1
|
Pliushcheuskaya P, Kesh S, Kaufmann E, Wucherpfennig S, Schwede F, Künze G, Nache V. Similar Binding Modes of cGMP Analogues Limit Selectivity in Modulating Retinal CNG Channels via the Cyclic Nucleotide-Binding Domain. ACS Chem Neurosci 2024; 15:1652-1668. [PMID: 38579109 PMCID: PMC11027099 DOI: 10.1021/acschemneuro.3c00665] [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: 10/16/2023] [Revised: 02/28/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
In treating retinitis pigmentosa, a genetic disorder causing progressive vision loss, selective inhibition of rod cyclic nucleotide-gated (CNG) channels holds promise. Blocking the increased Ca2+-influx in rod photoreceptors through CNG channels can potentially delay disease progression and improve the quality of life for patients. To find inhibitors for rod CNG channels, we investigated the impact of 16 cGMP analogues on both rod and cone CNG channels using the patch-clamp technique. Although modifications at the C8 position of the guanine ring did not change the ligand efficacy, modifications at the N1 and N2 positions rendered cGMP largely ineffective in activating retinal CNG channels. Notably, PET-cGMP displayed selective potential, favoring rod over cone, whereas Rp-cGMPS showed greater efficiency in activating cone over rod CNG channels. Ligand docking and molecular dynamics simulations on cyclic nucleotide-binding domains showed comparable binding energies and binding modes for cGMP and its analogues in both rod and cone CNG channels (CNGA1 vs CNGA3 subunits). Computational experiments on CNGB1a vs CNGB3 subunits showed similar binding modes albeit with fewer amino acid interactions with cGMP due to an inactivated conformation of their C-helix. In addition, no clear correlation could be observed between the computational scores and the CNG channel efficacy values, suggesting additional factors beyond binding strength determining ligand selectivity and potency. This study highlights the importance of looking beyond the cyclic nucleotide-binding domain and toward the gating mechanism when searching for selective modulators. Future efforts in developing selective modulators for CNG channels should prioritize targeting alternative channel domains.
Collapse
Affiliation(s)
- Palina Pliushcheuskaya
- Institute
for Drug Discovery, Medical Faculty, University
of Leipzig, Leipzig 04103, Germany
| | - Sandeep Kesh
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| | - Emma Kaufmann
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| | - Sophie Wucherpfennig
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| | - Frank Schwede
- BIOLOG
Life Science Institute GmbH & Co KG, Bremen 28199, Germany
| | - Georg Künze
- Institute
for Drug Discovery, Medical Faculty, University
of Leipzig, Leipzig 04103, Germany
- Interdisciplinary
Center for Bioinformatics, University of
Leipzig, Leipzig 04107, Germany
- Center
for Scalable Data Analytics and Artificial Intelligence, University of Leipzig, Leipzig 04105, Germany
| | - Vasilica Nache
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| |
Collapse
|
2
|
Erro R, Mencacci NE, Bhatia KP. The Emerging Role of Phosphodiesterases in Movement Disorders. Mov Disord 2021; 36:2225-2243. [PMID: 34155691 PMCID: PMC8596847 DOI: 10.1002/mds.28686] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Cyclic nucleotide phosphodiesterase (PDE) enzymes catalyze the hydrolysis and inactivation of the cyclic nucleotides cyclic adenosine monophosphate and cyclic guanosine monophosphate, which act as intracellular second messengers for many signal transduction pathways in the central nervous system. Several classes of PDE enzymes with specific tissue distributions and cyclic nucleotide selectivity are highly expressed in brain regions involved in cognitive and motor functions, which are known to be implicated in neurodegenerative diseases, such as Parkinson's disease and Huntington's disease. The indication that PDEs are intimately involved in the pathophysiology of different movement disorders further stems from recent discoveries that mutations in genes encoding different PDEs, including PDE2A, PDE8B, and PDE10A, are responsible for rare forms of monogenic parkinsonism and chorea. We here aim to provide a translational overview of the preclinical and clinical data on PDEs, the role of which is emerging in the field of movement disorders, offering a novel venue for a better understanding of their pathophysiology. Modulating cyclic nucleotide signaling, by either acting on their synthesis or on their degradation, represents a promising area for development of novel therapeutic approaches. The study of PDE mutations linked to monogenic movement disorders offers the opportunity of better understanding the role of PDEs in disease pathogenesis, a necessary step to successfully benefit the treatment of both hyperkinetic and hypokinetic movement disorders. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Niccoló E Mencacci
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
3
|
Susmi TF, Rahman A, Khan MMR, Yasmin F, Islam MS, Nasif O, Alharbi SA, Batiha GES, Hossain MU. Prognostic and clinicopathological insights of phosphodiesterase 9A gene as novel biomarker in human colorectal cancer. BMC Cancer 2021; 21:577. [PMID: 34016083 PMCID: PMC8136133 DOI: 10.1186/s12885-021-08332-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND PDE9A (Phosphodiesterase 9A) plays an important role in proliferation of cells, their differentiation and apoptosis via intracellular cGMP (cyclic guanosine monophosphate) signaling. The expression pattern of PDE9A is associated with diverse tumors and carcinomas. Therefore, PDE9A could be a prospective candidate as a therapeutic target in different types of carcinoma. The study presented here was designed to carry out the prognostic value as a biomarker of PDE9A in Colorectal cancer (CRC). The present study integrated several cancer databases with in-silico techniques to evaluate the cancer prognosis of CRC. RESULTS The analyses suggested that the expression of PDE9A was significantly down-regulated in CRC tissues than in normal tissues. Moreover, methylation in the DNA promoter region might also manipulate PDE9A gene expression. The Kaplan-Meier curves indicated that high level of expression of PDE9A gene was associated to higher survival in OS, RFS, and DSS in CRC patients. PDE9A demonstrated the highest positive correlation for rectal cancer recurrence with a marker gene CEACAM7. Furtheremore, PDE9A shared consolidated pathways with MAPK14 to induce survival autophagy in CRC cells and showed interaction with GUCY1A2 to drive CRPC. CONCLUSIONS Overall, the prognostic value of PDE9A gene could be used as a potential tumor biomarker for CRC.
Collapse
Affiliation(s)
- Tasmina Ferdous Susmi
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Atikur Rahman
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
- Department of Fermentation Engineering, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Md. Moshiur Rahman Khan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Farzana Yasmin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Md. Shariful Islam
- Department of Reproductive and Developmental Biology, Graduate School of Life Science, Hokkaido University, Sapporo, 5 Chome Kita 8 Jonishi, Kita Ward, Sapporo, Hokkaido 060-0808 Japan
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506-022 USA
| | - Omaima Nasif
- Department of Physiology, College of Medicine, King Saud University [Medical City], King Khalid University Hospital, PO Box 2925, Riyadh, 11461 Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany & Microbiology, College of Science, King Saud University, P.O Box 2455, Riyadh, 11451 Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511 Egypt
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349 Bangladesh
| |
Collapse
|
4
|
Pal S, Rashid M, Singh SK, Porwal K, Singh P, Mohamed R, Gayen JR, Wahajuddin M, Chattopadhyay N. Skeletal restoration by phosphodiesterase 5 inhibitors in osteopenic mice: Evidence of osteoanabolic and osteoangiogenic effects of the drugs. Bone 2020; 135:115305. [PMID: 32126313 DOI: 10.1016/j.bone.2020.115305] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/12/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Phosphodiesterases (PDEs) hydrolyze cyclic nucleotides and thereby regulate diverse cellular functions. The reports on the skeletal effects of PDE inhibitors are conflicting. Here, we screened 17 clinically used non-xanthine PDE inhibitors (selective and non-selective) using mouse calvarial osteoblasts (MCO) where the readout was osteoblast differentiation. From this screen, we identified sildenafil and vardenafil (both PDE5 inhibitors) having the least osteogenic EC50. Both drugs significantly increased vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) expressions in MCO and the nitric oxide synthase inhibitor L-NAME completely blocked VEGF expression induced by these drugs. Sunitinib, a tyrosine receptor kinase inhibitor that also blocks VEGFR2 blocked sildenafil-/vardenafil-induced osteoblast differentiation. At half of their human equivalent doses, i.e. 6.0 mg/kg sildenafil and 2.5 mg/kg vardenafil, the maximum bone marrow level of sildenafil was 32% and vardenafil was 21% of their blood levels. At these doses, both drugs enhanced bone regeneration at the femur osteotomy site and completely restored bone mass, microarchitecture, and strength in OVX mice. Furthermore, both drugs increased surface referent bone formation and serum bone formation marker (P1NP) without affecting the resorption marker (CTX-1). Both drugs increased the expression of VEGF and VEGFR2 in bones and osteoblasts and increased skeletal vascularity. Sunitinib completely blocked the bone restorative and vascular effects of sildenafil and vardenafil in OVX mice. Taken together, our study suggested that sildenafil and vardenafil at half of their adult human doses completely reversed osteopenia in OVX mice by an osteogenic mechanism that was associated with enhanced skeletal vascularity.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Mamunur Rashid
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | | | - Konica Porwal
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Priya Singh
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Riyazuddin Mohamed
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India.
| |
Collapse
|
5
|
Reverte-Salisa L, Sanyal A, Pfeifer A. Role of cAMP and cGMP Signaling in Brown Fat. Handb Exp Pharmacol 2019; 251:161-182. [PMID: 29633180 DOI: 10.1007/164_2018_117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cold-induced activation of brown adipose tissue (BAT) is mediated by norepinephrine and adenosine that are released during sympathetic nerve activation. Both signaling molecules induce an increase in intracellular levels of 3',5'-cyclic adenosine monophosphate (cAMP) in murine and human BAT. In brown adipocytes, cAMP plays a central role, because it activates lipolysis, glucose uptake, and thermogenesis. Another well-studied intracellular second messenger is 3',5'-cyclic guanosine monophosphate (cGMP), which closely resembles cAMP. Several studies have shown that intact cGMP signaling is essential for normal adipogenic differentiation and BAT-mediated thermogenesis in mice. This chapter highlights recent observations, demonstrating the physiological significance of cyclic nucleotide signaling in BAT as well as their potential to induce browning of white adipose tissue (WAT) in mice and humans.
