1
|
De Jong KA, Siddig S, Pfeifer A, Nikolaev VO. The role of compartmentalized β-AR/cAMP signaling in the regulation of lipolysis in white and brown adipocytes. FEBS J 2025; 292:261-271. [PMID: 38747241 PMCID: PMC11734871 DOI: 10.1111/febs.17157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/01/2024] [Accepted: 04/30/2024] [Indexed: 01/16/2025]
Abstract
White and brown adipocytes are central mediators of lipid metabolism and thermogenesis, respectively. Their function is tightly regulated by all three β-adrenergic receptor (β-AR) subtypes which are coupled to the production of the second messenger 3',5'-cyclic adenosine monophosphate (cAMP). While known for decades in other cell types, compartmentation of adipocyte β-AR/cAMP signaling by spatial organization of the pathway and by cAMP degrading phosphodiesterases (PDEs) as well as its role in the regulation of lipolysis is only beginning to emerge. Here, we provide a short overview of recent findings which shed light on compartmentalized signaling using live cell imaging of cAMP in adipocytes and discuss possible future directions of research which could open up new avenues for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Kirstie A. De Jong
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfGermany
| | - Sana Siddig
- Institute of Pharmacology and Toxicology, University Hospital BonnUniversity of BonnGermany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital BonnUniversity of BonnGermany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfGermany
| |
Collapse
|
2
|
Krause PN, McGeorge G, McPeek JL, Khalid S, Nelin LD, Liu Y, Chen B. Pde3a and Pde3b regulation of murine pulmonary artery smooth muscle cell growth and metabolism. Physiol Rep 2024; 12:e70089. [PMID: 39435735 PMCID: PMC11494452 DOI: 10.14814/phy2.70089] [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: 09/05/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/23/2024] Open
Abstract
A role for metabolically active adipose tissue in pulmonary hypertension (PH) pathogenesis is emerging. Alterations in cellular metabolism in metabolic syndrome are triggers of PH-related vascular dysfunction. Metabolic reprogramming in proliferative pulmonary vascular cells causes a metabolic switch from oxidative phosphorylation to glycolysis. PDE3A and PDE3B subtypes in the regulation of metabolism in pulmonary artery smooth muscle cells (PASMC) are poorly understood. We previously found that PDE3A modulates the cellular energy sensor, AMPK, in human PASMC. We demonstrate that global Pde3a knockout mice have right ventricular (RV) hypertrophy, elevated RV systolic pressures, and metabolic dysfunction with elevated serum free fatty acids (FFA). Therefore, we sought to delineate Pde3a/Pde3b regulation of metabolic pathways in PASMC. We found that PASMC Pde3a deficiency, and to a lesser extent Pde3b deficiency, downregulates AMPK, CREB and PPARγ, and upregulates pyruvate kinase dehydrogenase expression, suggesting decreased oxidative phosphorylation. Interestingly, siRNA Pde3a knockdown in adipocytes led to elevated FFA secretion. Furthermore, PASMC exposed to siPDE3A-transfected adipocyte media led to decreased α-SMA, AMPK and CREB phosphorylation, and greater viable cell numbers compared to controls under the same conditions. These data demonstrate that deficiencies of Pde3a and Pde3b alter pathways that affect cell growth and metabolism in PASMC.
Collapse
MESH Headings
- Animals
- Male
- Mice
- AMP-Activated Protein Kinases/metabolism
- AMP-Activated Protein Kinases/genetics
- Cell Proliferation
- Cells, Cultured
- Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 3/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/cytology
Collapse
Affiliation(s)
- Paulina N. Krause
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Gabrielle McGeorge
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Jennifer L. McPeek
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Sidra Khalid
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Leif D. Nelin
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Yusen Liu
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Bernadette Chen
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| |
Collapse
|
3
|
Menges L, Giesen J, Yilmaz K, Mergia E, Füchtbauer A, Füchtbauer EM, Koesling D, Russwurm M. It takes two to tango: cardiac fibroblast-derived NO-induced cGMP enters cardiac myocytes and increases cAMP by inhibiting PDE3. Commun Biol 2023; 6:504. [PMID: 37165086 PMCID: PMC10172304 DOI: 10.1038/s42003-023-04880-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
The occurrence of NO/cGMP signalling in cardiac cells is a matter of debate. Recent measurements with a FRET-based cGMP indicator in isolated cardiac cells revealed NO-induced cGMP signals in cardiac fibroblasts while cardiomyocytes were devoid of these signals. In a fibroblast/myocyte co-culture model though, cGMP formed in fibroblasts in response to NO entered cardiomyocytes via gap junctions. Here, we demonstrate gap junction-mediated cGMP transfer from cardiac fibroblasts to myocytes in intact tissue. In living cardiac slices of mice with cardiomyocyte-specific expression of a FRET-based cGMP indicator (αMHC/cGi-500), NO-dependent cGMP signals were shown to occur in myocytes, to depend on gap junctions and to be degraded mainly by PDE3. Stimulation of NO-sensitive guanylyl cyclase enhanced Forskolin- and Isoproterenol-induced cAMP and phospholamban phosphorylation. Genetic inactivation of NO-GC in Tcf21-expressing cardiac fibroblasts abrogated the synergistic action of NO-GC stimulation on Iso-induced phospholamban phosphorylation, identifying fibroblasts as cGMP source and substantiating the necessity of cGMP-transfer to myocytes. In sum, NO-stimulated cGMP formed in cardiac fibroblasts enters cardiomyocytes in native tissue where it exerts an inhibitory effect on cAMP degradation by PDE3, thereby increasing cAMP and downstream effects in cardiomyocytes. Hence, enhancing β-receptor-induced contractile responses appears as one of NO/cGMP's functions in the non-failing heart.
Collapse
Affiliation(s)
- Lukas Menges
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Jan Giesen
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Kerem Yilmaz
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Evanthia Mergia
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Annette Füchtbauer
- Department of Molecular Biology and Genetics, Aarhus University, 8000, Aarhus C, Denmark
| | | | - Doris Koesling
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Michael Russwurm
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780, Bochum, Germany.
| |
Collapse
|
4
|
Chen J, Liu N, Huang Y, Wang Y, Sun Y, Wu Q, Li D, Gao S, Wang HW, Huang N, Qi X, Wang X. Structure of PDE3A-SLFN12 complex and structure-based design for a potent apoptosis inducer of tumor cells. Nat Commun 2021; 12:6204. [PMID: 34707099 PMCID: PMC8551160 DOI: 10.1038/s41467-021-26546-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
Molecular glues are a class of small molecular drugs that mediate protein-protein interactions, that induce either the degradation or stabilization of target protein. A structurally diverse group of chemicals, including 17-β-estradiol (E2), anagrelide, nauclefine, and DNMDP, induces apoptosis by forming complexes with phosphodiesterase 3A (PDE3A) and Schlafen 12 protein (SLFN12). They do so by binding to the PDE3A enzymatic pocket that allows the compound-bound PDE3A to recruit and stabilize SLFN12, which in turn blocks protein translation, leading to apoptosis. In this work, we report the high-resolution cryo-electron microscopy structure of PDE3A-SLFN12 complexes isolated from cultured HeLa cells pre-treated with either anagrelide, or nauclefine, or DNMDP. The PDE3A-SLFN12 complexes exhibit a butterfly-like shape, forming a heterotetramer with these small molecules, which are packed in a shallow pocket in the catalytic domain of PDE3A. The resulting small molecule-modified interface binds to the short helix (E552-I558) of SLFN12 through hydrophobic interactions, thus "gluing" the two proteins together. Based on the complex structure, we designed and synthesized analogs of anagrelide, a known drug used for the treatment of thrombocytosis, to enhance their interactions with SLFN12, and achieved superior efficacy in inducing apoptosis in cultured cells as well as in tumor xenografts.
Collapse
Affiliation(s)
- Jie Chen
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Nan Liu
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yinpin Huang
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 100084, Beijing, China
| | - Yuanxun Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Yuxing Sun
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Qingcui Wu
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Dianrong Li
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Shuanhu Gao
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, 3663N Zhongshan Road, Shanghai, 200062, China
| | - Hong-Wei Wang
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Niu Huang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 100084, Beijing, China.
| | - Xiangbing Qi
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 100084, Beijing, China.
| | - Xiaodong Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
5
|
Vinogradova TM, Lakatta EG. Dual Activation of Phosphodiesterase 3 and 4 Regulates Basal Cardiac Pacemaker Function and Beyond. Int J Mol Sci 2021. [PMID: 34445119 DOI: 10.3390/ijms22168414.pmid:34445119;pmcid:pmc8395138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
The sinoatrial (SA) node is the physiological pacemaker of the heart, and resting heart rate in humans is a well-known risk factor for cardiovascular disease and mortality. Consequently, the mechanisms of initiating and regulating the normal spontaneous SA node beating rate are of vital importance. Spontaneous firing of the SA node is generated within sinoatrial nodal cells (SANC), which is regulated by the coupled-clock pacemaker system. Normal spontaneous beating of SANC is driven by a high level of cAMP-mediated PKA-dependent protein phosphorylation, which rely on the balance between high basal cAMP production by adenylyl cyclases and high basal cAMP degradation by cyclic nucleotide phosphodiesterases (PDEs). This diverse class of enzymes includes 11 families and PDE3 and PDE4 families dominate in both the SA node and cardiac myocardium, degrading cAMP and, consequently, regulating basal cardiac pacemaker function and excitation-contraction coupling. In this review, we will demonstrate similarities between expression, distribution, and colocalization of various PDE subtypes in SANC and cardiac myocytes of different species, including humans, focusing on PDE3 and PDE4. Here, we will describe specific targets of the coupled-clock pacemaker system modulated by dual PDE3 + PDE4 activation and provide evidence that concurrent activation of PDE3 + PDE4, operating in a synergistic manner, regulates the basal cardiac pacemaker function and provides control over normal spontaneous beating of SANCs through (PDE3 + PDE4)-dependent modulation of local subsarcolemmal Ca2+ releases (LCRs).
