Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Science ; 382(6670): 519, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37917686
2.
Cells ; 12(3)2023 02 01.
Article in English | MEDLINE | ID: mdl-36766813

ABSTRACT

NO-stimulated guanylyl cyclase (SGC) is a hemoprotein that plays key roles in various physiological functions. SGC is a typical enzyme-linked receptor that combines the functions of a sensor for NO gas and cGMP generator. SGC possesses exclusive selectivity for NO and exhibits a very fast binding of NO, which allows it to function as a sensitive NO receptor. This review describes the effect of various cellular factors, such as additional NO, cell thiols, cell-derived small molecules and proteins on the function of SGC as cellular NO receptor. Due to its vital physiological function SGC is an important drug target. An increasing number of synthetic compounds that affect SGC activity via different mechanisms are discovered and brought to clinical trials and clinics. Cellular factors modifying the activity of SGC constitute an opportunity for improving the effectiveness of existing SGC-directed drugs and/or the creation of new therapeutic strategies.


Subject(s)
Guanylate Cyclase , Nitric Oxide , Soluble Guanylyl Cyclase/metabolism , Nitric Oxide/metabolism , Guanylate Cyclase/metabolism , Sulfhydryl Compounds
3.
Front Mol Biosci ; 9: 1007768, 2022.
Article in English | MEDLINE | ID: mdl-36304925

ABSTRACT

Nitric oxide (NO), carbon monoxide (CO), oxygen (O2), hydrogen sulfide (H2S) are gaseous molecules that play important roles in the physiology and pathophysiology of eukaryotes. Tissue concentrations of these physiologically relevant gases vary remarkable from nM range for NO to high µM range of O2. Various hemoproteins play a significant role in sensing and transducing cellular signals encoded by gaseous molecules or in transporting them. Soluble guanylyl cyclase (sGC) is a hemoprotein that plays vital roles in a wide range of physiological functions and combines the functions of gaseous sensor and signal transducer. sGC uniquely evolved to sense low non-toxic levels of NO and respond to elevated NO levels by increasing its catalytic ability to generate the secondary signaling messenger cyclic guanosine monophosphate (cGMP). This review discusses sGC's gaseous ligand selectivity and the molecular basis for sGC function as high-affinity and selectivity NO receptor. The effects of other gaseous molecules and small molecules of cellular origin on sGC's function are also discussed.

4.
J Pharmacol Exp Ther ; 381(2): 164-175, 2022 05.
Article in English | MEDLINE | ID: mdl-35197320

ABSTRACT

12-lipoxigenase (12-LOX) is implicated in regulation of platelet activation processes and can be a new promising target for antiplatelet therapy. However, investigations of 12-LOX were restricted by the lack of specific and potent 12-LOX inhibitors and by controversial data concerning the role of 12-LOX metabolites in platelet functions. A novel specific 12-LOX inhibitor ML355 was shown to inhibit platelet aggregation without adverse side effects on hemostasis; however, the molecular mechanisms of its action on platelets are poorly understood. Here, we showed that ML355 inhibited platelet activation induced by thrombin or thromboxane A2, but not by collagen-related peptide. ML355 blocked protein kinase B, phosphoinositide 3-kinase, and extracellular signal-regulated kinase, but not p38 kinase, spleen tyrosine kinase (Syk), or phospholipase Cγ2 phosphorylation in activated platelets. The main inhibitory effect of low doses of ML355 (1-20 µM) on thrombin activated platelets was mediated by the decrease in reactive oxygen species level, whereas high doses of ML355 (50 µM) caused cyclic adenosine monophosphate activation. ML355 did not affect the activity of nitric oxide-dependent soluble guanylyl cyclase, nor did it affect the relaxation of preconstricted aortic rings in mice. ML355 itself did not affect platelet viability, but at 50 µM dose blocked caspase-dependent apoptosis induced by B-cell lymphoma II inhibitor ABT-737. SIGNIFICANCE STATEMENT: The current paper provides novel and original data concerning molecular mechanisms of 12-LOX inhibitor ML355 action on platelets. These data reveal antiplatelet and protective effects of ML355 on platelets and may be of importance for both antiplatelet and anticancer therapy.