Collapse
Affiliation(s)
- Laia Reverte-Salisa
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Abhishek Sanyal
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
6
|
Pan Y, Rong Y, You M, Ma Q, Chen M, Hu F. Royal jelly causes hypotension and vasodilation induced by increasing nitric oxide production. Food Sci Nutr 2019; 7:1361-1370. [PMID: 31024709 PMCID: PMC6475742 DOI: 10.1002/fsn3.970] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/21/2019] [Accepted: 01/25/2019] [Indexed: 12/26/2022] Open
Abstract
Among royal jelly's (RJ) various biological activities, its possible antihypertension and vasorelaxation effects deserve particular attention, but the underlying mechanisms of action remain unclear. Therefore, this study used the spontaneously hypertensive rats (SHR) hypertension model and the isolated rabbit thoracic aorta rings model to explore the mechanisms underlying the hypotension and vasorelaxation effects of RJ. Rats were divided into the following groups (n = 6): WKY-control group, SHR-control group, and SHR-RJ group. SHR-RJ group was received 1 g/kg of RJ via oral administration daily for 4 weeks. Systolic blood pressure (SBP), diastolic blood pressure (DBP), heart rate (HR), and nitric oxide (NO) level were detected. In addition, the mechanism of vasodilation of RJ was investigated using an isolated rabbit aortic ring technique. RJ significantly reduced SBP and DBP as well as increased NO levels of SHR in vivo. RJ caused vasorelaxation of the isolated aorta rings, and this effect was inhibited by atropine (M3 receptor blocker), L-NAME (nitric oxide synthase inhibitor), methylene blue (guanylate cyclase inhibitor), and indomethacin (cyclooxygenase inhibitor). Moreover, RJ could markedly suppress the NE-induced intracellular Ca2+ releases and high K+-induced extracellular Ca2+ influx in denuded aortic rings. In addition, RJ can also increase cGMP levels and the production of NO in isolated aortic rings. The present study showed that RJ has antihypertensive effects and was associated with increased NO production. In addition, RJ contains muscarinic receptor agonist, possibly an acetylcholine-like substance, and induces vasodilation through NO/cGMP pathway and calcium channels.
Collapse
Affiliation(s)
- Yongming Pan
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Comparative Medical Research Institute, Experimental Animal Research CenterZhejiang Chinese Medical UniversityHangzhouChina
| | - Yili Rong
- Comparative Medical Research Institute, Experimental Animal Research CenterZhejiang Chinese Medical UniversityHangzhouChina
| | - Mengmeng You
- College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Quanxin Ma
- Comparative Medical Research Institute, Experimental Animal Research CenterZhejiang Chinese Medical UniversityHangzhouChina
| | - Minli Chen
- Comparative Medical Research Institute, Experimental Animal Research CenterZhejiang Chinese Medical UniversityHangzhouChina
| | - Fuliang Hu
- College of Animal SciencesZhejiang UniversityHangzhouChina
| |
Collapse
|
7
|
Veerappan R, Malarvili T. Chrysin Pretreatment Improves Angiotensin System, cGMP Concentration in L-NAME Induced Hypertensive Rats. Indian J Clin Biochem 2018; 34:288-295. [PMID: 31391718 PMCID: PMC6660528 DOI: 10.1007/s12291-018-0761-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 05/25/2018] [Indexed: 01/19/2023]
Abstract
Nω-nitro-l-arginine methyl ester (L-NAME) is a non-specific nitric oxide (NO) synthase inhibitor, commonly used for the induction of NO-deficient hypertension. The objective of the present study was to investigate the effects of chrysin with flavnoids, on L-NAME-induced hypertensive rats. Methods: An experimental hypertensive animal (180–220 g) model was induced by L-NAME intake on rats. In treatment chrysin was orally administered 25 mg/kg body weight (b.w.). Blood pressure was measured by tail cuff plethysmography system. Cardiac and vascular function was evaluated by Langendorff isolated heart system with Angiotensin II (Ang-II), Hexo oxygenase (HO-1), cyclic guanosine monophosphate (cGMP) concentration in tissues respectively. Rats with hypertension showed an elevated blood pressure (BP), left ventricular functions, ang II, and decreased cGMP concentration of tissues. Treatment of chrysin is reverse to near normal in left ventricular functions, Ang-II, Ho-1 and decreased cGMP concentration of tissues. The antihypertensive effect of chrysin appears to be mediated by a reduction in left ventricular functions, cardiac oxidative stress and Ang-II, an increase in cardiac HO-1, cGMP concentration and a prevention of plasma nitric oxide loss.
Collapse
Affiliation(s)
- Ramanathan Veerappan
- Research Department of Biochemistry, Enathi Rajappa Arts and Science College, Pattukkottai, Thanjavur, Tamilanadu India
| | | |
Collapse
|
8
|
Interactions between bradykinin and plasmin in the endothelial Ca2+ response. Mol Cell Biochem 2017; 445:179-186. [DOI: 10.1007/s11010-017-3263-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/23/2017] [Indexed: 10/18/2022]
|
9
|
Frigolet ME, Thomas G, Beard K, Lu H, Liu L, Fantus IG. The bradykinin-cGMP-PKG pathway augments insulin sensitivity via upregulation of MAPK phosphatase-5 and inhibition of JNK. Am J Physiol Endocrinol Metab 2017; 313:E321-E334. [PMID: 28679626 DOI: 10.1152/ajpendo.00298.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/13/2023]
Abstract
Bradykinin (BK) promotes insulin sensitivity and glucose uptake in adipocytes and other cell types. We demonstrated that in rat adipocytes BK enhances insulin-stimulated glucose transport via endothelial nitric oxide synthase, nitric oxide (NO) generation, and decreased activity of the mitogen-activated protein kinase (MAPK) JNK (c-Jun NH2-terminal kinase). In endothelial cells, NO increases soluble guanylate cyclase (sGC) activity, which, in turn, activates protein kinase G (PKG) by increasing cGMP levels. In this study, we investigated whether BK acts via the sGC-cGMP-PKG pathway to inhibit the negative effects of JNK on insulin signaling and glucose uptake in rat adipocytes. BK augmented cGMP concentrations. The BK-induced enhancement of insulin-stimulated glucose uptake was mimicked by the sGC activator YC-1 and a cell-permeable cGMP analog, CPT-cGMP, and inhibited by the sGC inhibitor ODQ and the PKG inhibitor KT 5823. Transfection of dominant-negative PKG reduced the BK augmentation of insulin-induced Akt phosphorylation. The activation of JNK and ERK1/2 by insulin was attenuated by BK, which was mediated by the sGC-cGMP-PKG pathway. Whereas insulin-stimulated phosphorylation of upstream activators of JNK and ERK, i.e., MKK4 and MEK1/2, was unaffected, BK augmented insulin-mediated induction of MKP-5 mRNA and protein levels. Furthermore, zaprinast, a phosphodiesterase inhibitor, enhanced cGMP and MKP-5 and prolonged the action of BK. These data indicate that BK enhances insulin action by inhibition of negative feedback by JNK and ERK via upregulation of MKP-5, mediated by the sGC-cGMP-PKG signaling pathway.
Collapse
Affiliation(s)
- María E Frigolet
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
| | - Garry Thomas
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kristin Beard
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Huogen Lu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lijiang Liu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - I George Fantus
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada;
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Dolgacheva LP, Turovskaya MV, Dynnik VV, Zinchenko VP, Goncharov NV, Davletov B, Turovsky EA. Angiotensin II activates different calcium signaling pathways in adipocytes. Arch Biochem Biophys 2016; 593:38-49. [PMID: 26850364 DOI: 10.1016/j.abb.2016.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/11/2016] [Accepted: 02/01/2016] [Indexed: 12/15/2022]
Abstract
Angiotensin II (Ang II) is an important mammalian neurohormone involved in reninangiotensin system. Ang II is produced both constitutively and locally by RAS systems, including white fat adipocytes. The influence of Ang II on adipocytes is complex, affecting different systems of signal transduction from early Са(2+) responses to cell proliferation and differentiation, triglyceride accumulation, expression of adipokine-encoding genes and adipokine secretion. It is known that white fat adipocytes express all RAS components and Ang II receptors (АТ1 and АТ2). The current work was carried out with the primary white adipocytes culture, and Са(2+) signaling pathways activated by Ang II were investigated using fluorescent microscopy. Са(2+)-oscillations and transient responses of differentiated adipocytes to Ang II were registered in cells with both small and multiple lipid inclusions. Using inhibitory analysis and selective antagonists, we now show that Ang II initiates periodic Са(2+)-oscillations and transient responses by activating АТ1 and АТ2 receptors and involving branched signaling cascades: 1) Ang II → Gq → PLC → IP3 → IP3Rs → Ca(2+) 2) Gβγ → PI3Kγ → PKB 3) PKB → eNOS → NO → PKG 4) CD38 → cADPR → RyRs → Ca(2+) In these cascades, AT1 receptors play the leading role. The results of the present work open a perspective of using Ang II for correction of signal resistance of adipocytes often observed during obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Lyudmila P Dolgacheva
- Laboratory of Intracellular Signalling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Maria V Turovskaya
- Laboratory of Intracellular Signalling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Vladimir V Dynnik
- Laboratory of Intracellular Signalling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia; Laboratory of System Biochemistry, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Valery P Zinchenko
- Laboratory of Intracellular Signalling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Nikolay V Goncharov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Bazbek Davletov
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, England, UK
| | - Egor A Turovsky
- Laboratory of Intracellular Signalling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia.
| |
Collapse
|
11
|
Börger C, Schünke S, Lecher J, Stoldt M, Winkhaus F, Kaupp UB, Willbold D. Resonance assignment of the ligand-free cyclic nucleotide-binding domain from the murine ion channel HCN2. BIOMOLECULAR NMR ASSIGNMENTS 2015; 9:243-246. [PMID: 25324217 DOI: 10.1007/s12104-014-9583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/26/2014] [Indexed: 06/04/2023]
Abstract
Hyperpolarization activated and cyclic nucleotide-gated (HCN) ion channels as well as cyclic nucleotide-gated (CNG) ion channels are essential for the regulation of cardiac cells, neuronal excitability, and signaling in sensory cells. Both classes are composed of four subunits. Each subunit comprises a transmembrane region, intracellular N- and C-termini, and a C-terminal cyclic nucleotide-binding domain (CNBD). Binding of cyclic nucleotides to the CNBD promotes opening of both CNG and HCN channels. In case of CNG channels, binding of cyclic nucleotides to the CNBD is sufficient to open the channel. In contrast, HCN channels open upon membrane hyperpolarization and their activity is modulated by binding of cyclic nucleotides shifting the activation potential to more positive values. Although several high-resolution structures of CNBDs from HCN and CNG channels are available, the gating mechanism for murine HCN2 channel, which leads to the opening of the channel pore, is still poorly understood. As part of a structural investigation, here, we report the complete backbone and side chain resonance assignments of the murine HCN2 CNBD with part of the C-linker.
