1
|
Foroshani S, Karp A, Aronow WS, Lanier GM. The role of phosphodiesterase 9A inhibitors in heart failure. Expert Opin Investig Drugs 2024; 33:543-547. [PMID: 38702878 DOI: 10.1080/13543784.2024.2349813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
INTRODUCTION There are currently limited effective treatments available to improve lusitropy in patients suffering from heart failure with preserved ejection fraction. The role of PDE9A in diastolic dysfunction has been well-studied over recent years, with a special focus on its association with myocardial hypertrophy. Recent insights into PDE9A inhibition have brought to light the potential for reversal of cardiac remodeling, with multiple studies showing promising results in preclinical data. AREAS COVERED This expert opinion provides an overview of the role of PDE9A in diastolic heart dysfunction along with the efficacy of PDE9A inhibitors in laboratory models of heart failure with preserved ejection fraction. EXPERT OPINION The available data on PDE9A inhibition in preclinical studies suggest that there is potential for reversal of diastolic dysfunction and myocardial hypertrophy, however, conflicting data suggests that further studies are required before progressing to clinical trials.
Collapse
Affiliation(s)
| | - Avrohom Karp
- Medicine, New York Medical College, Valhalla, NY, USA
| | - Wilbert S Aronow
- Departments of Cardiology and Medicine Westchester Medical Center, New York Medical College, Valhalla, NY, USA
| | - Gregg M Lanier
- Departments of Cardiology and Medicine Westchester Medical Center, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
2
|
Phosphodiesterase-1 in the cardiovascular system. Cell Signal 2022; 92:110251. [DOI: 10.1016/j.cellsig.2022.110251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 11/18/2022]
|
3
|
Zemskov EA, Wu X, Aggarwal S, Yegambaram M, Gross C, Lu Q, Wang H, Tang H, Wang T, Black SM. Nitration of protein kinase G-Iα modulates cyclic nucleotide crosstalk via phosphodiesterase 3A: Implications for acute lung injury. J Biol Chem 2021; 297:100946. [PMID: 34252457 PMCID: PMC8342797 DOI: 10.1016/j.jbc.2021.100946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 12/05/2022] Open
Abstract
Phosphodiesterase 3A (PDE3A) selectively cleaves the phosphodiester bond of cAMP and is inhibited by cGMP, making it an important regulator of cAMP-cGMP signaling crosstalk in the pulmonary vasculature. In addition, the nitric oxide-cGMP axis is known to play an important role in maintaining endothelial barrier function. However, the potential role of protein kinase G-Iα (PKG-Iα) in this protective process is unresolved and was the focus of our study. We describe here a novel mechanism regulating PDE3A activity, which involves a PKG-Iα-dependent inhibitory phosphorylation of PDE3A at serine 654. We also show that this phosphorylation is critical for maintaining intracellular cAMP levels in the pulmonary endothelium and endothelial barrier integrity. In an animal model of acute lung injury (ALI) induced by challenging mice with lipopolysaccharide (LPS), an increase in PDE3 activity and a decrease in cAMP levels in lung tissue was associated with reduced PKG activity upon PKG-Iα nitration at tyrosine 247. The peroxynitrite scavenger manganese (III) tetrakis(1-methyl-4-pyridyl)porphyrin prevented this increase in PDE3 activity in LPS-exposed lungs. In addition, site-directed mutagenesis of PDE3A to replace serine 654 with alanine yielded a mutant protein that was insensitive to PKG-dependent regulation. Taken together, our data demonstrate a novel functional link between nitrosative stress induced by LPS during ALI and the downregulation of barrier-protective intracellular cAMP levels. Our data also provide new evidence that PKG-Iα is critical for endothelial barrier maintenance and that preservation of its catalytic activity may be efficacious in ALI therapy.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Saurabh Aggarwal
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | | | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Hui Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haiyang Tang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA; Department of Internal Medicine, The University of Arizona, Phoenix, Arizona, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA; Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
4
|
Siri-Angkul N, Dadfar B, Jaleel R, Naushad J, Parambathazhath J, Doye AA, Xie LH, Gwathmey JK. Calcium and Heart Failure: How Did We Get Here and Where Are We Going? Int J Mol Sci 2021; 22:ijms22147392. [PMID: 34299010 PMCID: PMC8306046 DOI: 10.3390/ijms22147392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
The occurrence and prevalence of heart failure remain high in the United States as well as globally. One person dies every 30 s from heart disease. Recognizing the importance of heart failure, clinicians and scientists have sought better therapeutic strategies and even cures for end-stage heart failure. This exploration has resulted in many failed clinical trials testing novel classes of pharmaceutical drugs and even gene therapy. As a result, along the way, there have been paradigm shifts toward and away from differing therapeutic approaches. The continued prevalence of death from heart failure, however, clearly demonstrates that the heart is not simply a pump and instead forces us to consider the complexity of simplicity in the pathophysiology of heart failure and reinforces the need to discover new therapeutic approaches.