Subject(s)
Blood Platelets , Thrombin , Animals , Apoptosis , Biphenyl Compounds , Mice , Nitrophenols , Phosphatidylinositol 3-Kinases/metabolism , Piperazines , Platelet Activation , Platelet Aggregation , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Sulfonamides , Thrombin/metabolism
5.
Am J Physiol Heart Circ Physiol ; 322(3): H417-H426, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35089807

ABSTRACT

Nitric oxide (NO) binds soluble guanylyl cyclase ß (sGCß) to produce cGMP and relax vascular smooth muscle cells (SMCs) needed for vasodilation. Although the regulation of NO-stimulated sGC activity has been well characterized at the posttranslational level, the mechanisms that govern sGC transcription remain incompletely understood. Recently, we identified Forkhead box subclass O (FoxO) transcription factors as essential for expression of sGC; however, the specific FoxO family member responsible for the expression of sGCß in SMC remains unknown. Using FoxO shRNA knockdown adenovirus treatment in rat aortic SMCs, we show that FoxO1 or FoxO3 knockdown causes greater than twofold increases in Gucy1a3 and Gucy1b3 mRNA expression, without changes in NO-dependent cGMP production or cGMP-dependent phosphorylation. FoxO4 knockdown produced a 50% decrease in Gucy1a3 and Gucy1b3 mRNA with 70% loss of sGCα and 50% loss of sGCß protein expression. Knockdown of FoxO4 expression decreased cGMP production and downstream protein kinase G-dependent phosphorylation more than 50%. Triple FoxO knockdown exacerbated loss of sGC-dependent function, phenocopying previous FoxO inhibition studies. Using promoter luciferase and chromatin immunoprecipitation assays, we find that FoxO4 acts as a transcriptional activator by directly binding several FoxO DNA motifs in the promoter regions of GUCY1B3 in human aortic SMCs. Collectively, our data show FoxO4 is a critical transcriptional regulator of sGCß expression in SMC.NEW & NOTEWORTHY One of the key mechanisms of vascular smooth muscle cell (SMC) dilation occurs through nitric oxide (NO)-dependent induction of soluble guanylyl cyclase (sGC) by means of its ß-subunit. Herein, we are the first to identify Forkhead box subclass O protein 4 (FoxO4) as a key transcriptional regulator of GUCY1B3 expression, which codes for sGCß protein in human and animal SMCs. This discovery will likely have important implications for the future usage of antihypertensive and vasodilatory therapies which target NO production, sGC, or FoxO transcription factors.


Subject(s)
Forkhead Transcription Factors/metabolism , Muscle, Smooth, Vascular/metabolism , Soluble Guanylyl Cyclase/genetics , Animals , Aorta/cytology , Cells, Cultured , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Rats , Soluble Guanylyl Cyclase/metabolism
6.
Eur J Med Chem ; 224: 113729, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34365128

ABSTRACT

Previous studies demonstrated that anti-hyperlipidemic drug gemfibrozil acts as NO- and heme-independent activator of NO receptor soluble guanylyl cyclase. A series of new gemfibrozil derivatives were synthesized and evaluated for sGC activation. The structure-activity relationship study identified the positions in gemfibrozil's scaffold that are detrimental for sGC activation and those that are amendable for optimizing modifications. Compared with gemfibrozil, compounds 7c and 15b were more potent activators of cGMP-forming activity of purified sGC and exhibited enhanced relaxation of preconstricted mouse thoracic aorta rings. These studies established the overall framework needed for futher improvement of sGC activators based on gemfibrozil scaffold.