Collapse
Affiliation(s)
- Tatiana M Vinogradova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institute of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institute of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| |
Collapse
|
6
|
Dual Activation of Phosphodiesterase 3 and 4 Regulates Basal Cardiac Pacemaker Function and Beyond. Int J Mol Sci 2021; 22:ijms22168414. [PMID: 34445119 PMCID: PMC8395138 DOI: 10.3390/ijms22168414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
The sinoatrial (SA) node is the physiological pacemaker of the heart, and resting heart rate in humans is a well-known risk factor for cardiovascular disease and mortality. Consequently, the mechanisms of initiating and regulating the normal spontaneous SA node beating rate are of vital importance. Spontaneous firing of the SA node is generated within sinoatrial nodal cells (SANC), which is regulated by the coupled-clock pacemaker system. Normal spontaneous beating of SANC is driven by a high level of cAMP-mediated PKA-dependent protein phosphorylation, which rely on the balance between high basal cAMP production by adenylyl cyclases and high basal cAMP degradation by cyclic nucleotide phosphodiesterases (PDEs). This diverse class of enzymes includes 11 families and PDE3 and PDE4 families dominate in both the SA node and cardiac myocardium, degrading cAMP and, consequently, regulating basal cardiac pacemaker function and excitation-contraction coupling. In this review, we will demonstrate similarities between expression, distribution, and colocalization of various PDE subtypes in SANC and cardiac myocytes of different species, including humans, focusing on PDE3 and PDE4. Here, we will describe specific targets of the coupled-clock pacemaker system modulated by dual PDE3 + PDE4 activation and provide evidence that concurrent activation of PDE3 + PDE4, operating in a synergistic manner, regulates the basal cardiac pacemaker function and provides control over normal spontaneous beating of SANCs through (PDE3 + PDE4)-dependent modulation of local subsarcolemmal Ca2+ releases (LCRs).
Collapse
|
7
|
Dillard J, Meng X, Nelin L, Liu Y, Chen B. Nitric oxide activates AMPK by modulating PDE3A in human pulmonary artery smooth muscle cells. Physiol Rep 2021; 8:e14559. [PMID: 32914566 PMCID: PMC7507575 DOI: 10.14814/phy2.14559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 01/17/2023] Open
Abstract
Phosphodiesterase 3 (PDE3), of which there are two isoforms, PDE3A and PDE3B, hydrolyzes cAMP and cGMP—cyclic nucleotides important in the regulation of pulmonary vascular tone. PDE3 has been implicated in pulmonary hypertension unresponsive to nitric oxide (NO); however, contributions of the two isoforms are not known. Furthermore, adenosine monophosphate‐activated protein kinase (AMPK), a critical regulator of cellular energy homeostasis, has been shown to be modulated by PDE3 in varying cell types. While AMPK has recently been implicated in pulmonary hypertension pathogenesis, its role and regulation in the pulmonary vasculature remain to be elucidated. Therefore, we utilized human pulmonary artery smooth muscle cells (hPASMC) to test the hypothesis that NO increases PDE3 expression in an isoform‐specific manner, thereby activating AMPK and inhibiting hPASMC proliferation. We found that in hPASMC, NO treatment increased PDE3A protein expression and PDE3 activity with a concomitant decrease in cAMP concentrations and increase in AMPK phosphorylation. Knockdown of PDE3A using siRNA transfection blunted the NO‐induced AMPK activation, indicating that PDE3A plays an important role in AMPK regulation in hPASMC. Treatment with a soluble guanylate cyclase (sGC) stimulator increased PDE3A expression and AMPK activation similar to that seen with NO treatment, whereas treatment with a sGC inhibitor blunted the NO‐induced increase in PDE3A and AMPK activation. These results suggest that NO increases PDE3A expression, decreases cAMP, and activates AMPK via the sGC‐cGMP pathway. We speculate that NO‐induced increases in PDE3A and AMPK may have implications in the pathogenesis and the response to therapies in pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- Julie Dillard
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Xiaomei Meng
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Leif Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
8
|
Ai Y, He H, Chen P, Yan B, Zhang W, Ding Z, Li D, Chen J, Ma Y, Cao Y, Zhu J, Li J, Ou J, Du S, Wang X, Ma J, Gao S, Qi X. An alkaloid initiates phosphodiesterase 3A-schlafen 12 dependent apoptosis without affecting the phosphodiesterase activity. Nat Commun 2020; 11:3236. [PMID: 32591543 PMCID: PMC7319972 DOI: 10.1038/s41467-020-17052-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
The promotion of apoptosis in tumor cells is a popular strategy for developing anti-cancer drugs. Here, we demonstrate that the plant indole alkaloid natural product nauclefine induces apoptosis of diverse cancer cells via a PDE3A-SLFN12 dependent death pathway. Nauclefine binds PDE3A but does not inhibit the PDE3A's phosphodiesterase activity, thus representing a previously unknown type of PDE3A modulator that can initiate apoptosis without affecting PDE3A's canonical function. We demonstrate that PDE3A's H840, Q975, Q1001, and F1004 residues-as well as I105 in SLFN12-are essential for nauclefine-induced PDE3A-SLFN12 interaction and cell death. Extending these molecular insights, we show in vivo that nauclefine inhibits tumor xenograft growth, doing so in a PDE3A- and SLFN12-dependent manner. Thus, beyond demonstrating potent cytotoxic effects of an alkaloid natural product, our study illustrates a potentially side-effect-reducing strategy for targeting PDE3A for anti-cancer therapeutics without affecting its phosphodiesterase activity.
Collapse
Affiliation(s)
- Youwei Ai
- College of Wildlife and Protected Area, Northeast Forestry University, Hexing Road, 150040, Harbin, China.
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| | - Haibing He
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, 3663N Zhongshan Road, 200062, Shanghai, China
| | - Peihao Chen
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Bo Yan
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Wenbin Zhang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Zhangcheng Ding
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Dianrong Li
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Jie Chen
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
| | - Yan Ma
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
| | - Yang Cao
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
| | - Jie Zhu
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
| | - Jiaojiao Li
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
| | - Jinjie Ou
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, 3663N Zhongshan Road, 200062, Shanghai, China
| | - Shan Du
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, 3663N Zhongshan Road, 200062, Shanghai, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Jianzhang Ma
- College of Wildlife and Protected Area, Northeast Forestry University, Hexing Road, 150040, Harbin, China.
| | - Shuanhu Gao
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, 3663N Zhongshan Road, 200062, Shanghai, China.
| | - Xiangbing Qi
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, 102206, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
9
|
Shekouhy M, Karimian S, Moaddeli A, Faghih Z, Delshad Y, Khalafi-Nezhad A. The synthesis and biological evaluation of nucleobases/tetrazole hybrid compounds: A new class of phosphodiesterase type 3 (PDE3) inhibitors. Bioorg Med Chem 2020; 28:115540. [PMID: 32503691 DOI: 10.1016/j.bmc.2020.115540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/22/2020] [Accepted: 04/29/2020] [Indexed: 01/12/2023]
Abstract
Spired by the chemical structure of Cilostazol, a selective phosphodiesterase 3A (PDE3A) inhibitor, several novel hybrid compounds of nucleobases (uracil, 6-azauracil, 2-thiuracil, adenine, guanine, theophylline and theobromine) and tetrazole were designed and successfully synthesized and their inhibitory effects on PDE3A as well as their cytotoxicity on HeLa and MCF-7 cancerous cell lines were studied. Obtained results show the linear correlation between the inhibitory effect of synthesized compounds and their cytotoxicity. In some cases, the PDE3A inhibitory effects of synthesized compounds are higher than the Cilostazol. Besides, compared to a standard anticancer drug methotrexate, some of the synthesized compounds showed the higher cytotoxicity against the HeLa and MCF-7 cancerous cell lines.
Collapse
Affiliation(s)
- Mohsen Shekouhy
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran.
| | - Somaye Karimian
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Moaddeli
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran; Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Zeinab Faghih
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yousef Delshad
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran
| | - Ali Khalafi-Nezhad
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran.
| |
Collapse
|
10
|
Dillard J, Perez M, Chen B. Therapies that enhance pulmonary vascular NO-signaling in the neonate. Nitric Oxide 2019; 95:45-54. [PMID: 31870967 DOI: 10.1016/j.niox.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/25/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
There are several pulmonary hypertensive diseases that affect the neonatal population, including persistent pulmonary hypertension of the newborn (PPHN) and bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH). While the indication for inhaled nitric oxide (iNO) use is for late-preterm and term neonates with PPHN, there is a suboptimal response to this pulmonary vasodilator in ~40% of patients. Additionally, there are no FDA-approved treatments for BPD-associated PH or for preterm infants with PH. Therefore, investigating mechanisms that alter the nitric oxide-signaling pathway has been at the forefront of pulmonary vascular biology research. In this review, we will discuss the various mechanistic pathways that have been targets in neonatal PH, including NO precursors, soluble guanylate cyclase modulators, phosphodiesterase inhibitors and antioxidants. We will review their role in enhancing NO-signaling at the bench, in animal models, as well as highlight their role in the treatment of neonates with PH.
Collapse
Affiliation(s)
- Julie Dillard
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Marta Perez
- Division of Neonatology, Stanley Manne Children's Research Institute, Ann and Robert H Lurie Children's Hospital, Chicago, IL, USA; Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
11
|
Baillie GS, Tejeda GS, Kelly MP. Therapeutic targeting of 3',5'-cyclic nucleotide phosphodiesterases: inhibition and beyond. Nat Rev Drug Discov 2019; 18:770-796. [PMID: 31388135 PMCID: PMC6773486 DOI: 10.1038/s41573-019-0033-4] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2019] [Indexed: 01/24/2023]
Abstract
Phosphodiesterases (PDEs), enzymes that degrade 3',5'-cyclic nucleotides, are being pursued as therapeutic targets for several diseases, including those affecting the nervous system, the cardiovascular system, fertility, immunity, cancer and metabolism. Clinical development programmes have focused exclusively on catalytic inhibition, which continues to be a strong focus of ongoing drug discovery efforts. However, emerging evidence supports novel strategies to therapeutically target PDE function, including enhancing catalytic activity, normalizing altered compartmentalization and modulating post-translational modifications, as well as the potential use of PDEs as disease biomarkers. Importantly, a more refined appreciation of the intramolecular mechanisms regulating PDE function and trafficking is emerging, making these pioneering drug discovery efforts tractable.