Collapse
Affiliation(s)
- Natthaphat Siri-Angkul
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Behzad Dadfar
- Department of General Medicine, School of Medicine, Mazandaran University of Medical Sciences, Sari 1471655836, Iran
| | - Riya Jaleel
- School of International Education, Zhengzhou University, Zhengzhou 450001, China
| | - Jazna Naushad
- Weill Cornell Medicine Qatar, Doha P. O. Box 24144, Qatar
| | | | | | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence: ; Tel.: +973-972-2411; Fax: +973-972-7489
| |
Collapse
|
5
|
Novel pyrazole derivatives with oxa/thiadiazolyl, pyrazolyl moieties and pyrazolo[4,3-d]-pyrimidine derivatives as potential antimicrobial and anticancer agents. Bioorg Med Chem Lett 2016; 26:2428-2433. [DOI: 10.1016/j.bmcl.2016.03.117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/21/2022]
|
6
|
Phosphodiesterase inhibitor KMUP-3 displays cardioprotection via protein kinase G and increases cardiac output via G-protein-coupled receptor agonist activity and Ca(2+) sensitization. Kaohsiung J Med Sci 2016; 32:55-67. [PMID: 26944323 DOI: 10.1016/j.kjms.2016.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/08/2016] [Accepted: 01/11/2016] [Indexed: 11/20/2022] Open
Abstract
KMUP-3 (7-{2-[4-(4-nitrobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) displays cardioprotection and increases cardiac output, and is suggested to increase cardiac performance and improve myocardial infarction. To determine whether KMUP-3 improves outcomes in hypoperfused myocardium by inducing Ca(2+) sensitization to oppose protein kinase (PK)G-mediated Ca(2+) blockade, we measured left ventricular systolic blood pressure, maximal rates of pressure development, mean arterial pressure and heart rate in rats, and measured contractility and expression of PKs/RhoA/Rho kinase (ROCK)II in beating guinea pig left atria. Hemodynamic changes induced by KMUP-3 (0.5-3.0 mg/kg, intravenously) were inhibited by Y27632 [(R)-(+)-trans-4-1-aminoethyl)-N-(4-Pyridyl) cyclohexane carboxamide] and ketanserin (1 mg/kg, intravenously). In electrically stimulated left guinea pig atria, positive inotropy induced by KMUP-3 (0.1-100μM) was inhibited by the endothelial NO synthase (eNOS) inhibitors N-nitro-l-arginine methyl ester (L-NAME) and 7-nitroindazole, cyclic AMP antagonist SQ22536 [9-(terahydro-2-furanyl)-9H-purin-6-amine], soluble guanylyl cyclase (sGC) antagonist ODQ (1H-[1,2,4] oxadiazolo[4,3-a] quinoxalin-1-one), RhoA inhibitor C3 exoenzyme, β-blocker propranolol, 5-hydroxytryptamine 2A antagonist ketanserin, ROCK inhibitor Y27632 and KMUP-1 (7-{2-[4-(2-chlorobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) at 10μM. Western blotting assays indicated that KMUP-3 (0.1-10μM) increased PKA, RhoA/ROCKII, and PKC translocation and CIP-17 (an endogenous 17-kDa inhibitory protein) activation. In spontaneous