Subject(s)
Gemfibrozil/therapeutic use , Nitric Oxide/metabolism , Soluble Guanylyl Cyclase/drug effects , Animals , Gemfibrozil/pharmacology , Humans , Mice , Structure-Activity Relationship
7.
Br J Pharmacol ; 178(18): 3765-3782, 2021 09.
Article in English | MEDLINE | ID: mdl-33931865

ABSTRACT

BACKGROUND AND PURPOSE: l-cysteine or hydrogen sulfide (H2 S) donors induce a biphasic effect on precontracted isolated vessels. The contractile effect occurs within a concentration range of 10 nM to 3 µM followed by vasodilatation at 30-100 µM. Here, we have investigated the signalling involved in the H2 S-induced contraction. EXPERIMENTAL APPROACH: Vascular response to NaHS or l-cysteine is evaluated on isolated precontracted with phenylephrine vessel rings harvested from wild type, cystathionine γ-lyase (CSE-/- ), soluble guanylyl cyclase (sGCα1-/- ) and endothelial nitric oxide synthase (eNOS-/- ) knock-out mice. The cAMP, cGMP and inosine 3',5'-cyclic monophosphate (cIMP) levels are simultaneously quantified using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) analysis. The involvement of sGC, phosphodiesterase (PDE) 4A and PDE5 are also evaluated. KEY RESULTS: CSE-derived H2 S-induced contraction requires an intact eNOS/NO/sGC pathway and involves cIMP as a second messenger. H2 S contractile effect involves a transient increase of cGMP and cAMP metabolism caused by PDE5 and PDE4A, thus unmasking cIMP contracting action. The stable cell-permeable analogue of cIMP elicits concentration-dependent contraction on a stable background tone induced by phenylephrine. The lack of cIMP, coupled to the hypocontractility displayed by vessels harvested from CSE-/- mice, confirms that H2 S-induced contraction involves cIMP. CONCLUSION AND IMPLICATIONS: The endothelium dynamically regulates vessel homeostasis by modulating contractile tone. This also involves CSE-derived H2 S that is mediated by cIMP.


Subject(s)
Cystathionine gamma-Lyase , Hydrogen Sulfide , Animals , Chromatography, Liquid , Cyclic GMP , Inosine Monophosphate , Mice , Nitric Oxide , Tandem Mass Spectrometry
8.
Biochem Pharmacol ; 186: 114459, 2021 04.
Article in English | MEDLINE | ID: mdl-33571505

ABSTRACT

NO sensitive soluble guanylyl cyclase (sGC) plays a key role in mediating physiological functions of NO. Genetic alterations of the GUCY1A3 gene, coding for the α1 subunit of sGC, are associated with several cardiovascular dysfunctions. A rare sGC variant with Cys517 â†’ Tyr substitution in the α1subunit, has been associated with moyamoya disease and achalasia. In this report we characterize the properties of this rare sGC variant. Purified α1C517Yß1 sGC preserved only ~25% of its cGMP-forming activity and showed an elevated Km for GTP substrate. However, the mutant enzyme retained a high affinity for and robust activation by NO, similar to wild type sGC. Purified α1C517Yß1 enzyme was more sensitive to specific sGC heme oxidizers and less responsive to heme reducing agents. When expressed in COS7 cells, α1C517Yß1 sGC showed a much stronger response to cinaciguat or gemfibrozil, which targets apo-sGC or sGC with ferric heme, as compared to its NO response or the relative response of the wild type sGC. A stronger response to cinaciguat was also observed for purified α1C517Yß1 in the absence of reducing agents. In COS7 cells, αCys517ß sGC was less stable than the wild type enzyme under normal conditions and exhibited accelerated degradation upon induction of cellular oxidative stress. We conclude that diminished cGMP-forming activity of this sGC variant is aggravated by its high susceptibility to oxidative stress and diminished protein stability. The combination of these deficiencies contributes to the severity of observed moyamoya and achalasia symptoms in human carriers of this rare α1C517Yß1 sGC variant.


