Vascular Surgery, The University of Texas Health Science Center in Houston Medical School, Houston, Texas
| | - Iraida G Sharina
- Department of Internal Medicine/Cardiology, The University of Texas Health Science Center in Houston Medical School, Houston, Texas; and
| |
Collapse
|
23
|
Weissmann N, Lobo B, Pichl A, Parajuli N, Seimetz M, Puig-Pey R, Ferrer E, Peinado VI, Domínguez-Fandos D, Fysikopoulos A, Stasch JP, Ghofrani HA, Coll-Bonfill N, Frey R, Schermuly RT, García-Lucio J, Blanco I, Bednorz M, Tura-Ceide O, Tadele E, Brandes RP, Grimminger J, Klepetko W, Jaksch P, Rodriguez-Roisin R, Seeger W, Grimminger F, Barberà JA. Stimulation of soluble guanylate cyclase prevents cigarette smoke-induced pulmonary hypertension and emphysema. Am J Respir Crit Care Med 2014; 189:1359-73. [PMID: 24738736 DOI: 10.1164/rccm.201311-2037oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is a major cause of death worldwide. No therapy stopping progress of the disease is available. OBJECTIVES To investigate the role of the soluble guanylate cyclase (sGC)-cGMP axis in development of lung emphysema and pulmonary hypertension (PH) and to test whether the sGC-cGMP axis is a treatment target for these conditions. METHODS Investigations were performed in human lung tissue from patients with COPD, healthy donors, mice, and guinea pigs. Mice were exposed to cigarette smoke (CS) for 6 hours per day, 5 days per week for up to 6 months and treated with BAY 63-2521. Guinea pigs were exposed to CS from six cigarettes per day for 3 months, 5 days per week and treated with BAY 41-2272. Both BAY compounds are sGC stimulators. Gene and protein expression analysis were performed by quantitative real-time polymerase chain reaction and Western blotting. Lung compliance, hemodynamics, right ventricular heart mass alterations, and alveolar and vascular morphometry were performed, as well as inflammatory cell infiltrate assessment. In vitro assays of cell adhesion, proliferation, and apoptosis have been done. MEASUREMENTS AND MAIN RESULTS The functionally essential sGC β1-subunit was down-regulated in patients with COPD and in CS-exposed mice. sGC stimulators prevented the development of PH and emphysema in the two different CS-exposed animal models. sGC stimulation prevented peroxynitrite-induced apoptosis of alveolar and endothelial cells, reduced CS-induced inflammatory cell infiltrate in lung parenchyma, and inhibited adhesion of CS-stimulated neutrophils. CONCLUSIONS The sGC-cGMP axis is perturbed by chronic exposure to CS. Treatment of COPD animal models with sGC stimulators can prevent CS-induced PH and emphysema.
Collapse
Affiliation(s)
- Norbert Weissmann
- 1 Justus-Liebig University, Excellence Cluster Cardiopulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Giessen, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wei Y, Liao Y, Zavilowitz B, Ren J, Liu W, Chan P, Rohatgi R, Estilo G, Jackson EK, Wang WH, Satlin LM. Angiotensin II type 2 receptor regulates ROMK-like K⁺ channel activity in the renal cortical collecting duct during high dietary K⁺ adaptation. Am J Physiol Renal Physiol 2014; 307:F833-43. [PMID: 25100281 DOI: 10.1152/ajprenal.00141.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The kidney adjusts K⁺ excretion to match intake in part by regulation of the activity of apical K⁺ secretory channels, including renal outer medullary K⁺ (ROMK)-like K⁺ channels, in the cortical collecting duct (CCD). ANG II inhibits ROMK channels via the ANG II type 1 receptor (AT1R) during dietary K⁺ restriction. Because AT1Rs and ANG II type 2 receptors (AT2Rs) generally function in an antagonistic manner, we sought to characterize the regulation of ROMK channels by the AT2R. Patch-clamp experiments revealed that ANG II increased ROMK channel activity in CCDs isolated from high-K⁺ (HK)-fed but not normal K⁺ (NK)-fed rats. This response was blocked by PD-123319, an AT2R antagonist, but not by losartan, an AT1R antagonist, and was mimicked by the AT2R agonist CGP-42112. Nitric oxide (NO) synthase is present in CCD cells that express ROMK channels. Blockade of NO synthase with N-nitro-l-arginine methyl ester and free NO with 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide potassium salt completely abolished ANG II-stimulated ROMK channel activity. NO enhances the synthesis of cGMP, which inhibits phosphodiesterases (PDEs) that normally degrade cAMP; cAMP increases ROMK channel activity. Pretreatment of CCDs with IBMX, a broad-spectrum PDE inhibitor, or cilostamide, a PDE3 inhibitor, abolished the stimulatory effect of ANG II on ROMK channels. Furthermore, PKA inhibitor peptide, but not an activator of the exchange protein directly activated by cAMP (Epac), also prevented the stimulatory effect of ANG II. We conclude that ANG II acts at the AT2R to stimulate ROMK channel activity in CCDs from HK-fed rats, a response opposite to that mediated by the AT1R in dietary K⁺-restricted animals, via a NO/cGMP pathway linked to a cAMP-PKA pathway.