Collapse
Affiliation(s)
- Claudia Börger
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany
| | - Sven Schünke
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Justin Lecher
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany
| | - Matthias Stoldt
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany
| | - Friederike Winkhaus
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, 53175, Bonn, Germany
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377, Penzberg, Germany
| | - U Benjamin Kaupp
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, 53175, Bonn, Germany
| | - Dieter Willbold
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany.
| |
Collapse
|
12
|
Silva MPD, Cedraz-Mercez PL, Varanda WA. Effects of nitric oxide on magnocellular neurons of the supraoptic nucleus involve multiple mechanisms. Braz J Med Biol Res 2014; 47:90-100. [PMID: 24519124 PMCID: PMC4051181 DOI: 10.1590/1414-431x20133326] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/22/2013] [Indexed: 01/24/2023] Open
Abstract
Physiological evidence indicates that the supraoptic nucleus (SON) is an
important region for integrating information related to homeostasis of body
fluids. Located bilaterally to the optic chiasm, this nucleus is composed of
magnocellular neurosecretory cells (MNCs) responsible for the synthesis and
release of vasopressin and oxytocin to the neurohypophysis. At the cellular
level, the control of vasopressin and oxytocin release is directly linked to the
firing frequency of MNCs. In general, we can say that the excitability of these
cells can be controlled via two distinct mechanisms: 1) the intrinsic membrane
properties of the MNCs themselves and 2) synaptic input from circumventricular
organs that contain osmosensitive neurons. It has also been demonstrated that
MNCs are sensitive to osmotic stimuli in the physiological range. Therefore, the
study of their intrinsic membrane properties became imperative to explain the
osmosensitivity of MNCs. In addition to this, the discovery that several
neurotransmitters and neuropeptides can modulate their electrical activity
greatly increased our knowledge about the role played by the MNCs in fluid
homeostasis. In particular, nitric oxide (NO) may be an important player in
fluid balance homeostasis, because it has been demonstrated that the enzyme
responsible for its production has an increased activity following a hypertonic
stimulation of the system. At the cellular level, NO has been shown to change
the electrical excitability of MNCs. Therefore, in this review, we focus on some
important points concerning nitrergic modulation of the neuroendocrine system,
particularly the effects of NO on the SON.
Collapse
Affiliation(s)
- M P da Silva
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão PretoSP, Brasil, Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - P L Cedraz-Mercez
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão PretoSP, Brasil, Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - W A Varanda
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão PretoSP, Brasil, Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
13
|
Palma-Flores C, Ramírez-Sánchez I, Rosas-Vargas H, Canto P, Coral-Vázquez RM. Description of a utrophin associated protein complex in lipid raft domains of human artery smooth muscle cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1047-54. [PMID: 24060563 DOI: 10.1016/j.bbamem.2013.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 09/06/2013] [Accepted: 09/12/2013] [Indexed: 01/08/2023]
Abstract
The dystrophin-associated protein complex (DAPC) is a multimeric complex that links the extracellular matrix to the actin cytoskeleton, and in some cases dystrophin can be substituted by its autosomal homologue utrophin to form the utrophin-associated protein complex (UAPC). Both complexes maintain the stability of plasma membrane during contraction process and play an important role in transmembrane signaling. Mutations in members of the DAPC are associated with muscular dystrophy and dilated cardiomyopathy. In a previous study with human umbilical cord vessels, we observed that utrophin colocalize with caveolin-1 (Cav-1) which proposed the presence of UAPC in the plasma membrane of vascular smooth muscle (VSM). In the current study, we demonstrated by immunofluorescence analysis, co-immunoprecipitation assays, and subcellular fractionation by sucrose gradients, the existence of an UAPC in lipid raft domains of human umbilical artery smooth muscle cells (HUASMC). This complex is constituted by utrophin, β-DG, ε-SG, α-smooth muscle actin, Cav-1, endothelial nitric oxide synthase (eNOS) and cavin-1. It was also observed the presence of dystrophin, utrophin Dp71, β-SG, δ-SG, δ-SG3 and sarcospan in non-lipid raft fractions. Furthermore, the knockdown of α/β-DG was associated with the decrease in both the synthesis of nitric oxide (NO) and the presence of the phosphorylated (active) form of eNOS; and with a reduction in the downstream activation of some cGMP signaling transduction pathway components. Together these results show the presence of an UAPC complex in HUASMC that may participate in the activity regulation of eNOS and in the vascular function.
Collapse
Affiliation(s)
- Carlos Palma-Flores
- División de Investigación Biomédica, Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, México, D.F., Mexico
| | - Israel Ramírez-Sánchez
- Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, D.F., Mexico
| | - Haydeé Rosas-Vargas
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Medico Nacional Siglo XXI-IMSS, Av. Cuauhtémoc No 330, Col Doctores, Delegación Cuauhtémoc, 06725 México, D.F., Mexico
| | - Patricia Canto
- División de Investigación Biomédica, Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, México, D.F., Mexico
| | - Ramón Mauricio Coral-Vázquez
- Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, D.F., Mexico; Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, México, D.F., Mexico.
| |
Collapse
|
14
|
Cywinski PJ, Moro AJ, Löhmannsröben HG. Cyclic GMP recognition using ratiometric QD-fluorophore conjugate nanosensors. Biosens Bioelectron 2013; 52:288-92. [PMID: 24071364 DOI: 10.1016/j.bios.2013.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 09/02/2013] [Indexed: 11/19/2022]
Abstract
Novel luminescent ratiometric nanosensors (QD-NAPTHs) were prepared based on cadmium telluride (CdTe655) quantum dots as luminescent nanoscaffolds with naphthyridine dyes as fluorescent receptors. This biosensing bifluorophoric nanosystem has been designed to achieve detection of guanosine 3',5'-cyclic monophosphate (cyclic GMP) in buffered media. Cyclic GMP is a secondary messenger that is an important factor for detecting cancer, diabetes and, cardiovascular diseases. Due to low concentration levels, even in pathological conditions, sensitive cGMP detection remains a challenge for modern biomedical diagnostics. Here, QD-NAPTH nanosensors were tested in the presence of a target nucleotide and with various structural cGMP analogues. Steady-state fluorescence spectroscopy was used to monitor a change in the nucleotide concentration. A 5-fold increase in naphthyridine fluorescence with a simultaneous decrease in QD luminescence was observed after adding 50 μM of cGMP. Using this novel nanosystem with ratiometric detection, it was possible to recognize cGMP with limit of detection (3σ) equal to 70 ng/ml. Moreover, the enhancement in fluorescence upon interaction with the target nucleotide constitutes a favourable approach towards the detection of cGMP in buffered media. These bifluorophoric nanosensors have a potential for application in fluorescence microscopy imaging and in-vitro assays.
Collapse
Affiliation(s)
- Piotr J Cywinski
- Physical Chemistry, Department of Chemistry, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476 Potsdam-Golm, Germany.
| | | | | |
Collapse
|
15
|
Phosphodiesterases: Regulators of cyclic nucleotide signals and novel molecular target for movement disorders. Eur J Pharmacol 2013; 714:486-97. [PMID: 23850946 DOI: 10.1016/j.ejphar.2013.06.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 06/16/2013] [Accepted: 06/21/2013] [Indexed: 12/21/2022]
Abstract
Movement disorders rank among the most common neurological disorders. During the last two decades substantial progress has been made in understanding of the pathological basis of these disorders. Although, several mechanisms have been proposed, downregulation of cyclic nucleotide mediated signaling cascade has consistently been shown to contribute to the striatal dysfunctioning as seen in movement disorders. Thus, counteracting dysregulated cyclic nucleotide signaling has been considered to be beneficial in movement disorders. Cyclic nucleotide phosphodiesterases (PDEs) are the enzymes responsible for the breakdown of cyclic nucleotides and upregulation in PDE activity has been reported in various movement disorders. Thus, PDE inhibition is considered to be a novel strategy to restore cerebral cyclic nucleotide levels and their downstream signalling cascade. Indeed, various PDE inhibitors have been tested pre-clinically and were reported to be neuroprotective in various neurodegenerative disorders associated with movement disabilities. In this review, we have discussed a putative role of PDE inhibitors in movement disorders and associated abnormalities.
Collapse
|
16
|
Xu Z, Lee S, Han J. Dual role of cyclic GMP in cardiac cell survival. Int J Biochem Cell Biol 2013; 45:1577-84. [PMID: 23660294 DOI: 10.1016/j.biocel.2013.04.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/08/2013] [Accepted: 04/29/2013] [Indexed: 10/26/2022]
Abstract
It is well known that cyclic guanosine 3',5'-monophosphate plays an important role in cardioprotection against ischemia/reperfusion injury through activation of protein kinase G (PKG). We found that cGMP prevents the mitochondrial permeability transition pore (mPTP) opening by inactivating glycogen synthase kinase 3β (GSK-3β) via protein kinase G (PKG) in cardiac H9c2 cells. While GSK-3β and its major upstream regulator phosphoinositide 3-kinase (PI3K)/Akt are critical for acute cardioprotection, an excessive activation of PI3K/Akt or GSK-3β inactivation can also lead to cardiac hypertrophy. Here, we show that cGMP not only inactivates GSK-3β through PKG (this leads to acute cardioprotection) but also negatively regulates Akt activity (this may lead to prevention of hypertrophy and heart failure, and the regulation of NO synthesis) in cardiac cells. We further found that the negative regulatory effect of cGMP on Akt activity is not mediated by PKG but may be through up-regulation of protein phosphatase PP2A activity. We propose that cGMP is a versatile signal with dual beneficial role in cardiac cell survival.
Collapse
Affiliation(s)
- Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China.
| | | | | |
Collapse
|
17
|
Mak DOD. A mechanism for different receptors coupled to the same G protein to generate different responses mediated by different second messengers. J Gen Physiol 2013; 141:513-6. [PMID: 23630335 PMCID: PMC3639579 DOI: 10.1085/jgp.201311006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Don-On Daniel Mak
- Physiology Department, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Abstract
cGMP-dependent protein kinases (cGK) are serine/threonine kinases that are widely distributed in eukaryotes. Two genes-prkg1 and prkg2-code for cGKs, namely, cGKI and cGKII. In mammals, two isozymes, cGKIα and cGKIβ, are generated from the prkg1 gene. The cGKI isozymes are prominent in all types of smooth muscle, platelets, and specific neuronal areas such as cerebellar Purkinje cells, hippocampal neurons, and the lateral amygdala. The cGKII prevails in the secretory epithelium of the small intestine, the juxtaglomerular cells, the adrenal cortex, the chondrocytes, and in the nucleus suprachiasmaticus. Both cGKs are major downstream effectors of many, but not all, signalling events of the NO/cGMP and the ANP/cGMP pathways. cGKI relaxes smooth muscle tone and prevents platelet aggregation, whereas cGKII inhibits renin secretion, chloride/water secretion in the small intestine, the resetting of the clock during early night, and endochondral bone growth. This chapter focuses on the involvement of cGKs in cardiovascular and non-cardiovascular processes including cell growth and metabolism.