Subject(s)
Genetic Variation/physiology , Heme/metabolism , Moyamoya Disease/genetics , Moyamoya Disease/metabolism , Soluble Guanylyl Cyclase/genetics , Soluble Guanylyl Cyclase/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Genetic Variation/drug effects , Humans , Oxadiazoles/pharmacology , Oxazines/pharmacology , Oxidation-Reduction/drug effects , Protein Stability/drug effects , Protein Structure, Secondary , Sf9 Cells
9.
Sci Rep ; 10(1): 17469, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060787

ABSTRACT

Cardiovascular diseases are the number one death worldwide. Nitric oxide (NO)-NO-sensitive (soluble) guanylyl cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway regulates diverse set of important physiological functions, including maintenance of cardiovascular homeostasis. Resting and activated sGC enzyme converts guanosine triphosphate to an important second messenger cGMP. In addition to traditional NO generators, a number of sGC activators and stimulators are currently in clinical trials aiming to support or increase sGC activity in various pathological conditions. cGMP-specific phosphodiesterases (PDEs), which degrade cGMP to guanosine monophosphate, play key role in controlling the cGMP level and the strength or length of the cGMP-dependent cellular signaling. Thus, PDE inhibitors also have clear clinical applications. Here, we introduce a homogeneous quenching resonance energy transfer (QRET) for cGMP to monitor both sGC and PDE activities using high throughput screening adoptable method. We demonstrate that using cGMP-specific antibody, sGC or PDE activity and the effect of small molecules modulating their function can be studied with sub-picomole cGMP sensitivity. The results further indicate that the method is suitable for monitoring enzyme reactions also in complex biological cellular homogenates and mixture.


Subject(s)
Cyclic GMP/metabolism , Nitric Oxide/metabolism , Phosphoric Diester Hydrolases/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , COS Cells , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Chlorocebus aethiops , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Enzyme Activators/therapeutic use , Homeostasis , Humans , Kinetics , Mice , Signal Transduction , Spectrometry, Fluorescence
10.
Article in English | MEDLINE | ID: mdl-30881924

ABSTRACT

The oral microbiome has the potential to provide an important symbiotic function in human blood pressure physiology by contributing to the generation of nitric oxide (NO), an essential cardiovascular signaling molecule. NO is produced by the human body via conversion of arginine to NO by endogenous nitric oxide synthase (eNOS) but eNOS activity varies by subject. Oral microbial communities are proposed to supplement host NO production by reducing dietary nitrate to nitrite via bacterial nitrate reductases. Unreduced dietary nitrate is delivered to the oral cavity in saliva, a physiological process termed the enterosalivary circulation of nitrate. Previous studies demonstrated that disruption of enterosalivary circulation via use of oral antiseptics resulted in increases in systolic blood pressure. These previous studies did not include detailed information on the oral health of enrolled subjects. Using 16S rRNA gene sequencing and analysis, we determined whether introduction of chlorhexidine antiseptic mouthwash for 1 week was associated with changes in tongue bacterial communities and resting systolic blood pressure in healthy normotensive individuals with documented oral hygiene behaviors and free of oral disease. Tongue cleaning frequency was a predictor of chlorhexidine-induced changes in systolic blood pressure and tongue microbiome composition. Twice-daily chlorhexidine usage was associated with a significant increase in systolic blood pressure after 1 week of use and recovery from use resulted in an enrichment in nitrate-reducing bacteria on the tongue. Individuals with relatively high levels of bacterial nitrite reductases had lower resting systolic blood pressure. These results further support the concept of a symbiotic oral microbiome contributing to human health via the enterosalivary nitrate-nitrite-NO pathway. These data suggest that management of the tongue microbiome by regular cleaning together with adequate dietary intake of nitrate provide an opportunity for the improvement of resting systolic blood pressure.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Chlorhexidine/administration & dosage , Microbiota/drug effects , Nitrates/metabolism , Tongue/microbiology , Blood Pressure/drug effects , Cluster Analysis , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Healthy Volunteers , Humans , Mouthwashes/administration & dosage , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
11.
Antioxid Redox Signal ; 26(3): 122-136, 2017 01 20.
Article in English | MEDLINE | ID: mdl-26972233