Collapse
Affiliation(s)
- Yuan Wei
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacology, New York Medical College, Valhalla, New York; Department of Cell Biology, New York University Medical Center, New York, New York
| | - Yi Liao
- Department of Cell Biology, New York University Medical Center, New York, New York
| | - Beth Zavilowitz
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jin Ren
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wen Liu
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pokman Chan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rajeev Rohatgi
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, James J. Peters Veterans Affairs Medical Center, Bronx, New York; and
| | - Genevieve Estilo
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Edwin K Jackson
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
25
|
Rapôso C, Luna RLDA, Nunes AKS, Thomé R, Peixoto CA. Role of iNOS-NO-cGMP signaling in modulation of inflammatory and myelination processes. Brain Res Bull 2014; 104:60-73. [PMID: 24727400 DOI: 10.1016/j.brainresbull.2014.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/27/2014] [Accepted: 04/03/2014] [Indexed: 01/25/2023]
Abstract
Nitric oxide (NO) is the main activator of the soluble guanylate cyclase (sGC)-guanosine 3'5' cyclic monophosphate (cGMP) pathway. The level of cGMP is regulated by phosphodiesterases (PDEs), which break down cGMP. It has been reported that levels of NO in the central nervous system (CNS) can greatly increase during demyelination and/or neuroinflammation. Controversially, in demyelination models, mice without iNOS may develop more severe cases of disease. Furthermore, cGMP accumulation caused by PDE inhibitors has an anti-inflammatory/neuroprotective effect in MS-models. The role of the NO-cGMP pathway in the nervous tissue is, therefore, complex and not fully understood. The aim of the present study was to contribute to existing knowledge of the role of this pathway in the CNS. Wild type (WT - C57BL/6) and iNOS(-/-) animals were treated with sildenafil (25mg/kg) for 8 weeks. Control animals were not treated. VCAM and ICAM (adhesion proteins), GFAP and Iba-1 (astrocyte and microglia markers, respectively), PKG (cGMP-dependent protein kinase), sGC, eNOS (constitutive endothelial NO sinthase) and GSTpi (a marker of mature oligodendrocytes) were evaluated in the cerebellum using immunohistochemistry or western blotting. Myelin was assessed by luxol fast blue staining and electron transmission microscopy. Treatment with sildenafil reduced ICAM and VCAM levels (anti-inflammatory effect) and increased GFAP and Iba-1 expression (clearance phenotype) in WT animals. The expression of VCAM, ICAM, GFAP, PKG and sGC was lower in iNOS(-/-) mice than in WT control animals. The treatment of iNOS(-/-) animals with sildenafil resulted in an increase of all proteins (pro-inflammatory effect). There was overexpression of eNOS in untreated iNOS(-/-) mice. The myelin structure of iNOS(-/-) animals was damaged in comparison with WT control. Sildenafil increased GSTpi and resulted in an improved myelin structure in iNOS(-/-) mice. In conclusion, NO-cGMP signaling plays a role in the regulation of inflammation and myelination processes. The accumulation of cGMP produced opposite effects in WT and iNOS(-/-) mice. This can be explained by the overexpression of eNOS in iNOS(-/-) mice, unbalancing cGMP signaling, or cGMP has a dual role in inflammation. Drugs that modulate the NO-sGC-cGMP pathway may be clinically beneficial in the treatment of neuroinflammatory/demyelinating disorders, but further studies of the regulation of this pathway are required.
Collapse
Affiliation(s)
- Catarina Rapôso
- Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Rua Monteiro Lobato, 255, CEP 13083-862, Campinas, SP, Brazil.
| | - Rayana Leal de Almeida Luna
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, s/n, CEP 50670-420, Recife, PE, Brazil.
| | - Ana Karolina Santana Nunes
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, s/n, CEP 50670-420, Recife, PE, Brazil.
| | - Rodolfo Thomé
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Rua Monteiro Lobato, 255, CEP 13083-862, Campinas, SP, Brazil.
| | - Christina Alves Peixoto
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, s/n, CEP 50670-420, Recife, PE, Brazil.
| |
Collapse
|
26
|
El-Sehemy A, Chang AC, Azad AK, Gupta N, Xu Z, Steed H, Karsan A, Fu Y. Notch activation augments nitric oxide/soluble guanylyl cyclase signaling in immortalized ovarian surface epithelial cells and ovarian cancer cells. Cell Signal 2013; 25:2780-7. [PMID: 24041655 DOI: 10.1016/j.cellsig.2013.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 09/06/2013] [Indexed: 11/25/2022]
Abstract
Nitric oxide (NO) is generated by tumor, stromal and endothelial cells and plays a multifaceted role in tumor biology. Many physiological functions of NO are mediated by soluble guanylyl cyclase (sGC) and NO/sGC signaling has been shown to promote proliferation and survival of ovarian cancer cells. However, how NO/sGC signaling is modulated in ovarian cancer cells has not been studied. The evolutionarily conserved Notch signaling pathway plays an oncogenic role in ovarian cancer. Here, we report that all three ovarian cancer cell lines we examined express a higher level of GUCY1B3 (the β subunit of sGC) compared to non-cancerous immortalized ovarian surface epithelial (IOSE) cell lines. Interestingly, the highest expression of GUCY1B3 in ovarian cancer OVCAR3 cells is concurrent with the expression of Notch3. In IOSE cells, forced activation of Notch3 increases the expression of GUCY1B3, NO-induced cGMP production, and the expression of cGMP-dependent protein kinase (PKG), thereby enhancing NO- and cGMP-induced phosphorylation of vasodilator-stimulated phosphoprotein (VASP, a direct PKG substrate protein). In contrast, inhibition of Notch by DAPT reduces GUCY1B3 expression and NO-induced cGMP production and VASP phosphorylation in OVCAR3 cells. Finally, we confirmed that inhibition of sGC by ODQ decreases growth of ovarian cancer cells. Together, our work demonstrates that Notch is a positive regulator of NO/sGC signaling in IOSE and ovarian cancer cells, providing the first evidence that Notch and NO signaling pathways interact in IOSE and ovarian cancer cells.
Collapse
Affiliation(s)
- Ahmed El-Sehemy
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Stojkov NJ, Baburski AZ, Bjelic MM, Sokanovic SJ, Mihajlovic AI, Drljaca DM, Janjic MM, Kostic TS, Andric SA. In vivo blockade of α1-adrenergic receptors mitigates stress-disturbed cAMP and cGMP signaling in Leydig cells. Mol Hum Reprod 2013; 20:77-88. [PMID: 23894150 DOI: 10.1093/molehr/gat052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The molecular mechanism of stress-associated reproductive dysfunction is complex and largely unknown. This study was designed to systematically analyze molecular effects of systemic in vivo blockade of α1-adrenergic receptors (α1-ADRs) on stress-induced disturbance of cAMP/cGMP signaling in testosterone-producing Leydig cells using the following parameters (i) level of circulating stress hormones, LH and testosterone; (ii) level of main molecular markers of Leydig cell functionality (testosterone, Insl3, cAMP); (iii) expression of cAMP signaling (cAMP 'producers'/'effectors'/'removers') and (iv) expression of NO-cGMP signaling (NO-cGMP 'producers'/'effectors'/'removers'). The results showed that oral administration of α1-ADR blocker before stress increased cGMP and diminished stress-reduced cAMP production in Leydig cells. In the same cells, stress-induced effects on cAMP/cGMP signaling pathways elements were changed. Sustained in vivo α1-ADR blockade completely abolished stress-increased transcription of most abundantly expressed phosphodiesterase that remove cAMP (Pde4b) and potentiated stress-increased expression of PRKA, the main stimulator of Leydig cell steroidogenesis. In the same Leydig cells, stress-decreased NOS3 expression was abolished, while stress-increased GUCY1 (cGMP 'producer') and PRKG1 (cGMP 'effector') were potentiated. It is possible that all molecules mentioned could contribute, at least in part, in recovery of Leydig cell testosterone production. Presented data provide new role of α1-ADRs in stress-triggered disturbance of cAMP/cGMP signaling, and new molecular insights into the relationship between stress and mammalian reproduction. Regardless of whether the effects of α1-blocker + stress are direct or indirect, the results are important in terms of human reproductive health and the wide use of α1-ADR antagonists, alone or in combination, to treat post-traumatic stress disorders, hypertension, benign prostatic hyperplasia symptoms and potential drugs for prostate cancer prevention/treatment.