Collapse
Affiliation(s)
- Franz Hofmann
- FOR 923, Institut für Pharmakologie und Toxikologie, der Technischen Universität München, Munich, Germany
| | | |
Collapse
|
19
|
Sharma DK, Manral A, Saini V, Singh A, Srinivasan BP, Tiwari M. Novel diallyldisulfide analogs ameliorate cardiovascular remodeling in rats with L-NAME-induced hypertension. Eur J Pharmacol 2012; 691:198-208. [PMID: 22819707 DOI: 10.1016/j.ejphar.2012.07.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 06/20/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
Abstract
Diallyldisulfide (DADS), an active principle of garlic (Allium sativum) is known for its antihypertensive properties. The present study was designed to evaluate the effect of novel DADS analogs, against L-NAME induced hypertension in Wistar rats. The daily administration of L-NAME (50mg/kg) for six weeks along with DADS analogs (20 mg/kg) significantly decreased the elevated systolic blood pressure (SBP) and the activity of angiotensin converting enzyme (ACE) and also inhibited the decline in nitrite/nitrate (NO(x)) concentrations and cyclic guanosine monophosphate (cGMP) levels. Adverse changes such as lipid peroxidation, protein damage and a decrease in the levels of antioxidant enzymes, were rectified after the administration of DADS analogs. Oral administration of DADS analogs preserved the expression of endothelial nitric oxide synthase (eNOS). The ability of the DADS analogs to inhibit L-NAME induced hypertension was compared with Enalapril (15 mg/kg), which was taken as a standard. The DADS analogs prevented L-NAME-induced cardio toxicity, which was also reflected at the microscopic level indicative of its cardio protective effects. DADS analogs induced vasorelaxation was completely abolished by the removal of the endothelium or by pre-treatment with L-NAME, an inhibitor of nitric oxide synthase. DADS analogs inhibited the calcium influx induced by phenylephrine (0.3 μM) and high K(+) (60mM) and this effect was completely abolished by pretreatment of L-NAME. Taken together, our results show that the DADS analogs induce vasorelaxation and have antihypertensive properties, which may be mediated through activation of eNOS.
Collapse
Affiliation(s)
- Dinesh Kumar Sharma
- Dr. BR Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India
| | | | | | | | | | | |
Collapse
|
20
|
Hwang JH, Moon SA, Lee CH, Byun MR, Kim AR, Sung MK, Park HJ, Hwang ES, Sung SH, Hong JH. Idesolide inhibits the adipogenic differentiation of mesenchymal cells through the suppression of nitric oxide production. Eur J Pharmacol 2012; 685:218-23. [DOI: 10.1016/j.ejphar.2012.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 03/23/2012] [Accepted: 04/05/2012] [Indexed: 11/16/2022]
|
21
|
Saravani R, Karami-Tehrani F, Hashemi M, Aghaei M, Edalat R. Inhibition of phosphodiestrase 9 induces cGMP accumulation and apoptosis in human breast cancer cell lines, MCF-7 and MDA-MB-468. Cell Prolif 2012; 45:199-206. [PMID: 22469131 DOI: 10.1111/j.1365-2184.2012.00819.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 02/21/2012] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Phosphodiesterase 9 (PDE9) is a major isoform of phosphodiesterase hydrolysing cGMP and plays a key role in proliferation of cells, their differentiation and apoptosis, via intracellular cGMP signalling. The study described here was designed to investigate expression, activity and apoptotic effect of PDE9 on human breast cancer cell lines, MCF-7 and MDA-MB-468. MATERIALS AND METHODS Activity and expression of PDE9 were examined using colorimetric cyclic nucleotide phosphodiesterase assay and real-time RT-PCR methods respectively; cGMP concentration was also measured. MTT viability test, annexin V-FITC staining, Hoechst 33258 staining and caspase3 activity assay were used to detect apoptosis. RESULTS Treatment of both cell lines with BAY 73-6691 lead to reduction in PDE9 mRNA expression, PDE9 cGMP-hydrolytic activity and elevation of the intracellular cGMP response. BAY 73-6691 significantly reduced cell proliferation in a dose- and time-dependent manner and caused marked increase in apoptosis through caspase3 activation. CONCLUSION Our results revealed that BAY 73-6691 induced apoptosis in these breast cancer cell lines through the cGMP pathway. These data suggest that BAY 73-6691 could be utilized as an agent in treatment of breast cancer.
Collapse
Affiliation(s)
- R Saravani
- Clinical Biochemistry Department, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | |
Collapse
|
22
|
Adderley SP, Joshi CN, Martin DN, Tulis DA. Phosphodiesterases Regulate BAY 41-2272-Induced VASP Phosphorylation in Vascular Smooth Muscle Cells. Front Pharmacol 2012; 3:10. [PMID: 22347188 PMCID: PMC3273712 DOI: 10.3389/fphar.2012.00010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 01/18/2012] [Indexed: 11/13/2022] Open
Abstract
BAY 41-2272 (BAY), a stimulator of soluble guanylyl cyclase, increases cyclic nucleotides and inhibits proliferation of vascular smooth muscle cells (VSMCs). In this study, we elucidated mechanisms of action of BAY in its regulation of vasodilator-stimulated phosphoprotein (VASP) with an emphasis on VSMC phosphodiesterases (PDEs). BAY alone increased phosphorylation of VASP(Ser239) and VASP(Ser157), respective indicators of PKG and PKA signaling. IBMX, a non-selective inhibitor of PDEs, had no effect on BAY-induced phosphorylation at VASP(Ser239) but inhibited phosphorylation at VASP(Ser157). Selective inhibitors of PDE3 or PDE4 attenuated BAY-mediated increases at VASP(Ser239) and VASP(Ser157), whereas PDE5 inhibition potentiated BAY-mediated increases only at VASP(Ser157). In comparison, 8Br-cGMP increased phosphorylation at VASP(Ser239) and VASP(Ser157) which were not affected by selective PDE inhibitors. In the presence of 8Br-cAMP, inhibition of either PDE4 or PDE5 decreased VASP(Ser239) phosphorylation and inhibition of PDE3 increased phosphorylation at VASP(Ser239), while inhibition of PDE3 or PDE4 increased and PDE5 inhibition had no effect on VASP(Ser157) phosphorylation. These findings demonstrate that BAY operates via cAMP and cGMP along with regulation by PDEs to phosphorylate VASP in VSMCs and that the mechanism of action of BAY in VSMCs is different from that of direct cyclic nucleotide analogs with respect to VASP phosphorylation and the involvement of PDEs. Given a role for VASP as a critical cytoskeletal protein, these findings provide evidence for BAY as a regulator of VSMC growth and a potential therapeutic agent against vasculoproliferative disorders.
Collapse
Affiliation(s)
- Shaquria P Adderley
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | | | | | | |
Collapse
|
23
|
Morgado M, Cairrão E, Santos-Silva AJ, Verde I. Cyclic nucleotide-dependent relaxation pathways in vascular smooth muscle. Cell Mol Life Sci 2012; 69:247-66. [PMID: 21947498 PMCID: PMC11115151 DOI: 10.1007/s00018-011-0815-2] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 08/21/2011] [Accepted: 08/23/2011] [Indexed: 02/07/2023]
Abstract
Vascular smooth muscle tone is controlled by a balance between the cellular signaling pathways that mediate the generation of force (vasoconstriction) and release of force (vasodilation). The initiation of force is associated with increases in intracellular calcium concentrations, activation of myosin light-chain kinase, increases in the phosphorylation of the regulatory myosin light chains, and actin-myosin crossbridge cycling. There are, however, several signaling pathways modulating Ca(2+) mobilization and Ca(2+) sensitivity of the contractile machinery that secondarily regulate the contractile response of vascular smooth muscle to receptor agonists. Among these regulatory mechanisms involved in the physiological regulation of vascular tone are the cyclic nucleotides (cAMP and cGMP), which are considered the main messengers that mediate vasodilation under physiological conditions. At least four distinct mechanisms are currently thought to be involved in the vasodilator effect of cyclic nucleotides and their dependent protein kinases: (1) the decrease in cytosolic calcium concentration ([Ca(2+)]c), (2) the hyperpolarization of the smooth muscle cell membrane potential, (3) the reduction in the sensitivity of the contractile machinery by decreasing the [Ca(2+)]c sensitivity of myosin light-chain phosphorylation, and (4) the reduction in the sensitivity of the contractile machinery by uncoupling contraction from myosin light-chain phosphorylation. This review focuses on each of these mechanisms involved in cyclic nucleotide-dependent relaxation of vascular smooth muscle under physiological conditions.
Collapse
Affiliation(s)
- Manuel Morgado
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Elisa Cairrão
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - António José Santos-Silva
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ignacio Verde
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
24
|
Fu JY, Qian LB, Zhu LG, Liang HT, Tan YN, Lu HT, Lu JF, Wang HP, Xia Q. Betulinic acid ameliorates endothelium-dependent relaxation in L-NAME-induced hypertensive rats by reducing oxidative stress. Eur J Pharm Sci 2011; 44:385-391. [PMID: 21907795 DOI: 10.1016/j.ejps.2011.08.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/27/2011] [Indexed: 02/07/2023]
Abstract
Zizyphi Spinosi semen (ZSS) is one of the most widely used traditional Chinese herbs with protective effects on the cardiovascular system. It is not clear whether betulinic acid (BA), the key active constituent of ZSS, has beneficial cardiovascular effects on N(ω)-nitro-L-arginine methyl ester hydrochloride (L-NAME)-induced hypertensive rats. The objective of this study was to investigate the effect of BA on endothelium-dependent vasorelaxation in isolated aortic rings from L-NAME-induced hypertensive rats and its underlying mechanisms. Male Sprague-Dawley rats were injected with L-NAME (15 mg/kg/d, i.p.) for 4 weeks to induce hypertension. After treatment with L-NAME for 2 weeks, rats with mean blood pressure >120 mm Hg measured by tail-cuff method were considered hypertensive and then injected with BA (0.8, 4, 20 mg/kg/d, i.p.) for the last 2 weeks. The effect of BA on the tension of rat thoracic aortic rings was measured in an organ bath system. The levels of nitric oxide (NO), reactive oxygen species (ROS), and the activity of superoxide dismutase (SOD) and nitric oxide synthase (NOS) in aortas were assayed. We found that BA (0.1-100 μM) evoked a concentration-dependent vasorelaxation in endothelium-intact normal rat aortic rings, which was significantly attenuated by pretreatment with L-NAME (100 μM) or methylene blue (MB, 10 μM), but not by indomethacin (10 μM). Pretreatment with EC(50) (1.67 μM) concentration of BA enhanced the acetylcholine (ACh)-induced vasorelaxation, which was also markedly reversed by both L-NAME and MB. The blood pressure in hypertensive rats increased to 135.22±5.38 mm Hg (P<0.01 vs. control group), which was markedly attenuated by high dose of BA. The ACh-induced vasorelaxation in hypertensive rat aortic rings was impaired, which was markedly improved by chronic treatment with BA (20 mg/kg/d) for 2 weeks. The increase of ROS level and the decrease of NO level, SOD and eNOS activities in hypertensive rat aortas were all markedly inhibited by BA. These results indicate that BA decreased blood pressure and improved ACh-induced endothelium-dependent vasorelaxation in L-NAME-induced hypertension rats, which may be mediated by reducing oxidative stress and retaining the bioavailability of NO in the cardiovascular system.