Collapse
Affiliation(s)
- George S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Gonzalo S Tejeda
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
12
|
Vinogradova TM, Sirenko S, Lukyanenko YO, Yang D, Tarasov KV, Lyashkov AE, Varghese NJ, Li Y, Chakir K, Ziman B, Lakatta EG. Basal Spontaneous Firing of Rabbit Sinoatrial Node Cells Is Regulated by Dual Activation of PDEs (Phosphodiesterases) 3 and 4. Circ Arrhythm Electrophysiol 2019; 11:e005896. [PMID: 29880528 DOI: 10.1161/circep.117.005896] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 03/27/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Spontaneous firing of sinoatrial node cells (SANCs) is regulated by cAMP-mediated, PKA (protein kinase A)-dependent (cAMP/PKA) local subsarcolemmal Ca2+ releases (LCRs) from RyRs (ryanodine receptors). LCRs occur during diastolic depolarization and activate an inward Na+/Ca2+ exchange current that accelerates diastolic depolarization rate prompting the next action potential. PDEs (phosphodiesterases) regulate cAMP-mediated signaling; PDE3/PDE4 represent major PDE activities in SANC, but how they modulate LCRs and basal spontaneous SANC firing remains unknown. METHODS Real-time polymerase chain reaction, Western blot, immunostaining, cellular perforated patch clamping, and confocal microscopy were used to elucidate mechanisms of PDE-dependent regulation of cardiac pacemaking. RESULTS PDE3A, PDE4B, and PDE4D were the major PDE subtypes expressed in rabbit SANC, and PDE3A was colocalized with α-actinin, PDE4D, SERCA (sarcoplasmic reticulum Ca2+ ATP-ase), and PLB (phospholamban) in Z-lines. Inhibition of PDE3 (cilostamide) or PDE4 (rolipram) alone increased spontaneous SANC firing by ≈20% (P<0.05) and ≈5% (P>0.05), respectively, but concurrent PDE3+PDE4 inhibition increased spontaneous firing by ≈45% (P<0.01), indicating synergistic effect. Inhibition of PDE3 or PDE4 alone increased L-type Ca2+ current (ICa,L) by ≈60% (P<0.01) or ≈5% (P>0.05), respectively, and PLB phosphorylation by ≈20% (P>0.05) each, but dual PDE3+PDE4 inhibition increased ICa,L by ≈100% (P<0.01) and PLB phosphorylation by ≈110% (P<0.05). Dual PDE3+PDE4 inhibition increased the LCR number and size (P<0.01) and reduced the SR (sarcoplasmic reticulum) Ca2+ refilling time (P<0.01) and the LCR period (time from action potential-induced Ca2+ transient to subsequent LCR; P<0.01), leading to decrease in spontaneous SANC cycle length (P<0.01). When RyRs were disabled by ryanodine and LCRs ceased, dual PDE3+PDE4 inhibition failed to increase spontaneous SANC firing. CONCLUSIONS Basal cardiac pacemaker function is regulated by concurrent PDE3+PDE4 activation which operates in a synergistic manner via decrease in cAMP/PKA phosphorylation, suppression of LCR parameters, and prolongation of the LCR period and spontaneous SANC cycle length.
Collapse
Affiliation(s)
- Tatiana M Vinogradova
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD.
| | - Syevda Sirenko
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Yevgeniya O Lukyanenko
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Dongmei Yang
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Kirill V Tarasov
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Alexey E Lyashkov
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Nevin J Varghese
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Yue Li
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Khalid Chakir
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Bruce Ziman
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD
| |
Collapse
|
13
|
Kim NJ, Baek JH, Lee J, Kim H, Song JK, Chun KH. A PDE1 inhibitor reduces adipogenesis in mice via regulation of lipolysis and adipogenic cell signaling. Exp Mol Med 2019; 51:1-15. [PMID: 30635550 PMCID: PMC6329698 DOI: 10.1038/s12276-018-0198-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 09/03/2018] [Accepted: 09/11/2018] [Indexed: 12/15/2022] Open
Abstract
Vinpocetine, a phosphodiesterase (PDE) type-1 inhibitor, increases cAMP and cGMP levels and is currently used for the management of cerebrovascular disorders, such as stroke, cerebral hemorrhage, and cognitive dysfunctions. In this study, we first determined that vinpocetine effectively suppressed adipogenesis and lipid accumulation. However, we questioned which molecular mechanism is involved because the role of PDE in adipogenesis is still controversial. Vinpocetine decreased adipogenic cell signaling, including the phosphorylation of ERK, AKT, JAK2, and STAT3, and adipokine secretion, including IL-6, IL-10, and IFN-α. Interestingly, vinpocetine increased the phosphorylation of HSL, suggesting the induction of the lipolysis pathway. Moreover, vinpocetine increased UCP1 expression via increasing cAMP and PKA phosphorylation. The administration of vinpocetine with a normal-chow diet (NFD) or a high-fat diet (HFD) in mice attenuated body weight gain in mice fed both the NFD and HFD. These effects were larger in the HFD-fed mice, without a difference in food intake. Vinpocetine drastically decreased fat weight and adipocyte cell sizes in gonadal and inguinal white adipose tissues and in the liver in both diet groups. Serum triacylglycerol levels and fasting blood glucose levels were reduced by vinpocetine treatment. This study suggested that vinpocetine prevents adipocyte differentiation through the inhibition of adipogenesis-associated cell signaling in the early stages of adipogenesis. Moreover, upregulating cAMP levels leads to an increase in lipolysis and UCP1 expression and then inhibits lipid accumulation. Therefore, we suggest that vinpocetine could be an effective agent for treating obesity, as well as improving cognition and cardiovascular function in older individuals. A compound extracted from the periwinkle plant can limit the over-production of fat cells and may be a useful agent for treating obesity. Being overweight is the result of changes in the size and number of fat cells, or adipocytes, in the body. Scientists are searching for molecules that can limit the growth and replication of adipocytes, but many anti-obesity agents found to date have unpleasant side-effects. Kyung-Hee Chun at Yonsei University in Seoul, South Korea and co-workers examined the effects of 502 naturally occuring compounds on adipocyte differentiation in cell culture. One compound called vincamine, which is safely used to treat vascular diseases in the brain, decreased cell signaling pathways involved in adipocyte generation in mice and also lowered fasting blood glucose levels.
Collapse
Affiliation(s)
- Nam-Jun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Jung-Hwan Baek
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - JinAh Lee
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - HyeNa Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.,Tumor Microenvironment Research Branch, Division of Cancer Biology, National Cancer Center, Goyang, Republic of Korea
| | - Jun-Kyu Song
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Kyung-Hee Chun
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Vinogradova TM, Kobrinsky E, Lakatta EG. Dual Activation of Phosphodiesterases 3 and 4 Regulates Basal Spontaneous Beating Rate of Cardiac Pacemaker Cells: Role of Compartmentalization? Front Physiol 2018; 9:1301. [PMID: 30356755 PMCID: PMC6189467 DOI: 10.3389/fphys.2018.01301] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/29/2018] [Indexed: 12/17/2022] Open
Abstract
Spontaneous firing of sinoatrial (SA) node cells (SANCs) is regulated by cyclic adenosine monophosphate (cAMP)-mediated, protein kinase A (PKA)-dependent (cAMP/PKA) local subsarcolemmal Ca2+ releases (LCRs) from ryanodine receptors (RyR). The LCRs occur during diastolic depolarization (DD) and activate an inward Na+/Ca2+ exchange current that accelerates the DD rate prompting the next action potential (AP). Basal phosphodiesterases (PDEs) activation degrades cAMP, reduces basal cAMP/PKA-dependent phosphorylation, and suppresses normal spontaneous firing of SANCs. The cAMP-degrading PDE1, PDE3, and PDE4 represent major PDE activities in rabbit SANC, and PDE inhibition by 3-isobutyl-1-methylxanthine (IBMX) increases spontaneous firing of SANC by ∼50%. Though inhibition of single PDE1–PDE4 only moderately increases spontaneous SANC firing, dual PDE3 + PDE4 inhibition produces a synergistic effect hastening the spontaneous SANC beating rate by ∼50%. Here, we describe the expression and distribution of different PDE subtypes within rabbit SANCs, several specific targets (L-type Ca2+ channels and phospholamban) regulated by basal concurrent PDE3 + PDE4 activation, and critical importance of RyR Ca2+ releases for PDE-dependent regulation of spontaneous SANC firing. Colocalization of PDE3 and PDE4 beneath sarcolemma or in striated patterns inside SANCs strongly suggests that PDE-dependent regulation of cAMP/PKA signaling might be executed at the local level; this idea, however, requires further verification.
Collapse
Affiliation(s)
- Tatiana M Vinogradova
- Laboratory of Cardiovascular Science, Intramural Research Program, NIA, NIH, Baltimore, MD, United States
| | - Evgeny Kobrinsky
- Laboratory of Cardiovascular Science, Intramural Research Program, NIA, NIH, Baltimore, MD, United States
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, NIA, NIH, Baltimore, MD, United States
| |
Collapse
|
15
|
Ahsan MK, Tchernychev B, Kessler MM, Solinga RM, Arthur D, Linde CI, Silos-Santiago I, Hannig G, Ameen NA. Linaclotide activates guanylate cyclase-C/cGMP/protein kinase-II-dependent trafficking of CFTR in the intestine. Physiol Rep 2018; 5:5/11/e13299. [PMID: 28592587 PMCID: PMC5471438 DOI: 10.14814/phy2.13299] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/10/2017] [Accepted: 05/03/2017] [Indexed: 12/12/2022] Open
Abstract
The transmembrane receptor guanylyl cyclase‐C (GC‐C), expressed on enterocytes along the intestine, is the molecular target of the GC‐C agonist peptide linaclotide, an FDA‐approved drug for treatment of adult patients with Irritable Bowel Syndrome with Constipation and Chronic Idiopathic Constipation. Polarized human colonic intestinal cells (T84, CaCo‐2BBe) rat and human intestinal tissues were employed to examine cellular signaling and cystic fibrosis transmembrane conductance regulator (CFTR)‐trafficking pathways activated by linaclotide using confocal microscopy, in vivo surface biotinylation, and protein kinase‐II (PKG‐II) activity assays. Expression and activity of GC‐C/cGMP pathway components were determined by PCR, western blot, and cGMP assays. Fluid secretion as a marker of CFTR cell surface translocation was determined using in vivo rat intestinal loops. Linaclotide treatment (30 min) induced robust fluid secretion and translocation of CFTR from subapical compartments to the cell surface in rat intestinal loops. Similarly, linaclotide treatment (30 min) of T84 and CaCo‐2BBe cells increased cell surface CFTR levels. Linaclotide‐induced activation of the GC‐C/cGMP/PKGII signaling pathway resulted in elevated intracellular cGMP and pVASPser239 phosphorylation. Inhibition or silencing of PKGII significantly attenuated linaclotide‐induced CFTR trafficking to the apical membrane. Inhibition of protein kinase‐A (PKA) also attenuated linaclotide‐induced CFTR cell surface trafficking, implying cGMP‐dependent cross‐activation of PKA pathway. Together, these findings support linaclotide‐induced activation of the GC‐C/cGMP/PKG‐II/CFTR pathway as the major pathway of linaclotide‐mediated intestinal fluid secretion, and that linaclotide‐dependent CFTR activation and recruitment/trafficking of CFTR from subapical vesicles to the cell surface is an important step in this process.