Collapse
Affiliation(s)
- Natasa J Stojkov
- Reproductive Endocrinology and Signaling Group, Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Square 2, Novi Sad 21000, Serbia
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Biophysical techniques for detection of cAMP and cGMP in living cells. Int J Mol Sci 2013; 14:8025-46. [PMID: 23584022 PMCID: PMC3645729 DOI: 10.3390/ijms14048025] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/22/2013] [Accepted: 04/07/2013] [Indexed: 11/16/2022] Open
Abstract
Cyclic nucleotides cAMP and cGMP are ubiquitous second messengers which regulate myriads of functions in virtually all eukaryotic cells. Their intracellular effects are often mediated via discrete subcellular signaling microdomains. In this review, we will discuss state-of-the-art techniques to measure cAMP and cGMP in biological samples with a particular focus on live cell imaging approaches, which allow their detection with high temporal and spatial resolution in living cells and tissues. Finally, we will describe how these techniques can be applied to the analysis of second messenger dynamics in subcellular signaling microdomains.
Collapse
|
29
|
Chen Y, Chitapanarux T, Wu J, Soon RK, Melton AC, Yee HF. Inducible NOS mediates CNP-induced relaxation of intestinal myofibroblasts. Am J Physiol Gastrointest Liver Physiol 2013; 304:G673-9. [PMID: 23348803 PMCID: PMC3625877 DOI: 10.1152/ajpgi.00214.2012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Contraction of intestinal myofibroblasts (IMF) contributes to the development of strictures and fistulas seen in inflammatory bowel disease, but the mechanisms that regulate tension within these cells are poorly understood. In this study we investigated the role of nitric oxide (NO) signaling in C-type natriuretic peptide (CNP)-induced relaxation of IMF. We found that treatment with ODQ, a soluble guanylyl cyclase (sGC) inhibitor, or N(G)-nitro-L-arginine (L-NNA) or N(G)-monomethyl-L-arginine (L-NMMA), inhibitors of NO production, all impaired the relaxation of human and mouse IMF in response to CNP. ODQ, L-NNA, and L-NMMA also prevented CNP-induced elevations in cGMP concentrations, and L-NNA or L-NMMA blocked CNP-induced decreases in myosin light phosphorylation. IMF isolated from transgenic mice deficient in inducible nitric oxide synthase (iNOS) had reduced relaxation responses to CNP compared with IMF from control mice and were insensitive to the effects of ODQ, L-NNA, and L-NMMA on CNP treatment. Together these data indicate that stimulation of sGC though NO produced by iNOS activation is required for maximal CNP-induced relaxation in IMF.
Collapse
Affiliation(s)
- Yishi Chen
- 1Department of Medicine and Liver Center, University of California San Francisco, San Francisco, California; ,2AllCells, Emeryville, California; and
| | - Taned Chitapanarux
- 3Division of Gastrohepatology, Department of Medicine, Chiang Mai University, Thailand
| | - Jianfeng Wu
- 1Department of Medicine and Liver Center, University of California San Francisco, San Francisco, California;
| | - Russell K. Soon
- 1Department of Medicine and Liver Center, University of California San Francisco, San Francisco, California;
| | - Andrew C. Melton
- 1Department of Medicine and Liver Center, University of California San Francisco, San Francisco, California;
| | - Hal F. Yee
- 1Department of Medicine and Liver Center, University of California San Francisco, San Francisco, California;
| |
Collapse
|
30
|
Fritz BG, Roberts SA, Ahmed A, Breci L, Li W, Weichsel A, Brailey JL, Wysocki VH, Tama F, Montfort WR. Molecular model of a soluble guanylyl cyclase fragment determined by small-angle X-ray scattering and chemical cross-linking. Biochemistry 2013; 52:1568-82. [PMID: 23363317 PMCID: PMC3607398 DOI: 10.1021/bi301570m] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Soluble guanylyl/guanylate cyclase (sGC) converts GTP to cGMP after binding nitric oxide, leading to smooth muscle relaxation and vasodilation. Impaired sGC activity is common in cardiovascular disease, and sGC stimulatory compounds are vigorously sought. sGC is a 150 kDa heterodimeric protein with two H-NOX domains (one with heme, one without), two PAS domains, a coiled-coil domain, and two cyclase domains. Binding of NO to the sGC heme leads to proximal histidine release and stimulation of catalytic activity. To begin to understand how binding leads to activation, we examined truncated sGC proteins from Manduca sexta (tobacco hornworm) that bind NO, CO, and stimulatory compound YC-1 but lack the cyclase domains. We determined the overall shape of truncated M. sexta sGC using analytical ultracentrifugation and small-angle X-ray scattering (SAXS), revealing an elongated molecule with dimensions of 115 Å × 90 Å × 75 Å. Binding of NO, CO, or YC-1 had little effect on shape. Using chemical cross-linking and tandem mass spectrometry, we identified 20 intermolecular contacts, allowing us to fit homology models of the individual domains into the SAXS-derived molecular envelope. The resulting model displays a central parallel coiled-coil platform upon which the H-NOX and PAS domains are assembled. The β1 H-NOX and α1 PAS domains are in contact and form the core signaling complex, while the α1 H-NOX domain can be removed without a significant effect on ligand binding or overall shape. Removal of 21 residues from the C-terminus yields a protein with dramatically increased proximal histidine release rates upon NO binding.