Collapse
Affiliation(s)
- Jia-Yin Fu
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Armani A, Marzolla V, Rosano GMC, Fabbri A, Caprio M. Phosphodiesterase type 5 (PDE5) in the adipocyte: a novel player in fat metabolism? Trends Endocrinol Metab 2011; 22:404-11. [PMID: 21741267 DOI: 10.1016/j.tem.2011.05.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/18/2011] [Accepted: 05/25/2011] [Indexed: 12/19/2022]
Abstract
Phosphodiesterase type 5 (PDE5) is expressed in many tissues (e.g. heart, lung, pancreas, penis) and plays a specific role in hydrolyzing cyclic guanosine monophosphate (cGMP). In adipocytes, cGMP regulates crucial functions by activating cGMP-dependent protein kinase (PKG). Interestingly, PDE5 was recently identified in adipose tissue, although its role remains unclear. Its inhibition, however, was recently shown to affect adipose differentiation and aromatase function. This review summarizes evidence supporting a role for the PDE5-regulated cGMP/PKG system in adipose tissue and its effects on adipocyte function. A better elucidation of the role of PDE5 in the adipocyte could reveal new therapeutic strategies for fighting obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Andrea Armani
- Center for Clinical and Basic Research, Scientific Institute for Research, Hospitalization and Health Care (IRCCS) San Raffaele Pisana, Rome, Italy
| | | | | | | | | |
Collapse
|
26
|
Fallahian F, Karami-Tehrani F, Salami S, Aghaei M. Cyclic GMP induced apoptosis via protein kinase G in oestrogen receptor-positive and -negative breast cancer cell lines. FEBS J 2011; 278:3360-9. [PMID: 21777390 DOI: 10.1111/j.1742-4658.2011.08260.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The activation of protein kinase G (PKG) by cyclic guanosine 3,5-monophosphate (cGMP) has become of considerable interest as a novel molecular approach for the induction of apoptosis in cancer cells. The present study was designed to examine the effects of cGMP and PKG on cell growth and apoptosis in the human breast cancer cell lines, MCF-7 and MDA-MB-468. To achieve this, 1-benzyl-3-(5P-hydroxymethyl-2P-furyl) indazole (YC-1), a soluble guanylyl cyclase activator, and 8-bromo-cGMP (8-br-cGMP), a membrane-permeant and phosphodiesterase-resistant analogue of cGMP, were employed in MCF-7 and MDA-MB-468 cells. Then, the role of PKG in the induction of apoptosis was evaluated using KT5823 and Rp-8-pCPT-cGMP as specific inhibitors of PKG. The expression of PKG isoforms in these cell lines was also investigated. KT5823 and Rp-8-pCPT-cGMP significantly attenuated the loss of cell viability caused by YC-1 and 8-br-cGMP in these cells. This study provides direct evidence that the activation of PKG by cGMP induces growth inhibition and apoptosis in MCF-7 and MDA-MB-468 breast cancer cell lines.
Collapse
Affiliation(s)
- Faranak Fallahian
- Department of Clinical Biochemistry, Cancer Research Laboratory, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | | | | | | |
Collapse
|
27
|
Kotlo KU, Hesabi B, Danziger RS. Implication of microRNAs in atrial natriuretic peptide and nitric oxide signaling in vascular smooth muscle cells. Am J Physiol Cell Physiol 2011; 301:C929-37. [PMID: 21734186 DOI: 10.1152/ajpcell.00088.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRs) are endogenous small RNA molecules that suppress gene expression by binding to complementary sequences in the 3' untranslated regions of their target genes. miRs have been implicated in many diseases, including heart failure, ischemic heart disease, hypertension, cardiac hypertrophy, and cancers. Nitric oxide (NO) and atrial natriuretic peptide (ANP) are potent vasorelaxants whose actions are mediated through receptor guanylyl cyclases and cGMP-dependent protein kinase. The present study examines miRs in signaling by ANP and NO in vascular smooth muscle cells. miR microarray analysis was performed on human vascular smooth muscle cells (HVSMC) treated with ANP (10 nM, 4 h) and S-nitroso-N-acetylpenicillamine (SNAP) (100 μM, 4 h), a NO donor. Twenty-two shared miRs were upregulated, and 21 shared miRs were downregulated, by both ANP and SNAP (P < 0.05). Expression levels of four miRs (miRs-21, -26b, -98, and -1826), which had the greatest change in expression, as determined by microarray analysis, were confirmed by quantitative RT-PCR. Rp-8-Br-PET-cGMPS, a cGMP-dependent protein kinase-specific inhibitor, blocked the regulation of these miRs by ANP and SNAP. 8-bromo-cGMP mimicked the effect of ANP and SNAP on their expression. miR-21 was shown to inhibit HVSMC contraction in collagen gel lattice contraction assays. We also identified by computational algorithms and confirmed by Western blot analysis new intracellular targets of miR-21, i.e., cofilin-2 and myosin phosphatase and Rho interacting protein. Transfection with pre-miR-21 contracted cells and ANP and SNAP blocked miR-21-induced HVSMC contraction. Transfection with anti-miR-21 inhibitor reduced contractility of HVSMC (P < 0.05). The present results implicate miRs in NO and ANP signaling in general and miR-21 in particular in cGMP signaling and vascular smooth muscle cell relaxation.
Collapse
Affiliation(s)
- Kumar U Kotlo
- Department of Medicine, University of Illinois at Chicago, 60612, USA.
| | | | | |
Collapse
|
28
|
Inhibition of hepatitis C virus replicon RNA synthesis by PSI-352938, a cyclic phosphate prodrug of β-D-2'-deoxy-2'-α-fluoro-2'-β-C-methylguanosine. Antimicrob Agents Chemother 2011; 55:2566-75. [PMID: 21444700 DOI: 10.1128/aac.00032-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PSI-352938 is a novel cyclic phosphate prodrug of β-D-2'-deoxy-2'-α-fluoro-2'-β-C-methylguanosine 5'-monophosphate that has potent activity against hepatitis C virus (HCV) in vitro. The studies described here characterize the in vitro anti-HCV activity of PSI-352938, alone and in combination with other inhibitors of HCV, and the cross-resistance profile of PSI-352938. The effective concentration required to achieve 50% inhibition for PSI-352938, determined using genotype 1a-, 1b-, and 2a-derived replicons stably expressed in the Lunet cell line, were 0.20, 0.13, and 0.14 μM, respectively. The active 5'-triphosphate metabolite, PSI-352666, inhibited recombinant NS5B polymerase from genotypes 1 to 4 with comparable 50% inhibitory concentrations. In contrast, PSI-352938 did not inhibit the replication of hepatitis B virus or human immunodeficiency virus in vitro. PSI-352666 did not significantly affect the activity of human DNA and RNA polymerases. PSI-352938 and its cyclic phosphate metabolites did not affect the cyclic GMP-mediated activation of protein kinase G. Clearance studies using replicon cells demonstrated that PSI-352938 cleared cells of HCV replicon RNA and prevented replicon rebound. An additive to synergistic effect was observed when PSI-352938 was combined with other classes of HCV inhibitors, including alpha interferon, ribavirin, NS3/4A inhibitors, an NS5A inhibitor, and nucleoside/nucleotide and nonnucleoside inhibitors. Cross-resistance studies showed that PSI-352938 remained fully active against replicons containing the S282T or the S96T/N142T amino acid alteration. Replicons that contain mutations conferring resistance to various classes of nonnucleoside inhibitors also remained sensitive to inhibition by PSI-352938. PSI-352938 is currently being evaluated in a phase I clinical study in genotype 1-infected individuals.