Collapse
Affiliation(s)
- Md Kaimul Ahsan
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut
| | - Boris Tchernychev
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | - Marco M Kessler
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | - Robert M Solinga
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | | | | | | | - Gerhard Hannig
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | - Nadia A Ameen
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut .,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
16
|
Sharma S, Visweswariah SS. Illuminating Cyclic Nucleotides: Sensors for cAMP and cGMP and Their Application in Live Cell Imaging. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0014-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
17
|
Richards JP, Bowles EA, Gordon WR, Ellsworth ML, Stephenson AH, Sprague RS. Mechanisms of C-peptide-mediated rescue of low O2-induced ATP release from erythrocytes of humans with type 2 diabetes. Am J Physiol Regul Integr Comp Physiol 2014; 308:R411-8. [PMID: 25552662 DOI: 10.1152/ajpregu.00420.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The circulating erythrocyte, by virtue of the regulated release of ATP in response to reduced oxygen (O2) tension, plays a key role in maintaining appropriate perfusion distribution to meet tissue needs. Erythrocytes from individuals with Type 2 diabetes (DM2) fail to release ATP in response to this stimulus. However, the administration of C-peptide and insulin at a 1:1 ratio was shown to restore this important physiological response in humans with DM2. To begin to investigate the mechanisms by which C-peptide influences low O2-induced ATP release, erythrocytes from healthy humans and humans with DM2 were exposed to reduced O2 in a thin-film tonometer, and ATP release under these conditions was compared with release during normoxia. We determined that 1) low O2-induced ATP release from DM2 erythrocytes is rescued by C-peptide in the presence and absence of insulin, 2) the signaling pathway activated by C-peptide in human erythrocytes involves PKC, as well as soluble guanylyl cyclase (sGC) and 3) inhibitors of cGMP degradation rescue low O2-induced ATP release from DM2 erythrocytes. These results provide support for the hypothesis that both PKC and sGC are components of a signaling pathway activated by C-peptide in human erythrocytes. In addition, since both C-peptide and phosphodiesterase 5 inhibitors rescue low O2-induced ATP release from erythrocytes of humans with DM2, their administration to humans with DM2 could aid in the treatment and/or prevention of the vascular disease associated with this condition.
Collapse
Affiliation(s)
- Jennifer P Richards
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Elizabeth A Bowles
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Weston R Gordon
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Mary L Ellsworth
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Alan H Stephenson
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Randy S Sprague
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
18
|
Davari AS, Abnous K, Mehri S, Ghandadi M, Hadizadeh F. Synthesis and biological evaluation of novel pyridine derivatives as potential anticancer agents and phosphodiesterase-3 inhibitors. Bioorg Chem 2014; 57:83-89. [DOI: 10.1016/j.bioorg.2014.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/28/2014] [Accepted: 09/08/2014] [Indexed: 01/04/2023]
|
19
|
Conti M, Mika D, Richter W. Cyclic AMP compartments and signaling specificity: role of cyclic nucleotide phosphodiesterases. ACTA ACUST UNITED AC 2014; 143:29-38. [PMID: 24378905 PMCID: PMC3874571 DOI: 10.1085/jgp.201311083] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Marco Conti
- Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143
| | | | | |
Collapse
|
20
|
Huang YY, Li Z, Cai YH, Feng LJ, Wu Y, Li X, Luo HB. The Molecular Basis for the Selectivity of Tadalafil toward Phosphodiesterase 5 and 6: A Modeling Study. J Chem Inf Model 2013; 53:3044-53. [DOI: 10.1021/ci400458z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Yi-You Huang
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhe Li
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Ying-Hong Cai
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Ling-Jun Feng
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yinuo Wu
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xingshu Li
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
21
|
Shi F, Perez E, Wang T, Peitz B, Lapolt PS. Stage- and Cell-Specific Expression of Soluble Guanylyl Cyclase Alpha and Beta Subunits, cGMP-Dependent Protein Kinase I Alpha and Beta, and Cyclic Nucleotide-Gated Channel Subunit 1 in the Rat Testis. ACTA ACUST UNITED AC 2013; 26:258-63. [PMID: 15713832 DOI: 10.1002/j.1939-4640.2005.tb01093.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several studies suggest that nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) modulate testicular function. In this study, we examined the expression of cGMP-dependent protein kinase G-I (PKG-I), and cyclic nucleotide-gated channel 1 (CNG-1), 2 known mediators of cGMP action, and the expression of soluble guanylyl cyclase (sGC) subunits in the rat testis. Immunohistochemical analysis revealed that the alpha subunit of sGC was expressed in the blood vessels and Leydig cells of adult rat testes. In addition, the sGC alpha subunit was observed in the acrosomal structures of spermatids undergoing the middle and later stages of spermiogenesis, but not in mature spermatozoa. Similar localization and expression patterns were seen for the sGC beta subunit, indicating coexpression of the sGC subunits. PKG-I was expressed in blood vessels and in the acrosomal region of spermatids during the early and middle stages of spermiogenesis but was not observed in Leydig cells or in mature spermatozoa. In contrast to sGC and PKG-I, CNG-1 was expressed only in cytoplasm and the residual bodies of late-stage (17-19) spermatids, with no staining observed in blood vessels and Leydig cells. These results demonstrate that sGC, PKG-I, and CNG-1 are expressed in a stage- and cell-specific manner in the rat testis. The distinct temporal patterns of expression of these components of cGMP signaling pathways suggest different physiological roles for sGC, PKG-I, and CNG-1 in spermiogenesis and steroidogenesis.
Collapse
Affiliation(s)
- Fangxiong Shi
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, 210095 China.
| | | | | | | | | |
Collapse
|
22
|
Çifci G, Aviyente V, Akten ED. Molecular Docking Study Based on Pharmacophore Modeling for Novel PhosphodiesteraseIV Inhibitors. Mol Inform 2012; 31:459-71. [DOI: 10.1002/minf.201100141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 05/15/2012] [Indexed: 11/06/2022]
|
23
|
Discovery of new inhibitor for PDE3 by virtual screening. Bioorg Med Chem Lett 2011; 21:1617-20. [PMID: 21330134 DOI: 10.1016/j.bmcl.2011.01.120] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 12/21/2010] [Accepted: 01/26/2011] [Indexed: 11/23/2022]
Abstract
In this work, we tried to find a new scaffold for a PDE3 using virtual screening for the obesity treatment. We first analyzed structural features for the known PDE3 inhibitors based on the PDE3B-ligand complex structure, and then carried out a docking study based on PDE3B 3D structure. We obtained a compound as potent PDE3 inhibitor stimulating lipolysis in murine adipocytes and human adipocytes.
Collapse
|
24
|
Francis SH, Busch JL, Corbin JD, Sibley D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol Rev 2010; 62:525-63. [PMID: 20716671 PMCID: PMC2964902 DOI: 10.1124/pr.110.002907] [Citation(s) in RCA: 733] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To date, studies suggest that biological signaling by nitric oxide (NO) is primarily mediated by cGMP, which is synthesized by NO-activated guanylyl cyclases and broken down by cyclic nucleotide phosphodiesterases (PDEs). Effects of cGMP occur through three main groups of cellular targets: cGMP-dependent protein kinases (PKGs), cGMP-gated cation channels, and PDEs. cGMP binding activates PKG, which phosphorylates serines and threonines on many cellular proteins, frequently resulting in changes in activity or function, subcellular localization, or regulatory features. The proteins that are so modified by PKG commonly regulate calcium homeostasis, calcium sensitivity of cellular proteins, platelet activation and adhesion, smooth muscle contraction, cardiac function, gene expression, feedback of the NO-signaling pathway, and other processes. Current therapies that have successfully targeted the NO-signaling pathway include nitrovasodilators (nitroglycerin), PDE5 inhibitors [sildenafil (Viagra and Revatio), vardenafil (Levitra), and tadalafil (Cialis and Adcirca)] for treatment of a number of vascular diseases including angina pectoris, erectile dysfunction, and pulmonary hypertension; the PDE3 inhibitors [cilostazol (Pletal) and milrinone (Primacor)] are used for treatment of intermittent claudication and acute heart failure, respectively. Potential for use of these medications in the treatment of other maladies continues to emerge.