Collapse
Affiliation(s)
- Bradley G. Fritz
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Sue A. Roberts
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Aqeel Ahmed
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Linda Breci
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Wenzhou Li
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Andrzej Weichsel
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Jacqueline L. Brailey
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Florence Tama
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - William R. Montfort
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| |
Collapse
|
31
|
Mujoo K, Krumenacker JS, Murad F. Nitric oxide-cyclic GMP signaling in stem cell differentiation. Free Radic Biol Med 2011; 51:2150-7. [PMID: 22019632 PMCID: PMC3232180 DOI: 10.1016/j.freeradbiomed.2011.09.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/27/2011] [Accepted: 09/29/2011] [Indexed: 12/15/2022]
Abstract
The nitric oxide-cyclic GMP (NO-cGMP) pathway mediates important physiological functions associated with various integrative body systems including the cardiovascular and nervous systems. Furthermore, NO regulates cell growth, survival, apoptosis, proliferation, and differentiation at the cellular level. To understand the significance of the NO-cGMP pathway in development and differentiation, studies have been conducted both in developing embryos and in stem cells. Manipulation of the NO-cGMP pathway, by employing activators and inhibitors as pharmacological probes, and genetic manipulation of NO signaling components have implicated the involvement of this pathway in the regulation of stem cell differentiation. This review focuses on some of the work pertaining to the role of NO-cGMP in the differentiation of stem cells into cells of various lineages, particularly into myocardial cells, and in stem cell-based therapy.
Collapse
Affiliation(s)
- Kalpana Mujoo
- Brown Foundation Institute of Molecular Medicine, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | | | | |
Collapse
|
32
|
Doherty GH. Nitric oxide in neurodegeneration: potential benefits of non-steroidal anti-inflammatories. Neurosci Bull 2011; 27:366-82. [PMID: 22108814 PMCID: PMC5560384 DOI: 10.1007/s12264-011-1530-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/13/2011] [Indexed: 12/28/2022] Open
Abstract
The cellular messenger nitric oxide (NO) has been linked to neurodegenerative disorders due to the increased expression of the enzymes that catalyze its synthesis in postmortem tissues derived from sufferers of these diseases. Nitrated proteins have also been detected in these samples, revealing that NO is biologically active in regions damaged during neurodegeneration. Modulation of NO levels has been reported not only in the neurons of the central nervous system, but also in the glial cells (microglia and astroglia) activated during the neuroinflammatory response. Neuroinflammation has been found in some neurodegenerative conditions, and inhibition of these neuroinflammatory signals has been shown to delay the progress of such disorders. Thus NO and the pathways triggering its release are emerging as an important research focus in the search for strategies to prevent, halt or cure neurodegenerative diseases.
Collapse
Affiliation(s)
- Gayle Helane Doherty
- School of Biology, St Andrews University, St Andrews, Fife KY169TS, United Kingdom.
| |
Collapse
|
33
|
Ramanathan S, Mazzalupo S, Boitano S, Montfort WR. Thrombospondin-1 and angiotensin II inhibit soluble guanylyl cyclase through an increase in intracellular calcium concentration. Biochemistry 2011; 50:7787-99. [PMID: 21823650 DOI: 10.1021/bi201060c] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nitric oxide (NO) regulates cardiovascular hemostasis by binding to soluble guanylyl cyclase (sGC), leading to cGMP production, reduced cytosolic calcium concentration ([Ca(2+)](i)), and vasorelaxation. Thrombospondin-1 (TSP-1), a secreted matricellular protein, was recently discovered to inhibit NO signaling and sGC activity. Inhibition of sGC requires binding to cell-surface receptor CD47. Here, we show that a TSP-1 C-terminal fragment (E3CaG1) readily inhibits sGC in Jurkat T cells and that inhibition requires an increase in [Ca(2+)](i). Using flow cytometry, we show that E3CaG1 binds directly to CD47 on the surface of Jurkat T cells. Using digital imaging microscopy on live cells, we further show that E3CaG1 binding results in a substantial increase in [Ca(2+)](i), up to 300 nM. Addition of angiotensin II, a potent vasoconstrictor known to increase [Ca(2+)](i), also strongly inhibits sGC activity. sGC isolated from calcium-treated cells or from cell-free lysates supplemented with Ca(2+) remains inhibited, while addition of kinase inhibitor staurosporine prevents inhibition, indicating inhibition is likely due to phosphorylation. Inhibition is through an increase in K(m) for GTP, which rises to 834 μM for the NO-stimulated protein, a 13-fold increase over the uninhibited protein. Compounds YC-1 and BAY 41-2272, allosteric stimulators of sGC that are of interest for treating hypertension, overcome E3CaG1-mediated inhibition of NO-ligated sGC. Taken together, these data suggest that sGC not only lowers [Ca(2+)](i) in response to NO, inducing vasodilation, but also is inhibited by high [Ca(2+)](i), providing a fine balance between signals for vasodilation and vasoconstriction.
Collapse
Affiliation(s)
- Saumya Ramanathan
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85721, United States
| | | | | | | |
Collapse
|
34
|
Sharina IG, Cote GJ, Martin E, Doursout MF, Murad F. RNA splicing in regulation of nitric oxide receptor soluble guanylyl cyclase. Nitric Oxide 2011; 25:265-74. [PMID: 21867767 DOI: 10.1016/j.niox.2011.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 08/01/2011] [Accepted: 08/04/2011] [Indexed: 11/19/2022]
Abstract
Soluble guanylyl cyclase (sGC) is a key protein in the nitric oxide (NO)/-cGMP signaling pathway. sGC activity is involved in a number of important physiological processes including smooth muscle relaxation, neurotransmission and platelet aggregation and adhesion. Regulation of sGC expression and activity emerges as a crucial factor in control of sGC function in normal and pathological conditions. Recently accumulated evidence strongly indicates that the regulation of sGC expression is a complex process modulated on several levels including transcription, post-transcriptional regulation, translation and protein stability. Presently our understanding of mechanisms governing regulation of sGC expression remains very limited and awaits systematic investigation. Among other ways, the expression of sGC subunits is modulated at the levels of mRNA abundance and transcript diversity. In this review we summarize available information on different mechanisms (including transcriptional activation, mRNA stability and alternative splicing) involved in the modulation of mRNA levels of sGC subunits in response to various environmental clues. We also summarize and cross-reference the information on human sGC splice forms available in the literature and in genomic databases. This review highlights the fact that the study of the biological role and regulation of sGC splicing will bring new insights to our understanding of NO/cGMP biology.