right atria, KMUP-3 induced negative chronotropy that was blunted by 7-nitroindazole and atropine. In neonatal myocytes, L-NAME inhibited KMUP-3-induced eNOS phosphorylation and RhoA/ROCK activation. In H9c2 cells, Y-27632 (50μM) and PKG antagonist KT5823 [2,3,9,10,11,12-hexahydro-10R- methoxy-2,9-dimethyl-1-oxo-9S,12R-epoxy-1H-diindolo(1,2,3-fg:3',2',1'-kl) pyrrolo(3,4-i)(1,6)benzodiazocine-10-carboxylic acid, methyl ester] (3μM) reversed KMUP-3 (1-100μM)-induced Ca(2+)-entry blockade. GPCR agonist activity of KMUP-3 appeared opposed to KMUP-1, and increased cardiac output via Ca(2+) sensitization, and displayed cardioprotection via cyclic GMP/PKG-mediated myocardial preconditioning in animal studies.
Collapse
|
7
|
Wu X, Yang T, Zhou Q, Li S, Huang L. Additional use of a phosphodiesterase 5 inhibitor in patients with pulmonary hypertension secondary to chronic systolic heart failure: a meta-analysis. Eur J Heart Fail 2013; 16:444-53. [PMID: 24464734 DOI: 10.1002/ejhf.47] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/10/2013] [Accepted: 11/15/2013] [Indexed: 11/05/2022] Open
Abstract
AIMS Increased indiscriminate use of pulmonary artery hypertension-targeted drugs has been observed in patients with pulmonary hypertension (PH) secondary to heart failure. We performed a meta-analysis to evaluate the chronic effects of using phosphodiesterase 5 (PDE5) inhibitors to treat patients with PH secondary to chronic systolic heart failure. METHODS AND RESULTS PubMed, EMBASE, and the Cochrane Library were searched up to October 2013 for randomized controlled trials (RCTs) assessing PDE5 inhibitor treatments in PH patients secondary to chronic heart failure. Six RCTs involving 206 chronic systolic heart failure patients with PH complications were included. Sildenafil was used in all trials. Sildenafil treatment resulted in fewer hospital admissions compared with the placebo treatment (3.15% vs. 12.20%; risk ratio 0.29; 95% confidence interval 0.11-0.77). Various haemodynamic parameters were improved with additional sildenafil treatment, including reduced mean pulmonary artery pressure [weighted mean difference (WMD) -5.71 mmHg, P<0.05] and pulmonary vascular resistance (WMD -81.5 dynes/cm(-5), P<0.00001), increased LVEF (WMD 3.95%, P<0.01), and unchanged heart rate and blood pressure. The exercise capacity improved (oxygen consumption at peak exercise, WMD 3.20 mL/min(-1)/kg(-1), P<0.00001; ventilation to CO2 production slope, WMD -5.89, P<0.00001), and the clinical symptoms were relieved based on the breathlessness (WMD 7.72, P<0.00001), fatigue (WMD 2.28, P<0.05), and emotional functioning (WMD 5.92, P<0.00001) scores. CONCLUSIONS Additional sildenafil treatment is a potential therapeutic method to improve pulmonary exercise capacity and quality of life by ameliorating PH in patients with chronic systolic heart failure.