Collapse
Affiliation(s)
- Sharron H Francis
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232-0615, USA.
| | | | | | | |
Collapse
|
25
|
Jensen JT, Stouffer RL, Stanley JE, Zelinski MB. Evaluation of the phosphodiesterase 3 inhibitor ORG 9935 as a contraceptive in female macaques: initial trials. Contraception 2009; 81:165-71. [PMID: 20103457 DOI: 10.1016/j.contraception.2009.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/28/2009] [Accepted: 09/30/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND The study was conducted to determine whether a phosphodiesterase (PDE) 3 inhibitor has potential as a novel contraceptive in primates. METHODS Regularly cycling adult female cynomolgus macaques of proven fertility (n=16) were treated for 7 months with placebo (controls) or the PDE3 inhibitor ORG 9935 as a daily food treat (150 mg/kg) or as a weekly depot injection (150 mg/kg, sc). After 1 month, a male of proven fertility was introduced into each group. Females underwent weekly monitoring of progesterone (P) and ultrasound evaluation for pregnancy if P remained elevated (1.0 ng/mL) >3 weeks. ORG 9935 values were evaluated using high-performance liquid chromatography. RESULTS Overall, the pregnancy rate in ORG 9935-treated monkeys (4/8, 50%) did not differ from controls (7/8, 88%; p=.5). However, no animal became pregnant in a cycle when the serum level of ORG 9935 exceeded 300 nmol/L. Moreover, two treated monkeys who mated throughout the treatment phase and did not conceive became pregnant within four cycles after stopping ORG 9935. The other two animals were discontinued prematurely from the protocol. CONCLUSIONS These results demonstrate that ORG 9935 may prevent pregnancy in primates at serum concentrations above 300 nmol/L and that the effect is reversible.
Collapse
Affiliation(s)
- Jeffrey T Jensen
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239, USA.
| | | | | | | |
Collapse
|
26
|
Mohamed RH. WITHDRAWN: Effect of cilostazol in alleviating cardiovascular complications through regulation of type 1 plasminogen activator inhibitor and transforming growth factor-β 1 overexpression in experimental rats. Biomed Pharmacother 2009:S0753-3322(09)00041-9. [PMID: 19303735 DOI: 10.1016/j.biopha.2009.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 02/24/2009] [Indexed: 10/21/2022] Open
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Rasha H Mohamed
- Biochemistry Department, Faculty of Pharmacy, Zagazig University 44511, Egypt
| |
Collapse
|
27
|
McEwan DG, Brunton VG, Baillie GS, Leslie NR, Houslay MD, Frame MC. Chemoresistant KM12C colon cancer cells are addicted to low cyclic AMP levels in a phosphodiesterase 4-regulated compartment via effects on phosphoinositide 3-kinase. Cancer Res 2007; 67:5248-57. [PMID: 17545604 DOI: 10.1158/0008-5472.can-07-0097] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One of the major problems in treating colon cancer is chemoresistance to cytotoxic chemotherapeutic agents. There is therefore a need to devise new strategies to inhibit colon cancer cell growth and survival. Here, we show that a combination of low doses of the adenylyl cyclase activator forskolin together with the specific cyclic AMP (cAMP) phosphodiesterase-4 (PDE4) inhibitor rolipram, but not the cAMP phosphodiesterase-3 (PDE3) inhibitor cilostamide, causes profound growth arrest of chemoresistant KM12C colon cancer cells. Low-dose forskolin causes KM12C cells to exit the cell cycle in G1 by inducing p27(Kip1) and primes cells for apoptosis on addition of rolipram. The effect of the low-dose forskolin/rolipram combination is mediated by displacement of the phosphatidylinositol 3,4,5-trisphosphate/phosphoinositide 3-kinase signaling module from the plasma membrane and suppression of the Akt/protein kinase-B oncogene pathway, to which KM12C cells are addicted for growth. The cAMP and phosphoinositide 3-kinase pathways form a critical intersection in this response, and reexpression of the tumor suppressor lipid phosphatase, phosphatase and tensin homologue, which is commonly lost or mutated in colon cancer, sensitizes KM12C cells to growth inhibition by challenge with low-dose forskolin. Certain chemoresistant colon cancer cells are therefore exquisitely sensitive to subtle elevation of cAMP by a synergistic low-dose adenylyl cyclase activator/PDE4 inhibitor combination. Indeed, these cells are addicted to maintenance of low cAMP concentrations in a compartment that is regulated by PDE4. Well-tolerated doses of PDE4 inhibitors that are already in clinical development for other therapeutic indications may provide an exciting new strategy for the treatment of colon cancer.
Collapse
Affiliation(s)
- David G McEwan
- The Beatson Institute for Cancer Research, Cancer Research UK Beatson Laboratories, Glasgow, United Kingdom
| | | | | | | | | | | |
Collapse
|
28
|
Willoughby D, Cooper DMF. Organization and Ca2+Regulation of Adenylyl Cyclases in cAMP Microdomains. Physiol Rev 2007; 87:965-1010. [PMID: 17615394 DOI: 10.1152/physrev.00049.2006] [Citation(s) in RCA: 327] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The adenylyl cyclases are variously regulated by G protein subunits, a number of serine/threonine and tyrosine protein kinases, and Ca2+. In some physiological situations, this regulation can be readily incorporated into a hormonal cascade, controlling processes such as cardiac contractility or neurotransmitter release. However, the significance of some modes of regulation is obscure and is likely only to be apparent in explicit cellular contexts (or stages of the cell cycle). The regulation of many of the ACs by the ubiquitous second messenger Ca2+provides an overarching mechanism for integrating the activities of these two major signaling systems. Elaborate devices have been evolved to ensure that this interaction occurs, to guarantee the fidelity of the interaction, and to insulate the microenvironment in which it occurs. Subcellular targeting, as well as a variety of scaffolding devices, is used to promote interaction of the ACs with specific signaling proteins and regulatory factors to generate privileged domains for cAMP signaling. A direct consequence of this organization is that cAMP will exhibit distinct kinetics in discrete cellular domains. A variety of means are now available to study cAMP in these domains and to dissect their components in real time in live cells. These topics are explored within the present review.
Collapse
Affiliation(s)
- Debbie Willoughby
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
29
|
Osadchii OE. Myocardial phosphodiesterases and regulation of cardiac contractility in health and cardiac disease. Cardiovasc Drugs Ther 2007; 21:171-94. [PMID: 17373584 DOI: 10.1007/s10557-007-6014-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Accepted: 02/21/2007] [Indexed: 01/14/2023]
Abstract
Phosphodiesterase (PDE) inhibitors are potent cardiotonic agents used for parenteral inotropic support in heart failure. Contractile effects of these agents are mediated through cAMP-protein kinase A-induced stimulation of I (Ca2+) which ultimately results in increased Ca(2+)-induced sarcoplasmic reticulum Ca(2+) release. A number of additional effects such as increases in sarcoplasmic reticulum Ca(2+) stores, stimulation of reverse mode Na(+)-Ca(2+) exchange, direct or cAMP-mediated effects on sarcoplasmic reticulum ryanodine receptor, stimulation of the voltage-sensitive sarcoplasmic reticulum Ca(2+) release mechanism, as well as A(1) adenosine receptor blockade could contribute to positive inotropic responses to PDE inhibitors. Moreover, some PDE inhibitors exhibit Ca(2+) sensitizer properties as they could increase the affinity of troponin C Ca(2+)-binding sites as well as reduce Ca(2+) threshold for thin myofilament sliding and facilitate cross-bridge cycling. Inotropic responses to PDE inhibitors are significantly reduced in cardiac disease, an effect largely attributed to downregulation of cAMP-mediated signalling due to sustained sympathetic activation. Four PDE isoenzymes (PDE1, PDE2, PDE3 and PDE4) are present in myocardial tissue of various mammalian species, of which PDE3 and PDE4 are particularly involved in regulation of cardiac myocyte contraction. PDE cAMP-hydrolysing activity is preserved in compensated cardiac hypertrophy but significantly reduced in animal models of heart failure. However, clinical studies have not revealed any changes in distribution profile as well as kinetic and regulatory properties of myocardial PDEs in failing human hearts. A reduction of PDE inhibitors-induced contractile responses in heart failure has therefore been ascribed to reduced cAMP synthesis due to uncoupling of adenylyl cyclase from beta-adrenoreceptor. In cardiac myocytes, PDEs are targeted to distinct subcellular compartments by scaffolding proteins such as myomegalin, mAKAP and beta-arrestins. Over subcellular microdomains, cAMP hydrolysis by PDE3 and PDE4 allows to control the activity of local pools of protein kinase A and therefore the extent of protein kinase A-mediated phosphorylation of cellular proteins.
Collapse
Affiliation(s)
- Oleg E Osadchii
- Cardiology Group, School of Clinical Sciences, University Clinical Departments, University of Liverpool, The Duncan Building, Liverpool, UK.
| |
Collapse
|
30
|
Abstract
Phosphodiesterases are a diverse family of enzymes that hydrolyse cyclic nucleotides and thus play a key role in regulating intracellular levels of the second messengers cAMP and cGMP, and hence cell function. Theophylline and papaverine have historically been used therapeutically and are known to be weak inhibitors of PDE, but to what extent this contributed toward their clinical efficacy was poorly defined. However, the discovery of 11 isoenzyme families and our increased understanding of their function at the cell and molecular level provides an impetus for the development of isoenzyme selective inhibitors for the treatment of various diseases. This review focuses on the development of PDE3 inhibitors for congestive heart failure, PDE4 inhibitors for inflammatory airways disease and most successfully, PDE5 inhibitors for erectile dysfunction.
Collapse
Affiliation(s)
- Victoria Boswell-Smith
- Sackler Institute of Pulmonary Pharmacology, Kings College London School of Biomedical Health and Life Sciences, 5th Floor, Hodgkin Building, Guys Campus, Kings College, London SE1 1UL, London
| | - Domenico Spina
- Sackler Institute of Pulmonary Pharmacology, Kings College London School of Biomedical Health and Life Sciences, 5th Floor, Hodgkin Building, Guys Campus, Kings College, London SE1 1UL, London
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Kings College London School of Biomedical Health and Life Sciences, 5th Floor, Hodgkin Building, Guys Campus, Kings College, London SE1 1UL, London
- Sackler Institute of Pulmonary Pharmacology, Kings College London School of Biomedical Health and Life Sciences, 5th Floor, Hodgkin Building, Guys Campus, Kings College, London SE1 1UL, London. E-mail:
| |
Collapse
|
31
|
Patel LS, Mitchell CK, Dubinsky WP, O’Brien J. Regulation of gap junction coupling through the neuronal connexin Cx35 by nitric oxide and cGMP. ACTA ACUST UNITED AC 2006; 13:41-54. [PMID: 16613779 PMCID: PMC2189984 DOI: 10.1080/15419060600631474] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Gap-junctional coupling among neurons is subject to regulation by a number of neurotransmitters including nitric oxide. We studied the mechanisms by which NO regulates coupling in cells expressing Cx35, a connexin expressed in neurons throughout the central nervous system. NO donors caused potent uncoupling of HeLa cells stably transfected with Cx35. This effect was mimicked by Bay 21-4272, an activator of guanylyl cyclase. A pharmacological analysis indicated that NO-induced uncoupling involved both PKG-dependent and PKG-independent pathways. PKA was involved in both pathways, suggesting that PKG-dependent uncoupling may be indirect. In vitro, PKG phosphorylated Cx35 at three sites: Ser110, Ser276, and Ser289. A mutational analysis indicated that phosphorylation on Ser110 and Ser276, sites previously shown also to be phosphorylated by PKA, had a significant influence on regulation. Ser289 phosphorylation had very limited effects. We conclude that NO can regulate coupling through Cx35 and that regulation is indirect in HeLa cells.