ABSTRACT

SIGNIFICANCE: Nitric oxide (NO)-dependent signaling is critical to many cellular functions and physiological processes. Soluble guanylyl cyclase (sGC) acts as an NO receptor and mediates the majority of NO functions. The signaling between NO and sGC is strongly altered by reactive oxygen and nitrogen species. Recent Advances: Besides NO scavenging, sGC is affected by oxidation/loss of sGC heme, oxidation, or nitrosation of cysteine residues and phosphorylation. Apo-sGC or sGC containing oxidized heme is targeted for degradation. sGC transcription and the stability of sGC mRNA are also affected by oxidative stress. CRITICAL ISSUES: Studies cited in this review suggest the existence of compensatory processes that adapt cellular processes to diminished sGC function under conditions of short-term or moderate oxidative stress. Alternative splicing of sGC transcripts is discussed as a mechanism with the potential to both enhance and reduce sGC function. The expression of α1 isoform B, a functional and stable splice variant of human α1 sGC subunit, is proposed as one of such compensatory mechanisms. The expression of dysfunctional splice isoforms is discussed as a contributor to decreased sGC function in vascular disease. FUTURE DIRECTIONS: Targeting the process of sGC splicing may be an important approach to maintain the composition of sGC transcripts that are expressed in healthy tissues under normal conditions. Emerging new strategies that allow for targeted manipulations of RNA splicing offer opportunities to use this approach as a preventive measure and to control the composition of sGC splice isoforms. Rational management of expressed sGC splice forms may be a valuable complementary treatment strategy for existing sGC-directed therapies. Antioxid. Redox Signal. 26, 122-136.


Subject(s)
Gene Expression Regulation , RNA Splicing , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Soluble Guanylyl Cyclase/genetics , Soluble Guanylyl Cyclase/metabolism , Alternative Splicing , Animals , Humans , Hydrogen Peroxide/metabolism , Nitrogen/metabolism , Oxidation-Reduction , Oxidative Stress , Oxygen/metabolism , Signal Transduction
12.
Cell Commun Signal ; 14(1): 16, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27515066

ABSTRACT

BACKGROUND: Direct interaction between Red blood cells (RBCs) and platelets is known for a long time. The bleeding time is prolonged in anemic patients independent of their platelet count and could be corrected by transfusion of RBCs, which indicates that RBCs play an important role in hemostasis and platelet activation. However, in the last few years, opposing mechanisms of platelet inhibition by RBCs derived nitric oxide (NO) were proposed. The aim of our study was to identify whether RBCs could produce NO and activate soluble guanylate cyclase (sGC) in platelets. METHODS: To test whether RBCs could activate sGC under different conditions (whole blood, under hypoxia, or even loaded with NO), we used our well-established and highly sensitive models of NO-dependent sGC activation in platelets and activation of purified sGC. The activation of sGC was monitored by detecting the phosphorylation of Vasodilator Stimulated Phosphoprotein (VASP(S239)) by flow cytometry and Western blot. ANOVA followed by Bonferroni's test and Student's t-test were used as appropriate. RESULTS: We show that in the whole blood, RBCs prevent NO-mediated inhibition of ADP and TRAP6-induced platelet activation. Likewise, coincubation of RBCs with platelets results in strong inhibition of NO-induced sGC activation. Under hypoxic conditions, incubation of RBCs with NO donor leads to Hb-NO formation which inhibits sGC activation in platelets. Similarly, RBCs inhibit activation of purified sGC, even under conditions optimal for RBC-mediated generation of NO from nitrite. CONCLUSIONS: All our experiments demonstrate that RBCs act as strong NO scavengers and prevent NO-mediated inhibition of activated platelets. In all tested conditions, RBCs were not able to activate platelet or purified sGC.


Subject(s)
Blood Platelets/metabolism , Erythrocytes/metabolism , Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Blood Platelets/enzymology , Humans
13.
Pharmacol Res ; 111: 556-562, 2016 09.
Article in English | MEDLINE | ID: mdl-27378567