Collapse
Affiliation(s)
- Iraida G Sharina
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX, USA.
| | | | | | | | | |
Collapse
|
35
|
Effects of nitric oxide on the survival and neuritogenesis of cerebellar Purkinje neurons. J Mol Neurosci 2011; 46:336-42. [PMID: 21728061 DOI: 10.1007/s12031-011-9590-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 06/22/2011] [Indexed: 01/21/2023]
Abstract
Nitric oxide has been investigated widely both during neurodevelopment and in neurological diseases. However, whilst it has been established that nitric oxide-producing enzymes of nitric oxide synthase family are expressed in cerebellar Purkinje neurons, the effects of nitric oxide on the viability and morphology of these neurons remain unknown. Here, we have demonstrated that the activity of neuronal nitric oxide synthase, but not the inducible or endothelial forms of this enzyme, is required to support the survival of a proportion of cerebellar Purkinje neurons in vitro. We discovered that donation of high concentrations of exogenous nitric oxide reduces Purkinje neuron survival in culture and that peroxynitrite is also toxic to these cells. Finally, we demonstrated that exogenous nitric oxide and peroxynitrite reduce both the magnitude and the complexity of the neurite arbour extended by cerebellar Purkinje neurons. Taken together, these findings reveal that whilst a low level of endogenous nitric oxide, released by the activity of neuronal nitric oxide synthase, is beneficial to cerebellar Purkinje neurons in vitro, high levels of exogenous nitric oxide and peroxynitrite are detrimental to both the survival of these neurons and to their ability to extend processes and form functional neural networks.
Collapse
|
36
|
Curatola AM, Xu J, Hendricks-Munoz KD. Cyclic GMP protects endothelial progenitors from oxidative stress. Angiogenesis 2011; 14:267-79. [PMID: 21499920 DOI: 10.1007/s10456-011-9211-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 03/30/2011] [Indexed: 12/24/2022]
Abstract
Endothelial progenitor cells (EPCs) play a critical role in the repair of damaged blood vessels and/or in the growth of new ones in ischemic tissues. Elevated levels of oxygen radicals, which accumulate in the ischemic tissue, could compromise the angiogenic potential of EPCs. To determine if oxidative stress alters the angiogenic response of EPCs and to identify possible cellular targets that protect EPCs from the damaging effects of oxidative stress, we have investigated vascular development in embryonic bodies (EBs) under hyperoxic conditions. Murine EBs at differentiaton day 2 were cultured for 3 days under normoxic (21% O(2)) or hyperoxic (60% O(2)) conditions. Hyperoxic EBs showed a moderate reduction in Pecam-1, Vegfr-2, eNOS and Tie2 mRNA levels compared to normoxic EBs. However, immunostaining of hyperoxic EBs with antibodies against PECAM-1 after 1 week recovery at room air revealed a defective vasculature completely deficient in branches, while normoxic EBs developed a normal vascular plexus. Oxygen-induced defective vascular development correlated with a dramatic decrease in soluble guanylyl cyclase, phosphodiesterase (Pde) 4B and Pde4C mRNAs. Oxidative stress did not affect the expression of adenylyl cyclase 6 and Pde5. The abnormal vascular development caused by hyperoxia was reverted by pharmacological treatments that increased cGMP levels, such as 8-bromo-cGMP or 4-{[3',4'-(methylenedioxy)benzyl]amino}-6-methoxyquinazoline, a specific inhibitor of PDE5. These results indicated that oxidative stress inhibits vascular development from EPCs through its effects on levels of cyclic nucleotides and suggested that therapies that target cyclic nucleotide turnover may be useful in protecting vascular repair under oxidative conditions.
Collapse
Affiliation(s)
- Anna Maria Curatola
- Division of Neonatology, Department of Pediatrics, New York University School of Medicine, NY 10016, USA.
| | | | | |
Collapse
|
37
|
1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one induces cell cycle arrest and apoptosis in HeLa cells by preventing microtubule polymerization. Biochem Biophys Res Commun 2011; 408:287-92. [PMID: 21501588 DOI: 10.1016/j.bbrc.2011.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 04/04/2011] [Indexed: 01/18/2023]
Abstract
1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) is known as a specific inhibitor of soluble guanylyl cyclase (sGC). Previously, however, ODQ was reported to induce cell death via sGC-dependent and sGC-independent means in a variety of cell types. The aim of this study was to investigate the mechanism by which ODQ induces cell death in HeLa cells. Treatment of HeLa cells with ODQ induced a concentration-dependent decrease in cell viability over the range from 10 to 100 μM. DNA fragmentation and fluorescence-activated cell sorting analysis using annexin V and propidium iodide staining revealed that ODQ triggered apoptosis at concentrations of 50 and 100 μM within 24 to 48 h. The addition of 8-Br-cGMP in the presence of ODQ failed to rescue HeLa cells from death, suggesting that the inhibition of sGC was not responsible for the pro-apoptotic action of ODQ. ODQ arrested the cell cycle at the G2/M phase and caused disassembly of the microtubule network. This process was reversed by dithiothreitol. In addition, ODQ was shown to inhibit the polymerization of purified tubulin, and this was also prevented by dithiothreitol. These results indicate that ODQ inhibits microtubule assembly by direct oxidation of tubulin, induces cell cycle arrest at the G2/M phase, and triggers apoptosis in HeLa cells.
Collapse
|
38
|
Castro LRV, Schittl J, Fischmeister R. Feedback control through cGMP-dependent protein kinase contributes to differential regulation and compartmentation of cGMP in rat cardiac myocytes. Circ Res 2010; 107:1232-40. [PMID: 20847310 DOI: 10.1161/circresaha.110.226712] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE We have shown recently that particulate (pGC) and soluble guanylyl (sGC) cyclases synthesize cGMP in different compartments in adult rat ventricular myocytes (ARVMs). OBJECTIVE We hypothesized that cGMP-dependent protein kinase (PKG) exerts a feedback control on cGMP concentration contributing to its intracellular compartmentation. METHODS AND RESULTS Global cGMP levels, cGMP-phosphodiesterase (PDE) and pGC enzymatic activities were determined in purified ARVMs. Subsarcolemmal cGMP signals were monitored in single cells by recording the cGMP-gated current (I(CNG)) in myocytes expressing the wild-type rat olfactory cyclic nucleotide-gated (CNG) channel. Whereas the NO donor S-nitroso-N-acetyl-penicillamine (SNAP) (100 μmol/L) produced little effect on I(CNG), the response increased 2-fold in the presence of the PKG inhibitors KT5823 (50 nmol/L) or DT-2 (2 μmol/L). The effect of KT5823 was abolished in the presence of the nonselective cyclic nucleotide PDE inhibitor 3-isobutyl-1-methylxantine (IBMX) (100 μmol/L) or the selective cGMP-PDE5 inhibitor sildenafil (100 nmol/L). PKG inhibition also potentiated the effect of SNAP on global cGMP levels and fully blocked the increase in cGMP-PDE5 activity. In contrast, PKG inhibition decreased by ≈50% the I(CNG) response to ANP (10 and 100 nmol/L), even in the presence of IBMX. Conversely, PKG activation increased the I(CNG) response to ANP and amplified the stimulatory effect of ANP on pGC activity. CONCLUSIONS PKG activation in adult cardiomyocytes limits the accumulation of cGMP induced by NO donors via PDE5 stimulation but increases that induced by natriuretic peptides. These findings support the paradigm that cGMP is not uniformly distributed in the cytosol and identifies PKG as a key component in this process.