Collapse
Affiliation(s)
- Leena S. Patel
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - Cheryl K. Mitchell
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - William P. Dubinsky
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
| | - John O’Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
- *Corresponding author: John O’Brien, Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 7.024, Houston, Texas 77030, Phone: (713) 500-5983, FAX: (713) 500-0682, e-mail:
| |
Collapse
|
32
|
Wang H, Liu Y, Huai Q, Cai J, Zoraghi R, Francis SH, Corbin JD, Robinson H, Xin Z, Lin G, Ke H. Multiple Conformations of Phosphodiesterase-5. J Biol Chem 2006; 281:21469-21479. [PMID: 16735511 DOI: 10.1074/jbc.m512527200] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphodiesterase-5 (PDE5) is the target for sildenafil, vardenafil, and tadalafil, which are drugs for treatment of erectile dysfunction and pulmonary hypertension. We report here the crystal structures of a fully active catalytic domain of unliganded PDE5A1 and its complexes with sildenafil or icarisid II. These structures together with the PDE5A1-isobutyl-1-methylxanthine complex show that the H-loop (residues 660-683) at the active site of PDE5A1 has four different conformations and migrates 7-35A upon inhibitor binding. In addition, the conformation of sildenafil reported herein differs significantly from those in the previous structures of chimerically hybridized or almost inactive PDE5. Mutagenesis and kinetic analyses confirm that the H-loop is particularly important for substrate recognition and that invariant Gly(659), which immediately precedes the H-loop, is critical for optimal substrate affinity and catalytic activity.
Collapse
Affiliation(s)
- Huanchen Wang
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260
| | - Yudong Liu
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260
| | - Qing Huai
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260
| | - Jiwen Cai
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510080, China
| | - Roya Zoraghi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0615
| | - Sharron H Francis
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0615
| | - Jackie D Corbin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0615
| | - Howard Robinson
- Biology Department, Brookhaven National Laboratory, Upton, New York 11973-5000
| | - Zhongcheng Xin
- Andrology Center, Peking University First Hospital, Peking University, 8 Xishiku Street, Beijing (100034), China
| | - Guiting Lin
- Department of Urology, University of California, San Francisco, California 94143-1695
| | - Hengming Ke
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260.
| |
Collapse
|
33
|
Lynch MJ, Hill EV, Houslay MD. Intracellular targeting of phosphodiesterase-4 underpins compartmentalized cAMP signaling. Curr Top Dev Biol 2006; 75:225-59. [PMID: 16984814 DOI: 10.1016/s0070-2153(06)75007-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The phosphodiesterase-4 (PDE4) enzyme belongs to a family of cAMP-dependent phosphodiesterases that provide the major means of hydrolyzing and, thereby, inactivating the key intracellular second messenger, cAMP. As such, PDE4s are central to the regulation of many diverse signaling processes that allow cells to respond to external stimuli. Four genes (4A, 4B, 4C, and 4D) encode around 20 distinct isoform members of the PDE4 family. Each isoform is characterized by a unique N-terminal region. PDE4s are multidomain metallohydrolases with each domain serving particular roles allowing them to be targeted to varying regions and organelles of intracellular space and regulated in distinct fashions by phosphorylation and protein-protein interaction. Although identical in catalytic function, each isoform locates to distinct regions within the cell so as to create and manage spatially distinct pools of cAMP. The multiplicity of partners associating with members of the four gene PDE4 family places these enzymes in key regulatory positions, permitting them to channel complex biological signals via fundamental signaling cohorts such as G-protein-coupled receptors (GPCRs), arrestins, A-kinase-anchoring proteins (AKAPs), and tyrosyl family kinases. The cAMP cascade has long been linked to cellular growth and embryogenesis and with this comes the implication that PDE4 may play considerable roles in the regulation of progeny development in maturing cells and tissues.
Collapse
Affiliation(s)
- Martin J Lynch
- Division of Biochemistry and Molecular Biology, IBLS, Wolfson Building University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | | | | |
Collapse
|
34
|
Shimizu K, Murata T, Hiramoto K, Sugiyama T, Nakagawa T, Manganiello VC, Tagawa T. Expression of cyclic nucleotide phosphodiesterase 3A in isolated rat submandibular acini. Arch Oral Biol 2005; 51:83-8. [PMID: 16102722 DOI: 10.1016/j.archoralbio.2005.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 06/20/2005] [Accepted: 06/22/2005] [Indexed: 11/24/2022]
Abstract
Phosphodiesterase (PDE) 3 has been characterized in isolated rat submandibular acini. PDE3 activity was detected in homogenates of isolated rat submandibular acini; little or no PDE3 activity was found in ducts. About 62% of PDE3 activity in the acini was recovered in the supernatant fractions; 38% in particulate fractions. In the acini, but not ducts, PDE3A mRNA was detected by reverse transcriptase-polymerase chain reaction (RT-PCR). The PDE3-specific inhibitor, cilostamide, increased the ratio of apomucin mRNA/18s rRNA, as quantified by real-time RT-PCR. Our results indicate that PDE3A may be important in regulating cAMP pools that control acini functions.
Collapse
Affiliation(s)
- Kasumi Shimizu
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie 514-8507, Japan.
| | | | | | | | | | | | | |
Collapse
|
35
|
Manallack DT, Hughes RA, Thompson PE. The next generation of phosphodiesterase inhibitors: structural clues to ligand and substrate selectivity of phosphodiesterases. J Med Chem 2005; 48:3449-62. [PMID: 15887951 DOI: 10.1021/jm040217u] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- David T Manallack
- Victorian College of Pharmacy, Monash University (Parkville Campus), 381 Royal Parade, Parkville Victoria, 3052, Australia.
| | | | | |
Collapse
|
36
|
Varnerin JP, Chung CC, Patel SB, Scapin G, Parmee ER, Morin NR, MacNeil DJ, Cully DF, Van der Ploeg LHT, Tota MR. Expression, refolding, and purification of recombinant human phosphodiesterase 3B: definition of the N-terminus of the catalytic core. Protein Expr Purif 2005; 35:225-36. [PMID: 15135397 DOI: 10.1016/j.pep.2004.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2003] [Revised: 01/12/2004] [Indexed: 10/26/2022]
Abstract
We have developed an expression, refolding, and purification protocol for the catalytic domain of human Phosphodiesterase 3B (PDE3B). High level expression in Escherichia coli has been achieved with yields of up to 20mg/L. The catalytic domain of the enzyme was purified by affinity chromatography utilizing a novel affinity ligand. PDE3B, purified by affinity chromatography, with no single impurity #10878;1% as determined by SDS-PAGE, has a specific activity of 2210+/-442nmol/min/mg and a KM for cAMP of 44+/-4.5nM. Reducing the size of the expressed catalytic domain from residues 387-1112 to residues 654-1086 greatly reduced the aggregation phenomena observed with the affinity purified PDE3B. The definition of the N-terminus of the catalytic core was examined through the generation of several truncation mutants spanning amino acid residues 636-674. Constructs starting at E665 and M674 were fully active and devoid of activity, respectively. A construct starting at D668 had a Vmax reduced by approximately 10-fold relative to the longer constructs, yet the KM was not affected. This indicates the minimal N-terminus of the catalytic core lies between E665 and Y667. Refolding and affinity purification of the 654-1073 catalytic core of PDE3B has been employed to produce large quantities of highly pure enzyme for structural studies.
Collapse
|
37
|
Fleming YM, Frame MC, Houslay MD. PDE4-regulated cAMP degradation controls the assembly of integrin-dependent actin adhesion structures and REF52 cell migration. J Cell Sci 2005; 117:2377-88. [PMID: 15126637 DOI: 10.1242/jcs.01096] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Plating of REF52 cells onto extracellular matrix components leads to the formation of integrin-dependent actin adhesion microspikes. We show that the formation of these structures is sensitive to chemical agents that regulate cAMP levels, such as forskolin and IBMX. In particular, by using the specific inhibitor rolipram, we identify the PDE4 family of cAMP-specific phosphodiesterases as critical regulators of this process. The effect of PDE4 on microspike formation is mediated by actions exerted through the activation of PKA - rather than through the alternative cAMP effector, Epac. We provide evidence that peripheral microspikes are RhoA-, ROCK- and myosin-dependent, and that this pathway is suppressed by PDE4 inhibition. In addition, PDE4 inhibition impairs cell locomotion that requires dynamic protrusion and retraction of peripheral spike structures. Our data demonstrate that PDE4 activity is a key modulator of integrin-induced actin assembly at the cell periphery which, in turn, controls cell migration.