Collapse
|
29
|
An Z, Winnick JJ, Farmer B, Neal D, Lautz M, Irimia JM, Roach PJ, Cherrington AD. A soluble guanylate cyclase-dependent mechanism is involved in the regulation of net hepatic glucose uptake by nitric oxide in vivo. Diabetes 2010; 59:2999-3007. [PMID: 20823104 PMCID: PMC2992759 DOI: 10.2337/db10-0138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 08/25/2010] [Indexed: 12/31/2022]
Abstract
OBJECTIVE We previously showed that elevating hepatic nitric oxide (NO) levels reduced net hepatic glucose uptake (NHGU) in the presence of portal glucose delivery, hyperglycemia, and hyperinsulinemia. The aim of the present study was to determine the role of a downstream signal, soluble guanylate cyclase (sGC), in the regulation of NHGU by NO. RESEARCH DESIGN AND METHODS Studies were performed on 42-h-fasted conscious dogs fitted with vascular catheters. At 0 min, somatostatin was given peripherally along with 4× basal insulin and basal glucagon intraportally. Glucose was delivered at a variable rate via a leg vein to double the blood glucose level and hepatic glucose load throughout the study. From 90 to 270 min, an intraportal infusion of the sGC inhibitor 1H-[1,2,4] oxadiazolo[4,3-a] quinoxalin-1-one (ODQ) was given in -sGC (n = 10) and -sGC/+NO (n = 6), whereas saline was given in saline infusion (SAL) (n = 10). The -sGC/+NO group also received intraportal SIN-1 (NO donor) to elevate hepatic NO from 180 to 270 min. RESULTS In the presence of 4× basal insulin, basal glucagon, and hyperglycemia (2× basal ), inhibition of sGC in the liver enhanced NHGU (mg/kg/min; 210-270 min) by ∼55% (2.9 ± 0.2 in SAL vs. 4.6 ± 0.5 in -sGC). Further elevating hepatic NO failed to reduce NHGU (4.5 ± 0.7 in -sGC/+NO). Net hepatic carbon retention (i.e., glycogen synthesis; mg glucose equivalents/kg/min) increased to 3.8 ± 0.2 in -sGC and 3.8 ± 0.4 in -sGC/+NO vs. 2.4 ± 0.2 in SAL (P < 0.05). CONCLUSIONS NO regulates liver glucose uptake through a sGC-dependent pathway. The latter could be a target for pharmacologic intervention to increase meal-associated hepatic glucose uptake in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Zhibo An
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee. USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhang YW, Rudnick G. Myristoylation of cGMP-dependent protein kinase dictates isoform specificity for serotonin transporter regulation. J Biol Chem 2010; 286:2461-8. [PMID: 21097501 DOI: 10.1074/jbc.m110.203935] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
By transporting serotonin (5-HT) into neurons and other cells, serotonin transporter (SERT) modulates the action of 5-HT at cell surface receptors. SERT itself is modulated by several processes, including the cGMP signaling pathway. Activation of SERT by cGMP requires the cGMP-dependent protein kinase (PKG). Here we show that in HeLa cells lacking endogenous PKG, expression of PKGIα or PKGIβ was required for 8-bromoguanosine-3',5'-cyclic monophosphate (8-Br-cGMP) to stimulate SERT phosphorylation and 5-HT influx. Catalytically inactive PKG mutants and wild-type PKGII did not support this stimulation. However, a mutant PKGII (G2A) that was not myristoylated substituted for functional PKGI, suggesting that myristoylation and subsequent membrane association blocked productive interaction with SERT. PKG also influenced SERT expression and localization. PKGI isoforms increased total and cell surface SERT levels, and PKGII decreased cell surface SERT without altering total expression. Remarkably, these changes did not require 8-Br-cGMP or functional kinase activity and were also observed with a SERT mutant resistant to activation by PKG. Both PKGIα and PKGIβ formed detergent-stable complexes with SERT, and this association did not require catalytic activity. The nonmyristoylated PKGII G2A mutant stimulated SERT expression similar to PKGI isoforms. These results suggest multiple mechanisms by which PKG can modulate SERT and demonstrate that the functional difference between PKG isoforms results from myristoylation of PKGII.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066, USA
| | | |
Collapse
|
31
|
Casserly B, Klinger JR. Brain natriuretic peptide in pulmonary arterial hypertension: biomarker and potential therapeutic agent. DRUG DESIGN DEVELOPMENT AND THERAPY 2009; 3:269-87. [PMID: 20054445 PMCID: PMC2802126 DOI: 10.2147/dddt.s4805] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
B-type natriuretic peptide (BNP) is a member of the natriuretic peptide family, a group of widely distributed, but evolutionarily conserved, polypeptide mediators that exert myriad cardiovascular effects. BNP is a potent vasodilator with mitogenic, hypertrophic and pro-inflammatory properties that is upregulated in pulmonary hypertensive diseases. Circulating levels of BNP correlate with mean pulmonary arterial pressure (mPAP) and pulmonary vascular resistance (PVR) in patients with pulmonary arterial hypertension (PAH). Elevated plasma BNP levels are associated with increased mortality in patients with PAH and a fall in BNP levels after therapy is associated with improved survival. These findings have important clinical implications in that a noninvasive blood test may be used to identify PAH patients at high-risk of decompensation and to guide pulmonary vasodilator therapy. BNP also has several biologic effects that could be beneficial to patients with PAH. However, lack of a convenient method for achieving sustained increases in circulating BNP levels has impeded the development of BNP as a therapy for treating pulmonary hypertension. New technologies that allow transdermal or oral administration of the natriuretic peptides have the potential to greatly accelerate research into therapeutic use of BNP for cor pulmonale and pulmonary vascular diseases. This review will examine the basic science and clinical research that has led to our understanding of the role of BNP in cardiovascular physiology, its use as a biomarker of right ventricular function and its therapeutic potential for managing patients with pulmonary vascular disease.
Collapse
Affiliation(s)
- Brian Casserly
- Division of Pulmonary and Critical Care Medicine, The Memorial Hospital of Rhode Island, Pawtucket, RI, USA
| | | |
Collapse
|
32
|
Tsai EJ, Kass DA. Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics. Pharmacol Ther 2009; 122:216-38. [PMID: 19306895 PMCID: PMC2709600 DOI: 10.1016/j.pharmthera.2009.02.009] [Citation(s) in RCA: 313] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) mediates a wide spectrum of physiologic processes in multiple cell types within the cardiovascular system. Dysfunctional signaling at any step of the cascade - cGMP synthesis, effector activation, or catabolism - have been implicated in numerous cardiovascular diseases, ranging from hypertension to atherosclerosis to cardiac hypertrophy and heart failure. In this review, we outline each step of the cGMP signaling cascade and discuss its regulation and physiologic effects within the cardiovascular system. In addition, we illustrate how cGMP signaling becomes dysregulated in specific cardiovascular disease states. The ubiquitous role cGMP plays in cardiac physiology and pathophysiology presents great opportunities for pharmacologic modulation of the cGMP signal in the treatment of cardiovascular diseases. We detail the various therapeutic interventional strategies that have been developed or are in development, summarizing relevant preclinical and clinical studies.
Collapse
Affiliation(s)
- Emily J Tsai
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
33
|
Aicher A, Heeschen C, Feil S, Hofmann F, Mendelsohn ME, Feil R, Dimmeler S. cGMP-dependent protein kinase I is crucial for angiogenesis and postnatal vasculogenesis. PLoS One 2009; 4:e4879. [PMID: 19287493 PMCID: PMC2654072 DOI: 10.1371/journal.pone.0004879] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2008] [Accepted: 02/10/2009] [Indexed: 01/13/2023] Open
Abstract
Background Endothelium-derived nitric oxide plays an important role for the bone marrow microenvironment. Since several important effects of nitric oxide are mediated by cGMP-dependent pathways, we investigated the role of the cGMP downstream effector cGMP-dependent protein kinase I (cGKI) on postnatal neovascularization. Methodology/Principal Findings In a disc neovascularization model, cGKI−/− mice showed an impaired neovascularization as compared to their wild-type (WT) littermates. Infusion of WT, but not cGKI−/− bone marrow progenitors rescued the impaired ingrowth of new vessels in cGKI-deficient mice. Bone marrow progenitors from cGKI−/− mice showed reduced proliferation and survival rates. In addition, we used cGKIα leucine zipper mutant (LZM) mice as model for cGKI deficiency. LZM mice harbor a mutation in the cGKIα leucine zipper that prevents interaction with downstream signaling molecules. Consistently, LZM mice exhibited reduced numbers of vasculogenic progenitors and impaired neovascularization following hindlimb ischemia compared to WT mice. Conclusions/Significance Our findings demonstrate that the cGMP-cGKI pathway is critical for postnatal neovascularization and establish a new role for cGKI in vasculogenesis, which is mediated by bone marrow-derived progenitors.
Collapse
Affiliation(s)
- Alexandra Aicher
- Department of Internal Medicine III, J. W. Goethe University, Frankfurt, Germany
- * E-mail: (AA); (SD)
| | - Christopher Heeschen
- Department of Internal Medicine III, J. W. Goethe University, Frankfurt, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, Universität Tübingen, Tübingen, Germany
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie, Technische Universität München, München, Germany
| | - Michael E. Mendelsohn
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Robert Feil
- Interfakultäres Institut für Biochemie, Universität Tübingen, Tübingen, Germany
| | - Stefanie Dimmeler
- Department of Internal Medicine III, J. W. Goethe University, Frankfurt, Germany
- * E-mail: (AA); (SD)
| |
Collapse
|
34
|
New aspects of adipogenesis: Radicals and oxidative stress. Differentiation 2009; 77:115-20. [DOI: 10.1016/j.diff.2008.09.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 07/06/2008] [Accepted: 07/25/2008] [Indexed: 01/04/2023]
|
35
|
González-Flores O, Gómora-Arrati P, Garcia-Juárez M, Gómez-Camarillo MA, Lima-Hernández FJ, Beyer C, Etgen AM. Nitric oxide and ERK/MAPK mediation of estrous behavior induced by GnRH, PGE2 and db-cAMP in rats. Physiol Behav 2009; 96:606-12. [PMID: 19162055 DOI: 10.1016/j.physbeh.2008.12.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 12/16/2008] [Accepted: 12/17/2008] [Indexed: 01/27/2023]
Abstract
We tested the hypothesis that GnRH, PGE2 and db-cAMP act via the nitric oxide (NO)-cGMP and MAPK pathways to facilitate estrous behavior (lordosis and proceptivity) in estradiol-primed female rats. Estradiol-primed rats received intracerebroventricular (icv) infusions of pharmacological antagonists of NO synthase (L-NAME), NO-dependent soluble guanylyl cyclase (ODQ), protein kinase G (KT5823), or the ERK1/2 inhibitor PD98059 15 min before icv administration of 50 ng of GnRH, 1 microg of PGE2 or 1 microg of db-cAMP. Icv infusions of GnRH, PGE2 and db-cAMP enhanced estrous behavior at 1 and 2 h after drug administration. Both L-NAME and ODQ blocked the estrous behavior induced by GnRH, PGE2 and db-cAMP at some of the times tested. The protein kinase G inhibitor KT5823 reduced PGE2 and db-cAMP facilitation of estrous behavior but did not affect the behavioral response to GnRH. In contrast, PD98059 blocked the estrous behavior induced by all three compounds. These data support the hypothesis that the NO-cGMP and ERK/MAPK pathways are involved in the lordosis and proceptive behaviors induced by GnRH, PGE2 and db-cAMP. However, cGMP mediation of GnRH-facilitated estrous behavior is independent of protein kinase G.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Apdo. Postal 62, Tlaxcala 90000, Mexico
| | | | | | | | | | | | | |
Collapse
|
36
|
LaPensee CR, Hugo ER, Ben-Jonathan N. Insulin stimulates interleukin-6 expression and release in LS14 human adipocytes through multiple signaling pathways. Endocrinology 2008; 149:5415-22. [PMID: 18617614 PMCID: PMC2584585 DOI: 10.1210/en.2008-0549] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
IL-6 is an important cytokine that regulates both immune and metabolic functions. Within adipose tissue, preadipocytes produce significant amounts of IL-6, but little is known about the factors or mechanisms that regulate IL-6 production in these cells. Using LS14, a newly developed human adipocyte cell line, our objective was to determine the mechanisms by which insulin stimulates IL-6 production and release in preadipocytes. Insulin increased IL-6 gene expression and secretion in a time- and dose-dependent manner. Insulin decreased cyclic AMP (cAMP) but increased cyclic GMP (cGMP) levels, and IL-6 expression/release was stimulated by a cGMP analog. The stimulatory effect of insulin and cGMP was abrogated by a specific inhibitor of protein kinase G (cyclic GMP-dependent protein kinase). Both insulin and cGMP rapidly induced phosphorylation of cAMP response element binding protein. Insulin also activated the MAPK signaling pathway, and its blockade prevented the insulin-stimulated increases in IL-6 cell content and release, but not IL-6 gene expression. Although inhibition of the proteosome increased IL-6 cell content and release, proteosome activity was unaffected by insulin. These data suggest that the stimulatory effects of insulin on IL-6 release involve several interrelated components: transcription, intracellular releasable pool, and secretion, which are differentially regulated and, thus, determine the size of the releasable pool of IL-6. Insulin-induced IL-6 gene expression is mediated by cGMP/cyclic GMP-dependent protein kinase/cAMP response element binding protein, whereas MAPK is involved in the insulin-stimulated IL-6 synthesis/release.