ABSTRACT

Soluble guanylate cyclase (sGC) is a receptor for nitric oxide (NO). Binding of NO to ferrous (Fe(2+)) heme increases its catalytic activity, leading to the production of cGMP from GTP. Hydrogen sulfide (H2S) is a signaling molecule that exerts both direct and indirect anti-oxidant effects. In the present, study we aimed to determine whether H2S could regulate sGC redox state and affect its responsiveness to NO-releasing agents and sGC activators. Using cultured rat aortic smooth muscle cells, we observed that treatment with H2S augmented the response to the NO donor DEA/NO, while attenuating the response to the heme-independent activator BAY58-2667 that targets oxidized sGC. Similarly, overexpression of H2S-synthesizing enzyme cystathionine-γ lyase reduced the ability of BAY58-2667 to promote cGMP accumulation. In experiments with phenylephrine-constricted mouse aortic rings, treatment with rotenone (a compound that increases ROS production), caused a rightward shift of the DEA/NO concentration-response curve, an effect partially restored by H2S. When rings were pre-treated with H2S, the concentration-response curve to BAY 58-2667 shifted to the right. Using purified recombinant human sGC, we observed that treatment with H2S converted ferric to ferrous sGC enhancing NO-donor-stimulated sGC activity and reducing BAY 58-2667-triggered cGMP formation. The present study identified an additional mechanism of cross-talk between the NO and H2S pathways at the level of redox regulation of sGC. Our results provide evidence that H2S reduces sGC heme Fe, thus, facilitating NO-mediated cellular signaling events.


Subject(s)
Heme/metabolism , Hydrogen Sulfide/pharmacology , Nitric Oxide/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Benzoates/pharmacology , Cells, Cultured , Cystathionine gamma-Lyase/metabolism , In Vitro Techniques , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Donors/pharmacology , Oxidation-Reduction , Phenylephrine , Quaternary Ammonium Compounds/pharmacology , Rats
14.
Am J Physiol Heart Circ Physiol ; 307(11): H1565-75, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25239802

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 ß1 sGC in healthy and diseased human vascular tissue. Our study found a variety of α1 and ß1 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.


Subject(s)
Alternative Splicing/genetics , Alternative Splicing/physiology , Aortic Aneurysm/enzymology , Aortic Aneurysm/genetics , Guanylate Cyclase/genetics , Guanylate Cyclase/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Adult , Aged , Aged, 80 and over , Animals , COS Cells , Cloning, Molecular , Cyclic GMP/physiology , Female , Genotype , Humans , Male , Middle Aged , Nitric Oxide/physiology , Precision Medicine , Signal Transduction/genetics , Signal Transduction/physiology , Soluble Guanylyl Cyclase , Young Adult
15.
ChemMedChem ; 9(10): 2344-50, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25044578

ABSTRACT

Specially designed B-ring-modified cobalamin derivatives were synthesized and tested as potential activators of soluble guanylyl cyclase (sGC). Herein, we disclose the influence of substituents at the c- and d-positions in hydrophilic and hydrophobic cobyrinic acid derivatives on their capacities to activate sGC. The presence of the amide group at c-/d-position in cobyrinic acid derivatives strongly influence the level of sGC activation. Removal of the d-position altogether has a profound effect for hydrophobic compounds. In contrast, little differences were observed in hydrophilic ones.


Subject(s)
Cobamides/chemistry , Guanylate Cyclase/metabolism , Enzyme Activation , Molecular Structure
16.
J Med Chem ; 56(18): 7260-77, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23961771

ABSTRACT

Hybrid molecules composed of PpIX and cobyrinic acid derivatives conjugated through linkers of varying length and composition were prepared via 1,3-dipolar cycloaddition (CuAAC) or amidation/esteryfication reactions. They were tested for activation of soluble guanylyl cyclase (sGC), a key enzyme in the NO/cGMP signaling pathway, by an in vitro GTP→cGMP conversion assay. Using purified heme-deficient sGC and truncated sGC variants lacking a heme-binding domain, we demonstrated that such hybrid molecules may activate sGC by targeting heme-binding and/or catalytic domain. While all conjugates activated sGC, only selected compounds served as bifunctional regulators and were capable of simultaneous targeting both heme and catalytic domains of sGC. The length and type of a linker connecting both components had a profound effect on the extent of sGC activation, indicating that the linker's type is crucial for their binding affinities with regulatory and catalytic domains. Only hybrids with the conjugated linker of 13-16 atom length synergistically target both domains and displayed the lowest EC50 and highest activating potency. Compounds with shorter connecting linkers were much less potent and were no more active than the cobyrinic acid component alone. The most active conjugate, which showed a 60-fold activation of sGC, was compound 11, in which PpIX and cobyrinic acid components are separated by 11 atoms chain with the triazole moiety in between.