Collapse
Affiliation(s)
- Yvonne M Fleming
- Institute of Biological and Life Sciences, Davidson and Wolfson Buildings, University of Glasgow, Glasgow G12 8QQ, UK.
| | | | | |
Collapse
|
38
|
Jensen JT, Zelinski-Wooten MB, Schwinof KM, Vance JE, Stouffer RL. The phosphodiesterase 3 inhibitor ORG 9935 inhibits oocyte maturation during gonadotropin-stimulated ovarian cycles in rhesus macaques. Contraception 2005; 71:68-73. [PMID: 15639077 DOI: 10.1016/j.contraception.2004.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Accepted: 07/02/2004] [Indexed: 10/26/2022]
Abstract
To determine whether phosphodiesterase (PDE) 3 inhibitors prevent the resumption of meiosis by primate oocytes in vivo, rhesus macaques were stimulated to develop multiple preovulatory follicles by administering human recombinant gonadotropins, and follicles were aspirated 34 h after an ovulatory stimulus (human chorionic gonadotropin [hCG]). Monkeys received no further treatment (controls) or the PDE3 inhibitor ORG 9935 (a) exclusively in the periovulatory interval beginning 6-12 h prior to receiving hCG at 200 mg/kg every 12 h orally (PER200) or a 200 mg/kg oral loading dose followed by 50 mg/kg sc every 6 h (PER50) or (b) throughout the ovarian stimulation protocol with daily increases until a dose of 200 mg/kg bid was administered onward from the eighth day of ovarian stimulation (EXT200). The primary outcome was the number of oocytes that had resumed meiosis (germinal vesicle breakdown [GVBD]) at collection. At initial aspiration, 85% of oocytes recovered from control animals (n = 4) had progressed to GVBD compared with 53% (p<.01), 23% (p<.01), and 13% (p<.01) recovered from animals in the PER200 (n = 2), PER50 (n = 1) and EXT200 (n = 3) groups, respectively. Although spontaneous maturation of oocytes was observed during follow-up culture in the absence of ORG 9935, none of the oocytes in the PER50 or EXT200 underwent normal fertilization in vitro. These results demonstrate that the PDE3 inhibitor ORG 9935 blocks oocyte maturation during gonadotropin-stimulated ovarian cycles in rhesus macaques and suggest that PDE3 inhibitors have potential clinical use as contraceptives in women.
Collapse
Affiliation(s)
- Jeffrey T Jensen
- Department of Obstetrics and Gynecology and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR 97239, USA.
| | | | | | | | | |
Collapse
|
39
|
Galantino-Homer HL, Florman HM, Storey BT, Dobrinski I, Kopf GS. Bovine sperm capacitation: assessment of phosphodiesterase activity and intracellular alkalinization on capacitation-associated protein tyrosine phosphorylation. Mol Reprod Dev 2004; 67:487-500. [PMID: 14991741 DOI: 10.1002/mrd.20034] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mammalian sperm capacitation is the obligatory maturational process leading to the development of the fertilization-competent state. Heparin is known to be a unique species-specific inducer of bovine sperm capacitation in vitro and glucose a unique inhibitor of this induction. Heparin-induced capacitation of bovine sperm has been shown to correlate with protein kinase A (PKA)-dependent protein tyrosine phosphorylation driven by an increase in intracellular cAMP. This study examines the possible roles of cyclic nucleotide phosphodiesterase (PDE) activity and intracellular alkalinization on bovine sperm capacitation and the protein tyrosine phosphorylation associated with it. Measurement of whole cell PDE kinetics during capacitation reveals neither a substantial change with heparin nor one with glucose: PDE activity is effectively constitutive in maintaining intracellular cAMP levels during capacitation. In contrast to a transient increase in intracellular pH, a sustained increase in medium pH by switching from 5% CO(2)/95% air incubation to 1% CO(2)/99% air incubation over 4 hr in the absence of heparin resulted in an increase in protein tyrosine phosphorylation and in the extent of induced acrosome reaction comparable to that observed following heparin-induced capacitation in 5% CO(2). These results suggest that increased bicarbonate-dependent adenylyl cyclase activity, driven by alkalinization, increases intracellular cAMP and so increases PKA activity mediating protein tyrosine phosphorylation. Quantitative analysis of the lactic acid production rate by bovine sperm glycolysis accounts fully for intracellular acidification sufficient to offset heparin-induced alkalinization, thus inhibiting capacitation. The mechanism by which heparin uniquely induces intracellular alkalinization in bovine sperm leading to capacitation remains obscure, inviting future investigation.
Collapse
Affiliation(s)
- Hannah L Galantino-Homer
- Center for Research on Reproduction and Women's Health, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania, USA.
| | | | | | | | | |
Collapse
|
40
|
Huai Q, Wang H, Zhang W, Colman RW, Robinson H, Ke H. Crystal structure of phosphodiesterase 9 shows orientation variation of inhibitor 3-isobutyl-1-methylxanthine binding. Proc Natl Acad Sci U S A 2004; 101:9624-9. [PMID: 15210993 PMCID: PMC470725 DOI: 10.1073/pnas.0401120101] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2004] [Accepted: 05/12/2004] [Indexed: 11/18/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are enzymes controlling cellular concentrations of the second messengers cAMP and cGMP. The crystal structure of the catalytic domain of PDE9A2, a member of a PDE family specifically hydrolyzing cGMP, has been determined at 2.23-A resolution. The PDE9A2 catalytic domain closely resembles the cAMP-specific PDE4D2 but is significantly different from the cGMP-specific PDE5A1, implying that each individual PDE family has its own characteristic substrate recognition mechanism. The different conformations of the H and M loops between PDE9A2 and PDE5A1 imply their less critical roles in nucleotide recognition. The nonselective inhibitor 3-isobutyl-1-methylxanthine (IBMX) binds to a similar subpocket in the active sites of PDE4, PDE5, and PDE9 and has a common pattern of the binding. However, significantly different orientations and interactions of IBMXs are observed among the three PDE families and also between two monomers of the PDE9A2 dimer. The kinetic properties of the PDE9A2 catalytic domain similar to those of full-length PDE9A imply that the N-terminal regulatory domain does not significantly alter the catalytic activity and the IBMX inhibition.
Collapse
Affiliation(s)
- Qing Huai
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | | | | | | | | | | |
Collapse
|
41
|
Huai Q, Liu Y, Francis SH, Corbin JD, Ke H. Crystal structures of phosphodiesterases 4 and 5 in complex with inhibitor 3-isobutyl-1-methylxanthine suggest a conformation determinant of inhibitor selectivity. J Biol Chem 2004; 279:13095-101. [PMID: 14668322 DOI: 10.1074/jbc.m311556200] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are a superfamily of enzymes controlling cellular concentrations of the second messengers cAMP and cGMP. Crystal structures of the catalytic domains of cGMP-specific PDE5A1 and cAMP-specific PDE4D2 in complex with the nonselective inhibitor 3-isobutyl-1-methylxanthine have been determined at medium resolution. The catalytic domain of PDE5A1 has the same topological folding as that of PDE4D2, but three regions show different tertiary structures, including residues 79-113, 208-224 (H-loop), and 341-364 (M-loop) in PDE4D2 or 535-566, 661-676, and 787-812 in PDE5A1, respectively. Because H- and M-loops are involved in binding of the selective inhibitors, the different conformations of the loops, thus the distinct shapes of the active sites, will be a determinant of inhibitor selectivity in PDEs. IBMX binds to a subpocket that comprises key residues Ile-336, Phe-340, Gln-369, and Phe-372 of PDE4D2 or Val-782, Phe-786, Gln-817, and Phe-820 of PDE5A1. This subpocket may be a common site for binding nonselective inhibitors of PDEs.
Collapse
Affiliation(s)
- Qing Huai
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, the University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | | | | | | | | |
Collapse
|
42
|
Edmondson SD, Mastracchio A, He J, Chung CC, Forrest MJ, Hofsess S, MacIntyre E, Metzger J, O'Connor N, Patel K, Tong X, Tota MR, Van der Ploeg LHT, Varnerin JP, Fisher MH, Wyvratt MJ, Weber AE, Parmee ER. Benzyl vinylogous amide substituted aryldihydropyridazinones and aryldimethylpyrazolones as potent and selective PDE3B inhibitors. Bioorg Med Chem Lett 2003; 13:3983-7. [PMID: 14592490 DOI: 10.1016/j.bmcl.2003.08.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Aryldihydropyridazinones and aryldimethylpyrazolones with 2-benzyl vinylogous amide substituents have been identified as potent PDE3B subtype selective inhibitors. Dihydropyridazinone 8a (PDE3B IC(50)=0.19 nM, 3A IC(50)=1.3 nM) was selected for in vivo evaluation of lipolysis induction, metabolic rate increase, and cardiovascular effects.
Collapse
Affiliation(s)
- Scott D Edmondson
- Department of Medicinal Chemistry, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Huai Q, Wang H, Sun Y, Kim HY, Liu Y, Ke H. Three-dimensional structures of PDE4D in complex with roliprams and implication on inhibitor selectivity. Structure 2003; 11:865-73. [PMID: 12842049 DOI: 10.1016/s0969-2126(03)00123-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Selective inhibitors against the 11 families of cyclic nucleotide phosphodiesterases (PDEs) are used to treat various human diseases. How the inhibitors selectively bind the conserved PDE catalytic domains is unknown. The crystal structures of the PDE4D2 catalytic domain in complex with (R)- or (R,S)-rolipram suggest that inhibitor selectivity is determined by the chemical nature of amino acids and subtle conformational changes of the binding pockets. The conformational states of Gln369 in PDE4D2 may play a key role in inhibitor recognition. The corresponding Y329S mutation in PDE7 may lead to loss of the hydrogen bonds between rolipram and Gln369 and is thus a possible reason explaining PDE7's insensitivity to rolipram inhibition. Docking of the PDE5 inhibitor sildenafil into the PDE4 catalytic pocket further helps understand inhibitor selectivity.
Collapse
Affiliation(s)
- Qing Huai
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
44
|
Haning H, Niewöhner U, Bischoff E. Phosphodiesterase type 5 (PDE5) inhibitors. PROGRESS IN MEDICINAL CHEMISTRY 2003; 41:249-306. [PMID: 12774696 DOI: 10.1016/s0079-6468(02)41007-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Helmut Haning
- BAYER AG Pharmaceutical Business Group, Medicinal Chemistry, D-42096 Wuppertal, Germany
| | | | | |
Collapse
|
45
|
Spoto G, Fioroni M, Rubini C, Contento A, Di Nicola M, Forcella S, Piattelli A. Cyclic guanosine monophosphate phosphodiesterase activity in human gingival carcinoma. J Oral Pathol Med 2003; 32:189-94. [PMID: 12653856 DOI: 10.1034/j.1600-0714.2003.00083.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cyclic guanosine monophosphate (cGMP) is an essential second messenger metabolized by phosphodiesterases (PDEs). OBJECTIVES We looked for a possible correlation of PDE activities in human oral squamous cell carcinoma (OSCC) with and without lymph node metastases. MATERIALS AND METHODS The analysis of phosphodiesterase activity and the cGMP assay were done by reverse-phase HPLC on samples of fresh or frozen gingival tissues. Analysis of cGMP was confirmed with the enzyme-linked immunoabsorption assay. RESULTS AND CONCLUSIONS cGMP PDE activity was 34.92 +/- 7.17 SD, 12.89 +/- 4.43 SD, and 35.88 +/- 8.76 SD (nmols/mg of protein), respectively, in controls, samples without lymph node involvement (N-), and specimens with lymph node metastases (N+). cGMP values were 1.97 +/- 0.63 SD, 3.30 +/- 1.47 SD, and 3.49 +/- 1.47 SD (nmols/mg of protein). Our data support the hypothesis of a role for cGMP and PDE in the progression of OSCC.