Collapse
Affiliation(s)
- Christopher R LaPensee
- Department of cancer and Cell Biology, University of Cincinnati School of Medicine, Cincinnati, Ohio 45267-0521, USA
| | | | | |
Collapse
|
37
|
The nitric oxide pathway in the human prostate: clinical implications in men with lower urinary tract symptoms. World J Urol 2008; 26:603-9. [PMID: 18607596 DOI: 10.1007/s00345-008-0303-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 06/16/2008] [Indexed: 12/15/2022] Open
Abstract
To date, there is an increasing interest in the nitric oxide (NO) pathway as a potential pharmacological target to treat male lower urinary tract symptomatology (LUTS). In the transition zone of the human prostate, a dense nitrinergic innervation has been shown of the fibromuscular stroma, glandular epithelium and blood vessels. The expression of key proteins of the NO pathway, such as the endothelial and neuronal nitric oxide synthase (eNOS, nNOS), cGMP-degrading phosphodiesterase type 5 (PDE5) and cGMP-binding protein kinase (cGK), has also been demonstrated. The hypothesis that an impaired NO/cGMP-signaling may contribute to the pathophysiology of benign prostatic hyperplasia (BPH) is supported by the results from randomized, placebo-controlled clinical studies, indicating that NO donor drugs and PDE5-inhibitors sildenafil, tadalafil and vardenafil may be useful to treat storage and voiding dysfunctions resulting from LUTS in men. Thus, given a potential role of the NO-pathway in the prostate and/or in other parts of lower urinary tract (e.g. bladder), the enhancement of the NO signaling by NO donor drugs, PDE5 inhibitors or activators of the soluble guanylyl cyclase (sGC) may represent a new therapeutic strategy for the treatment of LUTS. This review serves to focus on the role of NO and the NO-dependent signaling in the control of smooth muscle function in the human prostate. Results from clinical trials in men with LUTS/BPH are also discussed.
Collapse
|
38
|
Rabkin SW, Tsang MYC. The action of nitric oxide to enhance cell survival in chick cardiomyocytes is mediated through a cGMP and ERK1/2 pathway while p38 mitogen-activated protein kinase-dependent pathways do not alter cell death. Exp Physiol 2008; 93:834-42. [PMID: 18344257 DOI: 10.1113/expphysiol.2008.042176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The objective of this study was to determine whether the dual action of nitric oxide (NO) on cardiomyocyte cell viability is mediated through p38 mitogen-activated protein kinase (MAPK)-induced cell death and extracellular signal-regulated kinase (ERK1/2)-mediated cell survival pathways, and whether either of these is mediated through a cGMP-protein kinase G (PKG) pathway. Cell viability of embryonic chick cardiomyocytes was assessed by the MTT assay, which is based on the ability of viable cells to reduce 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide. The NO donor sodium nitroprusside (SNP) produced a significant (P < 0.01) concentration-dependent reduction in cell viability or increase in cell death. Sodium nitroprusside induced ERK1/2 phosphorylation, and the mitogen-activated protein kinase (MEK1/2) inhibitor PD 98059 significantly increased cell death. In contrast, SB202190, a relatively selective inhibitor of p38 MAPK, did not affect SNP-induced cell death. The cardioprotective effect of NO was prbably mediated in part via cGMP because 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a selective inhibitor of NO-sensitive guanylyl cyclase, produced a significant enhancement of SNP-induced cell death. In contrast, the PKG inhibitor KT5823 did not affect cell viability. In summary, these data suggest that NO, via stimulation of soluble guanylyl cyclase, activates MEK1/2 whose product, ERK1/2, protects against cell death. In contrast, SNP-induced p38 MAPK activation does not modulate NO-induced cardiomyocyte cell death. Not all cGMP targets affect NO-induced cell death, since the PKG pathway does not enhance or suppress NO-induced cardiomyocyte cell death. Enhancement of the ERK1/2 responses to NO may permit the beneficial effects of NO to predominate.
Collapse
Affiliation(s)
- Simon W Rabkin
- University of British Columbia, Level 9, 2775 Laurel Street, Vancouver, BC, Canada V5Z 1M9.
| | | |
Collapse
|
39
|
Role of neuronal nitric oxide synthase in the regulation of the neuroendocrine stress response in rodents: insights from mutant mice. Amino Acids 2008; 35:17-27. [DOI: 10.1007/s00726-007-0630-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Accepted: 10/31/2007] [Indexed: 02/04/2023]
|
40
|
Čokić VP, Schechter AN. Chapter 7 Effects of Nitric Oxide on Red Blood Cell Development and Phenotype. Curr Top Dev Biol 2008; 82:169-215. [DOI: 10.1016/s0070-2153(07)00007-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
41
|
Chen H, Levine YC, Golan DE, Michel T, Lin AJ. Atrial natriuretic peptide-initiated cGMP pathways regulate vasodilator-stimulated phosphoprotein phosphorylation and angiogenesis in vascular endothelium. J Biol Chem 2007; 283:4439-47. [PMID: 18079117 DOI: 10.1074/jbc.m709439200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nitric oxide (NO)- and atrial natriuretic peptide (ANP)-initiated cGMP signaling cascades are important in the maintenance of cardiovascular homeostasis. The molecular signaling mechanisms downstream of cGMP are not well understood, however. We have used small interfering RNA (siRNA) approaches to specifically knock down a series of signaling proteins in bovine aortic endothelial cells, and we have combined biochemical analyses with physiological assays to investigate cGMP-mediated signal transduction pathways. Activation of particulate guanylate cyclase (GC-A) by ANP leads to a substantial, dose-dependent, rapid, and sustained increase in intracellular cGMP. In contrast, stimulation of soluble guanylate cyclase by NO yields only a weak and transient increase in cGMP. ANP-induced cGMP production is selectively suppressed by siRNA-mediated knockdown of GC-A. ANP greatly enhances the phosphorylation at Ser-239 of the vasodilator-stimulated phosphoprotein (VASP), a major substrate of cGMP-dependent protein kinase (PKG) that significantly influences actin dynamics. Moreover, the ANP-induced phosphorylation of VASP at Ser-239 is accompanied by increased actin stress fiber formation and enhanced endothelial tube formation. siRNA-mediated knockdown of GC-A, VASP, or PKG abolishes ANP-induced VASP Ser-239 phosphorylation, stress fiber formation, and endothelial tube formation. We have demonstrated similar findings in human umbilical vein endothelial cells, where ANP substantially enhances intracellular cGMP content, phosphorylation of VASP at Ser-239, and endothelial tube formation. Taken together, our findings suggest that ANP-mediated cGMP signal transduction pathways regulate PKG phosphorylation of VASP Ser-239 in endothelial cells, resulting in reorganization of the actin cytoskeleton and enhancement of angiogenesis.
Collapse
Affiliation(s)
- Hongjie Chen
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
42
|
Green AK, Stratton RC, Squires PE, Simpson AWM. Atrial natriuretic peptide attenuates elevations in Ca2+ and protects hepatocytes by stimulating net plasma membrane Ca2+ efflux. J Biol Chem 2007; 282:34542-54. [PMID: 17893148 DOI: 10.1074/jbc.m707115200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Elevations in intracellular Ca(2+) concentration and calpain activity are common early events in cellular injury, including that of hepatocytes. Atrial natriuretic peptide is a circulating hormone that has been shown to be hepatoprotective. The aim of this study was to examine the effects of atrial natriuretic peptide on potentially harmful elevations in cytosolic free Ca(2+) and calpain activity induced by extracellular ATP in rat hepatocytes. We show that atrial natriuretic peptide, through protein kinase G, attenuated both the amplitude and duration of ATP-induced cytosolic Ca(2+) rises in single hepatocytes. Atrial natriuretic peptide also prevented stimulation of calpain activity by ATP, taurolithocholate, or Ca(2+) mobilization by thapsigargin and ionomycin. We therefore investigated the cellular Ca(2+) handling mechanisms through which ANP attenuates this sustained elevation in cytosolic Ca(2+). We show that atrial natriuretic peptide does not modulate the release from or re-uptake of Ca(2+) into intracellular stores but, through protein kinase G, both stimulates plasma membrane Ca(2+) efflux from and inhibits ATP-stimulated Ca(2+) influx into hepatocytes. These findings suggest that stimulation of net plasma membrane Ca(2+) efflux (to which both Ca(2+) efflux stimulation and Ca(2+) influx inhibition contribute) is the key process through which atrial natriuretic peptide attenuates elevations in cytosolic Ca(2+) and calpain activity. Moreover we propose that plasma membrane Ca(2+) efflux is a valuable, previously undiscovered, mechanism through which atrial natriuretic peptide protects rat hepatocytes, and perhaps other cell types, against Ca(2+)-dependent injury.
Collapse
Affiliation(s)
- Anne K Green
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry, UK.
| | | | | | | |
Collapse
|
43
|
PDE inhibitors in psychiatry--future options for dementia, depression and schizophrenia? Drug Discov Today 2007; 12:870-8. [PMID: 17933689 DOI: 10.1016/j.drudis.2007.07.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 07/21/2007] [Accepted: 07/23/2007] [Indexed: 02/07/2023]
Abstract
Phosphodiesterases are key enzymes in cellular signalling pathways. They degrade cyclic nucleotides and their inhibition via specific inhibitors offers unique 'receptor-independent' opportunities to modify cellular function. An increasing number of in vitro and animal model studies point to innovative treatment options in neurology and psychiatry. This review critiques a selection of recent studies and developments with a focus on dementia/neuroprotection, depression and schizophrenia. Despite increased interest among the clinical neurosciences, there are still no approved PDE inhibitors for clinical use in neurology or psychiatry. Adverse effects are a major impediment for clinical approval. It is therefore necessary to search for more specific inhibitors at the level of different PDE sub-families and isoforms.