Collapse
Affiliation(s)
- Liliana R V Castro
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Inserm U769, Châtenay-Malabry Cedex, France
| | | | | |
Collapse
|
39
|
Kostic TS, Stojkov NJ, Janjic MM, Andric SA. Structural complexity of the testis and PKG I / StAR interaction regulate the Leydig cell adaptive response to repeated immobilization stress. ACTA ACUST UNITED AC 2010; 33:717-29. [DOI: 10.1111/j.1365-2605.2009.01018.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
40
|
Teng Y, Xu W, Ma M. cGMP is required for seed germination in Arabidopsis thaliana. JOURNAL OF PLANT PHYSIOLOGY 2010; 167:885-9. [PMID: 20170981 DOI: 10.1016/j.jplph.2010.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 01/29/2010] [Accepted: 01/29/2010] [Indexed: 05/05/2023]
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) is an important second messenger in animals, and is emerging as a player in regulatory functions in plants. In this study, we investigated the role of cGMP in seed germination in Arabidopsis thaliana (Col-0). We demonstrated that both, a membrane-permeant analogue of cGMP (8-Br-cGMP) and the cyclic nucleotide phosphodiesterase (PDE) inhibitor Tadalafil promoted A. thaliana seed germination, whereas the guanylate cyclase inhibitor LY 83583 (6-anilino-5,8-quinolinedione; LY) inhibited it. LY blocked gibberellic acid (GA)-induced seed germination, whereas GA and 8-Br-cGMP co-treatment increased the germination rate and more effectively overcame LY-inhibition than 8-Br-cGMP alone. The gibberellin biosynthesis inhibitor paclobutrazol (PAC) also blocked 8-Br-cGMP and Tadalafil promotion of seed germination. Furthermore, 8-Br-cGMP and Tadalafil decreased abscisic acid (ABA) sensitivity during seed germination. These findings highlight that cGMP is a positive regulator and plays a crucial role in Arabidopsis seed germination. Furthermore, both GA and cGMP are required for seed germination.
Collapse
Affiliation(s)
- Yao Teng
- Key Laboratory of Photosynthesis and Environmental Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, PR China
| | | | | |
Collapse
|
41
|
Jones AW, Durante W, Korthuis RJ. Heme oxygenase-1 deficiency leads to alteration of soluble guanylate cyclase redox regulation. J Pharmacol Exp Ther 2010; 335:85-91. [PMID: 20605906 DOI: 10.1124/jpet.110.169755] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Heme oxygenase-1 knockout, H(mox)1(-/-), mice exhibit exacerbated vascular lesions after ischemia-reperfusion and mechanical injury. Surprisingly, we found no studies that reported contractile responses and sensitivity to vasorelaxants in H(mox)1(-/-) mice. The contractile responses [superior mesenteric arteries (SMA), from female H(mox)1(-/-) mice] exhibited increased sensitivity to phenylephrine (p < 0.001). Cumulative addition of acetylcholine relaxed SMA, with the residual contraction remaining 2 times higher in H(mox)1(-/-) mice (p < 0.001). Sodium nitroprusside (SNP, an NO donor) and 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole [YC-1; acts directly on soluble guanylate cyclase (sGC)] led to further relaxation, yet the residual contraction remained 2 to 3 times higher in H(mox)1(-/-) than H(mox)1(+/+) mice (p < 0.001). Branches from H(mox)1(-/-) mesenteric and renal arteries also showed reduced relaxation (p < 0.025). Relaxation of SMA was measured to 4-({(4-carboxybutyl) [2-(5-fluoro-2-{[4'-(trifluoromethyl) biphenyl-4-yl] methoxy}phenyl)ethyl]amino}benzoic acid (BAY 60-2770), which is a more effective activator of oxidized/heme-free sGC; and to 5-cyclopropyl-2-{1-(2-fluoro-benzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl}-pyrimidin-4-ylamine (BAY 41-2272), a more effective stimulator of reduced sGC. H(mox)1(-/-) arteries were 15 times more sensitive to BAY 60-2770 (p < 0.025) than were H(mox)1(+/+) arteries. Pretreatment with 1H-[1,2,4]oxadiazolo[3,4-a]quinoxalin-1-one (ODQ), an oxidizer of sGC, predictably shifted the BAY 60-2770 response of H(mox)1(+/+) to the left (p < 0.01) and BAY 41-2272 response to the right (p < 0.01). ODQ had little effect on the responses of H(mox)1(-/-) arteries, indicating that much of sGC was oxidized/heme-free. Western analyses of sGC in SMA indicated that both α1 and β1 subunit levels were reduced to <50% of H(mox)1(+/+) level (p < 0.025). These findings support the hypothesis that the antioxidant function of H(mox)1 plays a significant role in the maintenance of sGC in a reduced state, which is resistant to degradation and is sensitive to NO. This function may be especially important in reducing vascular damage during ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Allan W Jones
- Department of Medical Pharmacology, and Physiology, University of Missouri-Columbia, Columbia, MO 65212, USA.
| | | | | |
Collapse
|
42
|
Stratton RC, Squires PE, Green AK. 17Beta-estradiol elevates cGMP and, via plasma membrane recruitment of protein kinase GIalpha, stimulates Ca2+ efflux from rat hepatocytes. J Biol Chem 2010; 285:27201-27212. [PMID: 20566641 DOI: 10.1074/jbc.m110.103630] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rapid non-genomic effects of 17beta-estradiol, the principal circulating estrogen, have been observed in a wide variety of cell types. Here we investigate rapid signaling effects of 17beta-estradiol in rat hepatocytes. We show that, above a threshold concentration of 1 nm, 17beta-estradiol, but not 17alpha-estradiol, stimulates particulate guanylyl cyclase to elevate cGMP, which through activation and plasma membrane recruitment of protein kinase G isoform Ialpha, stimulates plasma membrane Ca(2+)-ATPase-mediated Ca(2+) efflux from rat hepatocytes. These effects are extremely rapid in onset and are mimicked by a membrane-impermeant 17beta-estradiol-BSA conjugate, suggesting that 17beta-estradiol acts at the extracellular face of the plasma membrane. We also show that 17beta-estradiol binds specifically to the intact hepatocyte plasma membrane through an interaction that is competed by an excess of atrial natriuretic peptide but also shows many similarities to the pharmacological characteristics of the putative gamma-adrenergic receptor. We, therefore, propose that the observed rapid signaling effects of 17beta-estradiol are mediated either through the guanylyl cyclase A receptor for atrial natriuretic peptide or through the gamma-adrenergic receptor, which is either itself a transmembrane guanylyl cyclase or activates a transmembrane guanylyl cyclase through cross-talk signaling.