Collapse
Affiliation(s)
- Giuseppe Spoto
- Department of Applied Sciences of Oral and Dental Diseases, G. D'Annunzio University, Via dei Vestini 31, 66013 Chieti, Italy.
| | | | | | | | | | | | | |
Collapse
|
46
|
Salomone S, Caruso A, Cutuli VM, Mangano NG, Prato A, Amico-Roxas M, Bianchi A, Clementi G. Effects of adrenomedullin on the contraction of gastric arteries during reserpine-induced gastric ulcer. Peptides 2003; 24:117-22. [PMID: 12576092 DOI: 10.1016/s0196-9781(02)00283-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Adrenomedullin (100 ng/kg, s.c.) prevents reserpine-induced damage of gastric mucosa. In the model of in vitro gastric arteries from reserpine-treated rats, adrenomedullin pre-treatment resulted in a decrease of the vasoconstriction in response to 5-hydroxytryptamine. In contrast, adrenomedullin pre-treatment of rat with intact gastric mucosa did not affect the vasoconstriction to 5-hydroxytryptamine. In the presence of the NOS inhibitor N(G)-nitro-L-arginine, the responsiveness to 5-hydroxytryptamine in gastric arteries from rats treated with reserpine + adrenomedullin was enhanced to the same level of rats treated with reserpine alone. The anti-ulcer effect of adrenomedullin could therefore be related, at least in part, to an increase of blood flow at the gastric mucosa, by a mechanism involving nitric oxide.
Collapse
Affiliation(s)
- Salvatore Salomone
- Dipartimento di Farmacologia Sperimentale e Clinica, Facoltà di Medicina e Chirurgia, Università di Catania, Viale A Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Effect of the phosphodiesterase 4 inhibitor, rolipram, on retinoic acid-increased alkaline phosphatase activity in the mouse fibroblastic C3H10T1/2 cell line. Arch Oral Biol 2003; 48:63-7. [PMID: 12615143 DOI: 10.1016/s0003-9969(02)00166-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have evaluated effects of a phosphodiesterase (PDE) 4 inhibitor on retinoic acid-increased alkaline phosphatase activity in the mouse fibroblastic C3H10T1/2 clone 8 (10T1/2) cell line. 10T1/2 cells were cultured in minimum essential medium (MEM) and 10% fetal bovine serum with or without 1 microM retinoic acid and/or the PDE 4 inhibitor, rolipram, and harvested at specific intervals before measurement of alkaline phosphatase activity, cAMP production in response to parathyroid hormone, osteocalcin synthesis and expression, and phosphodiesterase activity. Retinoic acid-increased alkaline phosphatase activity, and slightly enhanced cAMP production in response to parathyroid hormone. However, it did not affect osteocalcin synthesis and expression. In the presence of retinoic acid, PDE 4 activity was not changed. A PDE 4 inhibitor, rolipram, and cAMP analog, 8-bromo-cAMP dramatically increased retinoic acid's ability to induce alkaline phosphatase activity. This is the first report that PDE 4 may be involved in regulation of retinoic acid-increased alkaline phosphatase activity.
Collapse
|
48
|
Wykes V, Bellamy TC, Garthwaite J. Kinetics of nitric oxide-cyclic GMP signalling in CNS cells and its possible regulation by cyclic GMP. J Neurochem 2002; 83:37-47. [PMID: 12358727 DOI: 10.1046/j.1471-4159.2002.01106.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Physiologically, nitric oxide (NO) signal transduction occurs through soluble guanylyl cyclase (sGC), which catalyses cyclic GMP (cGMP) formation. Knowledge of the kinetics of NO-evoked cGMP signals is therefore critical for understanding how NO signals are decoded. Studies on cerebellar astrocytes showed that sGC undergoes a desensitizing profile of activity, which, in league with phosphodiesterases (PDEs), was hypothesized to diversify cGMP responses in different cells. The hypothesis was tested by examining the kinetics of cGMP in rat striatal cells, in which cGMP accumulated in neurones in response to NO. Based on the effects of selective PDE inhibitors, cGMP hydrolysis following exposure to NO was attributed to a cGMP-stimulated PDE (PDE 2). Analysis of NO-induced cGMP accumulation in the presence of a PDE inhibitor indicated that sGC underwent marked desensitization. However, the desensitization kinetics determined under these conditions described poorly the cGMP profile observed in the absence of the PDE inhibitor. An explanation shown plausible theoretically was that cGMP determines the level of sGC desensitization. In support, tests in cerebellar astrocytes indicated an inverse relationship between cGMP level and recovery of sGC from its desensitized state. We suggest that the degree of sGC desensitization is related to the cGMP concentration and that this effect is not mediated by (de)phosphorylation.
Collapse
Affiliation(s)
- Victoria Wykes
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | | | | |
Collapse
|
49
|
Shimizu K, Murata T, Okumura K, Manganiello VC, Tagawa T. Expression and role of phosphodiesterase 3 in human squamous cell carcinoma KB cells. Anticancer Drugs 2002; 13:875-80. [PMID: 12394274 DOI: 10.1097/00001813-200209000-00014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Phosphodiesterase (PDE) 3s have been characterized in human squamous cell carcinoma KB cells. PDE3 activity was detected in homogenates of KB cells. PDE3A and 3B mRNAs were detected by RT-PCR in RNA from KB cells; the nucleotide sequences of the fragments were identical to those of human PDE3A and 3B. Immunoblotting with anti-PDE3 antibodies detected both PDE3A- and 3B-immunoreactive proteins in KB cells. The PDE3-specific inhibitor, cilostamide, inhibited the proliferation of KB cells. Our results indicate that PDE3s may be important regulators of the growth of KB cells. Therefore, PDE3 inhibitors may be potential new drugs for antiproliferative therapies in squamous cell carcinoma in the head and neck.
Collapse
Affiliation(s)
- Kasumi Shimizu
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Mie University, Mie 514-8507, Japan.
| | | | | | | | | |
Collapse
|
50
|
Bolger GB, McCahill A, Yarwood SJ, Steele MR, Warwicker J, Houslay MD. Delineation of RAID1, the RACK1 interaction domain located within the unique N-terminal region of the cAMP-specific phosphodiesterase, PDE4D5. BMC BIOCHEMISTRY 2002; 3:24. [PMID: 12193273 PMCID: PMC126212 DOI: 10.1186/1471-2091-3-24] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2002] [Accepted: 08/23/2002] [Indexed: 11/10/2022]
Abstract
BACKGROUND The cyclic AMP specific phosphodiesterase, PDE4D5 interacts with the beta-propeller protein RACK1 to form a signaling scaffold complex in cells. Two-hybrid analysis of truncation and mutant constructs of the unique N-terminal region of the cAMP-specific phosphodiesterase, PDE4D5 were used to define a domain conferring interaction with the signaling scaffold protein, RACK1. RESULTS Truncation and mutagenesis approaches showed that the RACK1-interacting domain on PDE4D5 comprised a cluster of residues provided by Asn-22/Pro-23/Trp-24/Asn-26 together with a series of hydrophobic amino acids, namely Leu-29, Val-30, Leu-33, Leu-37 and Leu-38 in a 'Leu-Xaa-Xaa-Xaa-Leu' repeat. This was done by 2-hybrid analyses and then confirmed in biochemical pull down analyses using GST-RACK1 and mutant PDE4D5 forms expressed in COS cells. Mutation of Arg-34, to alanine, in PDE4D5 attenuated its interaction with RACK1 both in 2-hybrid screens and in pull down analyses. A 38-mer peptide, whose sequence reflected residues 12 through 49 of PDE4D5, bound to RACK1 with similar affinity to native PDE4D5 itself (Ka circa 6 nM). CONCLUSIONS The RACK1 Interaction Domain on PDE4D5, that we here call RAID1, is proposed to form an amphipathic helical structure that we suggest may interact with the C-terminal beta-propeller blades of RACK1 in a manner akin to the interaction of the helical G-gamma signal transducing protein with the beta-propeller protein, G-beta.
Collapse
Affiliation(s)
- Graeme B Bolger
- Veterans Affairs Medical Center, Huntsman Cancer Institute, Departments of Medicine (Division of Oncology) and Oncological Science, University of Utah Health Sciences Center, Salt Lake City, UT 84148 USA
- University of Alabama at Birmingham, Comprehensive Cancer Center, WTI 520, 1530 3rd Ave. S., Birmingham AL 35294-3300, USA
| | - Angela McCahill
- Molecular Pharmacology Group, Division of Biochemistry & Molecular Biology, Davidson Building, Institute of Biomedical & Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Stephen J Yarwood
- Molecular Pharmacology Group, Division of Biochemistry & Molecular Biology, Davidson Building, Institute of Biomedical & Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Michael R Steele
- Veterans Affairs Medical Center, Huntsman Cancer Institute, Departments of Medicine (Division of Oncology) and Oncological Science, University of Utah Health Sciences Center, Salt Lake City, UT 84148 USA
| | - Jim Warwicker
- Dept of Biomolecular Sciences, UMIST, Sackville Street, Manchester M60 1QD, UK
| | - Miles D Houslay
- Molecular Pharmacology Group, Division of Biochemistry & Molecular Biology, Davidson Building, Institute of Biomedical & Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
- Dept of Biomolecular Sciences, UMIST, Sackville Street, Manchester M60 1QD, UK
| |
Collapse
|