Collapse
|
44
|
Abstract
Extracellular matrix (ECM) remodeling with successive tissue fibrosis is a key feature of chronic cardiovascular diseases, including atherosclerosis and restenosis. The atherogenic changes underlying these pathologies result from chronification of an acute repair response towards injurious and inflammatory stimuli. Thereby functional tissue is replaced by excessive ECM deposition. In the kidney, impaired remodeling is a major cause of perivascular, interstitial, and glomerular fibrosis but also a common complication of chronic hypertension. Experimental evidence points to the matrix metalloproteases (MMPs) and their intrinsic inhibitors, the tissue inhibitors of MMPs as key mediators of atherogenic and fibrotic pathologies. Mechanistically, a deregulation in ECM turnover tightly correlates with an increased production and release of proinflammatory and profibrotic factors including interleukin-1beta, transforming growth factor beta, angiotensin II, and reactive oxygen species. Unlike these factors the pleiotropic messenger molecule nitric oxide (NO) by acting as the major physiological vasodilator has emerged as one of the most atheroprotective factors. However, under inflammatory conditions NO does acquire proatherogenic and profibrotic properties thereby exacerbating tissue fibrosis. In this review, the mechanisms underlying both opposing properties of NO on perivascular ECM remodeling will exemplarily be discussed for renal fibrosis with a particular focus on the MMPs and intrinsic protease inhibitors.
Collapse
Affiliation(s)
- W Eberhardt
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7 Frankfurt am Main, Germany.
| | | |
Collapse
|
45
|
Akata T. Cellular and molecular mechanisms regulating vascular tone. Part 2: regulatory mechanisms modulating Ca2+ mobilization and/or myofilament Ca2+ sensitivity in vascular smooth muscle cells. J Anesth 2007; 21:232-42. [PMID: 17458653 DOI: 10.1007/s00540-006-0488-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Accepted: 11/21/2006] [Indexed: 11/25/2022]
Abstract
Understanding the physiological mechanisms regulating vascular tone would lead to better circulatory management during general anesthesia. This two-part review provides an overview of current knowledge about the cellular and molecular mechanisms regulating the contractile state of vascular smooth muscle cells (i.e., vascular tone). The first part reviews basic mechanisms controlling the cytosolic Ca2+ concentration in vascular smooth muscle cells, and the Ca2+-dependent regulation of vascular tone. This second part reviews the regulatory mechanisms modulating Ca2+ mobilization and/or myofilament Ca2+ sensitivity in vascular smooth muscle cells-including Rho/Rho kinase, protein kinase C, arachidonic acid, Ca2+/calmodulin-dependent protein kinase II, caldesmon, calponin, mitogen-activated protein kinases, tyrosine kinases, cyclic nucleotides, Cl- channels, and K+ channels.
Collapse
Affiliation(s)
- Takashi Akata
- Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan
| |
Collapse
|
46
|
Rehmann H, Wittinghofer A, Bos JL. Capturing cyclic nucleotides in action: snapshots from crystallographic studies. Nat Rev Mol Cell Biol 2007; 8:63-73. [PMID: 17183361 DOI: 10.1038/nrm2082] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fifty years ago, cyclic AMP was discovered as a second messenger of hormone action, heralding the age of signal transduction. Many cellular processes were found to be regulated by cAMP and the related cyclic GMP. Cyclic nucleotides function by binding to and activating their effectors - protein kinase A, protein kinase G, cyclic-nucleotide-regulated ion channels and the guanine nucleotide-exchange factor Epac. Recent structural insights have now made it possible to propose a general structural mechanism for how cyclic nucleotides regulate these proteins.
Collapse
Affiliation(s)
- Holger Rehmann
- Department of Physiological Chemistry and Centre for Biomedical Genetics, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| | | | | |
Collapse
|
47
|
Cook ALM, Haynes JM. Phosphorylation of the PKG substrate, vasodilator-stimulated phosphoprotein (VASP), in human cultured prostatic stromal cells. Nitric Oxide 2007; 16:10-7. [PMID: 17049286 DOI: 10.1016/j.niox.2006.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 08/06/2006] [Accepted: 09/05/2006] [Indexed: 11/29/2022]
Abstract
Nitric oxide (NO) is known to regulate contractility and proliferation of cells within the prostate, however, the mechanism by which this occurs is unknown. The cGMP-dependent protein kinase (PKG) signalling pathway may be involved, and recent work has shown that activation of this pathway can be assessed by analysis of phosphorylation of vasodilator-stimulated phosphoprotein (VASP). The aim of the current study is to characterise the expression of VASP in the human prostate and human cultured prostatic stromal cells (HCPSCs), and to investigate whether NO activates PKG in these cells. Our studies revealed that VASP is expressed, and that incubation of HCPSCs with PKG-activating cGMP-analogues or the NO-donor, SNP, caused a significant PKG-dependent increase in VASP serine-239 phosphorylation. In addition, SNP elicited a reduction in intracellular K(+) in a time frame consistent with the phosphorylation of VASP and activation of PKG. These data demonstrate that VASP can be used to assess the NO/cGMP/PKG signalling pathway in HCPSCs. In addition, we demonstrate for the first time that SNP, probably via NO release, leads to phosphorylation of VASP in a manner consistent with PKG activation.
Collapse
Affiliation(s)
- Anna-Louise M Cook
- School of Biomedical Sciences, Curtin University of Technology, Bentley, WA, USA
| | | |
Collapse
|
48
|
Ewing JF, Maines MD. Regulation and expression of heme oxygenase enzymes in aged-rat brain: age related depression in HO-1 and HO-2 expression and altered stress-response. J Neural Transm (Vienna) 2006; 113:439-54. [PMID: 16467964 DOI: 10.1007/s00702-005-0408-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Accepted: 09/26/2005] [Indexed: 10/25/2022]
Abstract
The heme oxygenase isozymes, HO-1 and HO-2, oxidatively cleave the heme molecule to produce biliverdin and the gaseous messenger, CO. The cleavage results in the release of iron, a regulator of transferrin, ferritin, and nitric oxide (NO) synthase gene expression. Biliverdin reductase (BVR) then catalyzes the reduction of biliverdin, generating the potent intracellular antioxidant, bilirubin. We report an age-related decrease in HO-1 and HO-2 expression present in select brain regions including the hippocampus and the substantia nigra, that are involved in the high order cognitive processes of learning and memory. The age-related loss of monoxide-producing potential in select regions of the brain was not specific to the HO system but was also observed in neuronal NO-generating system. Furthermore, compared to 2-month old rats, the ability of aged brain tissue to respond to hypoxic/hyperthermia was compromised at both the protein and the transcription levels as judged by attenuated induction of HO-1 immunoreactive protein and its 1.8 Kb transcript. Neotrofin (AIT), a cognitive-enhancing and neuroprotective drug, caused a robust increase in HO-1 immunoreactive protein in select neuronal regions and increased the expression of HO-2 transcripts. The potential interplay between regulation of HO-2 gene expression and the serum levels of the adrenal steroids is discussed. We suggest the search for therapeutic agents that reverse the decline and aberrant stress response of HO enzymes may lead to effective treatment regimens for age-associated neuronal deficits.
Collapse
Affiliation(s)
- J F Ewing
- Department of Biochemistry/Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | | |
Collapse
|
49
|
Kissmehl R, Krüger TP, Treptau T, Froissard M, Plattner H. Multigene family encoding 3',5'-cyclic-GMP-dependent protein kinases in Paramecium tetraurelia cells. EUKARYOTIC CELL 2006; 5:77-91. [PMID: 16400170 PMCID: PMC1360248 DOI: 10.1128/ec.5.1.77-91.2006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Accepted: 11/01/2005] [Indexed: 01/28/2023]
Abstract
In the ciliate Paramecium tetraurelia, 3',5'-cyclic GMP (cGMP) is one of the second messengers involved in several signal transduction pathways. The enzymes for its production and degradation are well established for these cells, whereas less is known about the potential effector proteins. On the basis of a current Paramecium genome project, we have identified a multigene family with at least 35 members, all of which encode cGMP-dependent protein kinases (PKGs). They can be classified into 16 subfamilies with several members each. Two of the genes, PKG1-1 and PKG2-1, were analyzed in more detail after molecular cloning. They encode monomeric enzymes of 770 and 819 amino acids, respectively, whose overall domain organization resembles that in higher eukaryotes. The enzymes contain a regulatory domain of two tandem cyclic nucleotide-binding sites flanked by an amino-terminal region for intracellular localization and a catalytic domain with highly conserved regions for ATP binding and catalysis. However, some Paramecium PKGs show a different structure. In Western blots, PKGs are detected both as cytosolic and as structure-bound forms. Immunofluorescence labeling shows enrichment in the cell cortex, notably around the dense-core secretory vesicles (trichocysts), as well as in cilia. Immunogold electron microscopy analysis reveals consistent labeling of ciliary membranes, of the membrane complex composed of cell membrane and cortical Ca2+ stores, and of regions adjacent to ciliary basal bodies, trichocysts, and trafficking vesicles. Since PKGs (re)phosphorylate the exocytosis-sensitive phosphoprotein pp63/pf upon stimulation, the role of PKGs during stimulated exocytosis is discussed, in addition to a role in ciliary beat regulation.
Collapse
Affiliation(s)
- Roland Kissmehl
- Department of Biology, University of Konstanz, P.O. Box 5560, 78457 Konstanz, Germany.
| | | | | | | | | |
Collapse
|
50
|
Guix FX, Uribesalgo I, Coma M, Muñoz FJ. The physiology and pathophysiology of nitric oxide in the brain. Prog Neurobiol 2005; 76:126-52. [PMID: 16115721 DOI: 10.1016/j.pneurobio.2005.06.001] [Citation(s) in RCA: 480] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 06/10/2005] [Accepted: 06/14/2005] [Indexed: 12/11/2022]
Abstract
Nitric oxide (NO) is a molecule with pleiotropic effects in different tissues. NO is synthesized by NO synthases (NOS), a family with four major types: endothelial, neuronal, inducible and mitochondrial. They can be found in almost all the tissues and they can even co-exist in the same tissue. NO is a well-known vasorelaxant agent, but it works as a neurotransmitter when produced by neurons and is also involved in defense functions when it is produced by immune and glial cells. NO is thermodynamically unstable and tends to react with other molecules, resulting in the oxidation, nitrosylation or nitration of proteins, with the concomitant effects on many cellular mechanisms. NO intracellular signaling involves the activation of guanylate cyclase but it also interacts with MAPKs, apoptosis-related proteins, and mitochondrial respiratory chain or anti-proliferative molecules. It also plays a role in post-translational modification of proteins and protein degradation by the proteasome. However, under pathophysiological conditions NO has damaging effects. In disorders involving oxidative stress, such as Alzheimer's disease, stroke and Parkinson's disease, NO increases cell damage through the formation of highly reactive peroxynitrite. The paradox of beneficial and damaging effects of NO will be discussed in this review.
Collapse
Affiliation(s)
- F X Guix
- Laboratori de Fisiologia Molecular, Unitat de Senyalització Cellular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Carrer Dr. Aiguader, 80, Barcelona 08003, Spain
| | | | | | | |
Collapse
|