Subject(s)
Guanylate Cyclase/metabolism , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Chemistry Techniques, Synthetic , Copper/chemistry , Enzyme Activation/drug effects , Guanylate Cyclase/chemistry , Humans , Models, Molecular , Organometallic Compounds/chemistry , Protein Conformation , Protoporphyrins/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Soluble Guanylyl Cyclase , Structure-Activity Relationship
17.
PLoS One ; 7(7): e41099, 2012.
Article in English | MEDLINE | ID: mdl-22911749

ABSTRACT

BACKGROUND: Soluble guanylyl cyclase (sGC) plays a central role in nitric oxide (NO)-mediated signal transduction in the cardiovascular, nervous and gastrointestinal systems. Alternative RNA splicing has emerged as a potential mechanism to modulate sGC expression and activity. C-α1 sGC is an alternative splice form that is resistant to oxidation-induced protein degradation and demonstrates preferential subcellular distribution to the oxidized environment of endoplasmic reticulum (ER). METHODOLOGY/PRINCIPAL FINDINGS: Here we report that splicing of C-α1 sGC can be modulated by H(2)O(2) treatment in BE2 neuroblastoma and MDA-MD-468 adenocarcinoma human cells. In addition, we show that the H(2)O(2) treatment of MDA-MD-468 cells selectively decreases protein levels of PTBP1 and hnRNP A2/B1 splice factors identified as potential α1 gene splicing regulators by in silico analysis. We further demonstrate that down-regulation of PTBP1 by H(2)O(2) occurs at the protein level with variable regulation observed in different breast cancer cells. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate that H(2)O(2) regulates RNA splicing to induce expression of the oxidation-resistant C-α1 sGC subunit. We also report that H(2)O(2) treatment selectively alters the expression of key splicing regulators. This process might play an important role in regulation of cellular adaptation to conditions of oxidative stress.


Subject(s)
Alternative Splicing/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Guanylate Cyclase/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Hydrogen Peroxide/pharmacology , Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Cell Line, Tumor , Gene Expression Profiling , Guanylate Cyclase/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Hydrogen Peroxide/toxicity , Neoplasms/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Proteolysis/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase
18.
Biochem J ; 446(3): 445-53, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22690686

ABSTRACT

sGC (soluble guanylate cyclase) is the main mediator of NO signalling. Biochemical and physiological studies suggest that, besides NO, in vivo regulation of sGC involves direct interaction with other proteins. Using yeast two-hybrid screening, we identified that the multidomain LGN (Leu-Gly-Asn repeat-enriched protein) interacts with both α1 and ß1 sGC subunits. LGN and sGC co-localized in the cell cytoplasm, and the LGN-sGC complex was co-immunoprecipitated from cells expressing both proteins and from native tissues. Their interaction requires the N-terminal tetratricopeptide repeats of LGN, but does not require the N-terminal portions of α1 or ß1 sGC subunits. Overexpression of LGN decreases the activity of cellular sGC, whereas knockdown of LGN mRNA and protein correlated with increased sGC activity. Although purified LGN interacts directly with purified sGC, the inhibitory effect in vitro is observed only after supplementation of cell lysate to the reaction. Although resting sGC and sGC activated by the stimulator BAY41-2272 have very similar LGN-IC50 values to the NO-stimulated sGC, they have a much higher Hill coefficient, suggesting co-operative binding with respect to LGN in the low-activated state of sGC. AGS3 (activator of G-protein signalling 3), the closest LGN homologue, also inhibits sGC. The interaction of sGC with these scaffolding proteins may expand the cross-talk between NO/cGMP signalling and other cellular pathways and tailor sGC function to specific tissues or signals.