Collapse
Affiliation(s)
- Rebecca C Stratton
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom
| | - Paul E Squires
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom
| | - Anne K Green
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
43
|
Uray FP, de Alfonzo RG, de Becemberg IL, Alfonzo MJ. Muscarinic agonists acting through M2 acetylcholine receptors stimulate the migration of an NO-sensitive guanylyl cyclase to the plasma membrane of bovine tracheal smooth muscle. J Recept Signal Transduct Res 2010; 30:10-23. [PMID: 19911949 DOI: 10.3109/10799890903325585] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Muscarinic agonists acting on bovine tracheal smooth muscle (BTSM) induce two separate cGMP signals, one at 20 sec associated with NO-sensitive-soluble-guanylyl-cyclase (NO-sGC) and another at 60 sec, linked to natriuretic-peptide-GC. The 20-sec-cGMP novel cascade starts with mAChRs, via unknown components, activates an NO-sGC. To unravel this cascade, in crude membranes isolated from intact BTSM strips exposed to muscarinic agonists, we detected GC activities increments at 20 sec and 60 sec. The 20-sec-GC is a NO-sensitive-GC, identified as alpha(1)beta(1)-heterodimer. In reconstitution experiments with purified plasma membranes and cytosol, muscarinic agonists induced an NO-sGC migration in a dose-dependent manner, being inhibited by muscarinic antagonists displaying an M(2)AChR profile and blocked by PTX, suggesting the involvement of G(o)/G(i) proteins. The NO-sGC related to migration was isolated and identified as an alpha(1)beta(1)-heterodimer. This work shows that muscarinic agonists in BTSM induce a massive and selective alpha(1)beta(1)-NO-sGC migration from cytoplasm to plasma membranes being responsible for the 20-sec-cGMP signal.
Collapse
Affiliation(s)
- Fabiola Pláceres Uray
- Sección de Biomembranas, Instituto de Medicina Experimental, Cátedra de Patología General y Fisiopatología, Escuela Luís Razetti, Facultad de Medicina. Universidad Central de Venezuela, Caracas, Venezuela
| | | | | | | |
Collapse
|
44
|
Fukutani T, Iino S, Nojyo Y. The expression of soluble guanylate cyclase in the vasculature of rat skeletal muscle. ACTA ACUST UNITED AC 2010; 72:117-26. [PMID: 20009348 DOI: 10.1679/aohc.72.117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nitric oxide (NO) has various roles in the skeletal musculature in both normal and pathological conditions. NO primarily activates soluble guanylate cyclase (sGC) and mediates subsequent intracellular signaling in target cells. We sought to identify the target cells of NO in the rat skeletal musculature, using subtypes of sGCalpha1 and sGCbeta1 antibodies. Immunohistochemistry revealed that both antibodies stained the same cells with round or oval shapes, having several long processes. The sGC-immunopositive cells co-expressed NG2 chondroitin sulfate proteoglycan, a marker of pericytes. The sGC-immunopositive cells were associated with capillaries and formed cellular networks with elongated cytoplasmic processes. sGCalpha1 and sGCbeta1 were not found in muscle sarcolemma that were stained by anti-dystrophin, or neuromuscular junctions, as detected by anti-synaptophysin. Based on these findings, we concluded that sGC immunoreactivity was specifically distributed in capillary pericytes. Pericytes in the skeletal musculature have been shown to be target cells of NO and are involved in the microvascular blood flow.
Collapse
Affiliation(s)
- Tamotsu Fukutani
- Department of Physical Therapy, Nagano Rehabilitation College, Nagano, Japan.
| | | | | |
Collapse
|
45
|
Synthesis and biological evaluation of oxadiazole derivatives as inhibitors of soluble guanylyl cyclase. Bioorg Med Chem 2009; 18:1288-96. [PMID: 20036129 DOI: 10.1016/j.bmc.2009.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 12/02/2009] [Accepted: 12/08/2009] [Indexed: 11/20/2022]
Abstract
Soluble guanylyl cyclase (sGC) is an ubiquitously expressed enzyme that generates the second messenger cGMP and hence, leads to a number of physiological responses including vasodilation, inhibition of platelet aggregation and neurotransmission. Whilst many activating and stimulating modulators of sGC were identified and studied in recent years, only two selective inhibitors are known: ODQ and NS 2028. Furthermore, a synthetic approach to these inhibitors has not been reported yet. Herein, we describe a novel and efficient synthesis of these inhibitors, as well as the preparation of three different classes of NS 2028 analogues. Biological evaluation of this library using rat aortic smooth muscle cells revealed four new compounds with good to moderate sGC inhibitory activity. Our experiments underline the major importance of the oxadiazole ring in ODQ and NS 2028 for the efficiency of this class of inhibitors.
Collapse
|
46
|
Ritchie RH, Irvine JC, Rosenkranz AC, Patel R, Wendt IR, Horowitz JD, Kemp-Harper BK. Exploiting cGMP-based therapies for the prevention of left ventricular hypertrophy: NO* and beyond. Pharmacol Ther 2009; 124:279-300. [PMID: 19723539 DOI: 10.1016/j.pharmthera.2009.08.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 08/14/2009] [Indexed: 02/07/2023]
Abstract
Left ventricular hypertrophy (LVH), an increased left ventricular (LV) mass, is common to many cardiovascular disorders, initially developing as an adaptive response to maintain myocardial function. In the longer term, this LV remodelling becomes maladaptive, with progressive decline in LV contractility and diastolic function. Indeed LVH is recognised as an important blood-pressure independent predictor of cardiovascular morbidity and mortality. The clinical efficacy of current treatments for LVH is reduced, however, by their tendency to slow disease progression rather than induce its reversal, and thus the development of new therapies for LVH is paramount. The signalling molecule cyclic guanosine-3',5'-monophosphate (cGMP), well-recognised for its role in regulating vascular tone, is now being increasingly identified as an important anti-hypertrophic mediator. This review is focused on the various means by which cGMP can be stimulated in the heart, such as via the natriuretic peptides, to exert anti-hypertrophic actions. In particular we address the limitations of traditional nitric oxide (NO*) donors in the face of the potential therapeutic advantages offered by novel alternatives; NO* siblings, ligands of the cGMP-generating enzymes, soluble (sGC) and particulate guanylyl cyclases (pGC), and phosphodiesterase inhibitors. Further impact of cGMP within the cardiovascular system is also discussed with a view to representing cGMP-based therapies as innovative pharmacotherapy, alone or concurrent with standard care, for the management of LVH.