Subject(s)
Guanylate Cyclase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Cell Line, Tumor , Cyclic GMP/metabolism , Guanylate Cyclase/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase , Transfection
19.
Proc Natl Acad Sci U S A ; 109(23): 9161-6, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22570497

ABSTRACT

Hydrogen sulfide (H(2)S) is a unique gasotransmitter, with regulatory roles in the cardiovascular, nervous, and immune systems. Some of the vascular actions of H(2)S (stimulation of angiogenesis, relaxation of vascular smooth muscle) resemble those of nitric oxide (NO). Although it was generally assumed that H(2)S and NO exert their effects via separate pathways, the results of the current study show that H(2)S and NO are mutually required to elicit angiogenesis and vasodilatation. Exposure of endothelial cells to H(2)S increases intracellular cyclic guanosine 5'-monophosphate (cGMP) in a NO-dependent manner, and activated protein kinase G (PKG) and its downstream effector, the vasodilator-stimulated phosphoprotein (VASP). Inhibition of endothelial isoform of NO synthase (eNOS) or PKG-I abolishes the H(2)S-stimulated angiogenic response, and attenuated H(2)S-stimulated vasorelaxation, demonstrating the requirement of NO in vascular H(2)S signaling. Conversely, silencing of the H(2)S-producing enzyme cystathionine-γ-lyase abolishes NO-stimulated cGMP accumulation and angiogenesis and attenuates the acetylcholine-induced vasorelaxation, indicating a partial requirement of H(2)S in the vascular activity of NO. The actions of H(2)S and NO converge at cGMP; though H(2)S does not directly activate soluble guanylyl cyclase, it maintains a tonic inhibitory effect on PDE5, thereby delaying the degradation of cGMP. H(2)S also activates PI3K/Akt, and increases eNOS phosphorylation at its activating site S1177. The cooperative action of the two gasotransmitters on increasing and maintaining intracellular cGMP is essential for PKG activation and angiogenesis and vasorelaxation. H(2)S-induced wound healing and microvessel growth in matrigel plugs is suppressed by pharmacological inhibition or genetic ablation of eNOS. Thus, NO and H(2)S are mutually required for the physiological control of vascular function.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Hydrogen Sulfide/pharmacology , Neovascularization, Physiologic/physiology , Nitric Oxide/pharmacology , Vasodilation/physiology , Analysis of Variance , Animals , Blotting, Western , Cell Adhesion Molecules/metabolism , Cell Line , Collagen , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cystathionine gamma-Lyase/metabolism , Drug Combinations , Hydrogen Sulfide/metabolism , Laminin , Mice , Microfilament Proteins/metabolism , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteoglycans , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects , Wound Healing/drug effects
20.
Biochemistry ; 51(13): 2737-46, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22401134

ABSTRACT

Soluble guanylyl cyclase (sGC), the key enzyme for the formation of second messenger cyclic GMP, is an authentic sensor for nitric oxide (NO). Binding of NO to sGC leads to strong activation of the enzyme activity. Multiple molecules and steps of binding of NO to sGC have been implicated, but the target of the second NO and the detailed binding mechanism remain controversial. In this study, we used (15)NO and (14)NO and anaerobic sequential mixing-freeze-quench electron paramagnetic resonance to unambiguously confirm that the heme Fe is the target of the second NO. The linear dependence on NO concentration up to 600 s(-1) for the observed rate of the second step of NO binding not only indicates that the binding site of the second NO is different from that in the first step, i.e., the proximal site of the heme, but also supports a concerted mechanism in which the dissociation of the His105 proximal ligand occurs simultaneously with the binding of the second NO molecule. Computer modeling successfully predicts the kinetics of formation of a set of five-coordinate NO complexes with the ligand on either the distal or proximal site and supports the selective release of NO from the distal side of the transient bis-NO-sGC complex. Thus, as has been demonstrated with cytochrome c', a five-coordinate NO-sGC complex containing a proximal NO is formed after the binding of the second NO.


Subject(s)
Guanylate Cyclase/metabolism , Heme/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Binding Sites , Cell Line , Computer Simulation , Electron Spin Resonance Spectroscopy , Enzyme Activation , Kinetics , Soluble Guanylyl Cyclase , Spodoptera
SELECTION OF CITATIONS
SEARCH DETAIL
...