Collapse
Affiliation(s)
- Rebecca H Ritchie
- Heart Failure Pharmacology, Baker IDI Heart & Diabetes Institute Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Recent developments of biophysical and electrophysiological techniques have enabled researchers to monitor levels of free intracellular cGMP in real-time and in intact living cells. These techniques are based on the use of cGMP sensors, which respond to cGMP with changes in transmembrane ion current or changes in fluorescence. Here, we describe the principles of these techniques, compare them in terms of sensitivity and discuss possible application for current cell biology and physiology.
Collapse
|
48
|
Abstract
Melatonin influences the second messenger cyclic guanosine 3',5'-monophosphate (cGMP) signaling pathway in pancreatic beta-cells via a receptor-mediated mechanism. In the present study, it was determined how the regulation of cGMP concentrations by melatonin proceeds. The results provide evidence that melatonin acts via the soluble guanylate cyclase (sGC), as molecular investigations demonstrated that long-term incubation with melatonin significantly reduced the expression levels of the sGC mRNA in rat insulinoma beta-cells (INS1) cells, whereas mRNA expression of membrane guanylate cyclases was unaffected. Incubation with melatonin abolished the S-nitrosoacetyl penicillamine-induced increase of cGMP concentrations in INS1 cells. In addition, the cGMP-inhibitory effect of melatonin was reversed by preincubation with the sGC inhibitors 1H-(1,2,4)oxadiazolo(4,3-alpha)quinoxalin-1-one and 4H-8-bromo-1,2,4-oxadiazolo(3,4-d)benz(b)(1,4)oxazin-1-one. Nitric oxide (NO) production was not influenced after 1 hr of melatonin application, but was influenced after a 4 hr incubation period. Preincubation of INS1 cells with the NO synthase inhibitor N(G)-monomethyl-l-arginine did not abolish the cGMP-inhibitory effect of melatonin. Transcripts of cyclic nucleotide-gated (CNG) channels were significantly reduced after melatonin treatment in a dose-dependent manner, indicating the involvement of these channels in mediating the melatonin effect in INS1 cells. The results of this study demonstrate that melatonin mediates its inhibitory effect on cGMP concentrations in pancreatic beta-cells by inhibiting the sGC, but does not influence NO concentration or NO synthase activity in short-term incubation experiments. In addition, it was demonstrated that melatonin is involved in modulation of CNG channel mRNA.
Collapse
Affiliation(s)
- Ina Stumpf
- Institute of Anatomy and Cell Biology, Martin Luther University, Halle-Wittenberg, Halle/Saale, Germany
| | | | | |
Collapse
|
49
|
Isenberg JS, Martin-Manso G, Maxhimer JB, Roberts DD. Regulation of nitric oxide signalling by thrombospondin 1: implications for anti-angiogenic therapies. Nat Rev Cancer 2009; 9:182-94. [PMID: 19194382 PMCID: PMC2796182 DOI: 10.1038/nrc2561] [Citation(s) in RCA: 220] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In addition to long-term regulation of angiogenesis, angiogenic growth factor signalling through nitric oxide (NO) acutely controls blood flow and haemostasis. Inhibition of this pathway may account for the hypertensive and pro-thrombotic side effects of the vascular endothelial growth factor antagonists that are currently used for cancer treatment. The first identified endogenous angiogenesis inhibitor, thrombospondin 1, also controls tissue perfusion, haemostasis and radiosensitivity by antagonizing NO signalling. We examine the role of these and other emerging activities of thrombospondin 1 in cancer. Clarifying how endogenous and therapeutic angiogenesis inhibitors regulate vascular NO signalling could facilitate development of more selective inhibitors.
Collapse
Affiliation(s)
- Jeff S Isenberg
- Hemostasis and Vascular Biology Research Institute and the Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | | | | | |
Collapse
|
50
|
Fernandes D, Sordi R, Pacheco LK, Nardi GM, Heckert BT, Villela CG, Lobo AR, Barja-Fidalgo C, Assreuy J. Late, but not early, inhibition of soluble guanylate cyclase decreases mortality in a rat sepsis model. J Pharmacol Exp Ther 2008; 328:991-9. [PMID: 19073910 DOI: 10.1124/jpet.108.142034] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Overproduction of nitric oxide and activation of soluble guanylate cyclase (sGC) are important in sepsis-induced hypotension and hyporesponsiveness to vasoconstrictors. A time course of the expression and activity of sGC in a sepsis model [cecal ligation and puncture (CLP)] was evaluated in rats. Soluble GC alpha-1 and beta-1 subunit mRNA levels increased in the lungs, but not in the aorta. However, in both tissues, the protein levels increased 24 h after sepsis and remained high for up to 48 h. Sodium nitroprusside-stimulated cGMP accumulation was higher 48 h after CLP in the lung and aorta. NOS-2 protein expression peaked 24 h after CLP, decreasing thereafter. The impact of inhibiting the expression of sGC early (8 h) or late (20 h) on vascular reactivity and the indexes of organ damage and mortality were also studied. Late administration of methylene blue (MB) or ODQ (1H-[1,2,4]-oxadiazole[4,3-a]quinoxalin-1-one) restored the blood pressure and vascular responsiveness to vasoconstrictors to normal levels but was ineffective in early sepsis. Late MB injection reduced the plasma levels of urea, creatinine, and lactate. MB improved the survival if administered late, but it increased the mortality when administrated early after sepsis onset. The increased sGC expression/activity may be relevant for the late hypotension and hyporesponsiveness to vasoconstrictors in sepsis. In accordance, MB increased survival if administered in late sepsis, but not in early sepsis. Therefore, differential responsiveness to sGC during the course of sepsis may determine the success or failure of treatment with sGC inhibitors.
Collapse
Affiliation(s)
- Daniel Fernandes
- Department of Pharmacology, University Campus, Trindade, Biological Sciences Centre, Block "D," Florianopolis, Santa Catarina 88049-900, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|