Collapse
Affiliation(s)
- Xiaojing Wu
- Cardiovascular Department of Xinqiao Hospital, Third Military Medical University, No.183 Xinqiao Street, Chongqing, China
| | | | | | | | | |
Collapse
|
8
|
Boyle KL, Leech E. A review of the pharmacology and clinical uses of pimobendan. J Vet Emerg Crit Care (San Antonio) 2013; 22:398-408. [PMID: 22928748 DOI: 10.1111/j.1476-4431.2012.00768.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To review the pharmacology, research developments, and clinical uses of pimobendan DATA SOURCES Original research articles and clinical studies from 1984 to August 2011. VETERINARY DATA SYNTHESIS Pimobendan is approved for use in dogs for the treatment of congestive heart failure (CHF) secondary to chronic valvular heart disease (CVHD) and dilated cardiomyopathy (DCM). Expert-based veterinary guidelines recommend the use of pimobendan in the management of acute, hospital-based therapy for patients with CHF attributable to CVHD. CONCLUSIONS The use of pimobendan, an inodilator with phosphodiesterase 3 (PDE3) inhibitory and calcium-sensitizing properties, is regarded as a component of the standard of care in the management of dogs with CHF secondary to both DCM and CVHD. Further studies are warranted to confirm the safety and efficacy of pimobendan for the off-label use of this drug in asymptomatic CVHD, pulmonary arterial hypertension, asymptomatic myocardial diseases, CHF from all other causes and in cats with CHF.
Collapse
Affiliation(s)
- Kimberly L Boyle
- VCA All-Care Animal Referral Center, Fountain Valley, CA, 92708, USA.
| | | |
Collapse
|
9
|
Michalski JM, Golden G, Ikari J, Rennard SI. PDE4: a novel target in the treatment of chronic obstructive pulmonary disease. Clin Pharmacol Ther 2011; 91:134-42. [PMID: 22130119 DOI: 10.1038/clpt.2011.266] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Phosphodiesterases (PDEs) are important modulators of inflammation and wound healing. In this capacity, specific targeting of PDEs for the treatment of many diseases, including chronic obstructive pulmonary disease (COPD), has been investigated. Currently, treatment of COPD is suboptimal. PDE4 modulates the inflammatory response of the lung, and inhibition of PDE4 may be a novel, COPD-specific approach toward more effective treatment strategies. This review describes the state of PDE4-inhibitor therapy for use in COPD treatment.
Collapse
Affiliation(s)
- J M Michalski
- Section of Pulmonary, Critical Care, Sleep and Allergy, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| | | | | | | |
Collapse
|
10
|
Hicks CW, Cui X, Sweeney DA, Li Y, Barochia A, Eichacker PQ. The potential contributions of lethal and edema toxins to the pathogenesis of anthrax associated shock. Toxins (Basel) 2011; 3:1185-202. [PMID: 22069762 PMCID: PMC3202877 DOI: 10.3390/toxins3091185] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 09/08/2011] [Accepted: 09/09/2011] [Indexed: 12/22/2022] Open
Abstract
Outbreaks of Bacillus anthracis in the US and Europe over the past 10 years have emphasized the health threat this lethal bacteria poses even for developed parts of the world. In contrast to cutaneous anthrax, inhalational disease in the US during the 2001 outbreaks and the newly identified injectional drug use form of disease in the UK and Germany have been associated with relatively high mortality rates. One notable aspect of these cases has been the difficulty in supporting patients once shock has developed. Anthrax bacilli produce several different components which likely contribute to this shock. Growing evidence indicates that both major anthrax toxins may produce substantial cardiovascular dysfunction. Lethal toxin (LT) can alter peripheral vascular function; it also has direct myocardial depressant effects. Edema toxin (ET) may have even more pronounced peripheral vascular effects than LT, including the ability to interfere with the actions of conventional vasopressors. Additionally, ET also appears capable of interfering with renal sodium and water retention. Importantly, the two toxins exert their actions via quite different mechanisms and therefore have the potential to worsen shock and outcome in an additive fashion. Finally, both toxins have the ability to inhibit host defense and microbial clearance, possibly contributing to the very high bacterial loads noted in patients dying with anthrax. This last point is clinically relevant since emerging data has begun to implicate other bacterial components such as anthrax cell wall in the shock and organ injury observed with infection. Taken together, accumulating evidence regarding the potential contribution of LT and ET to anthrax-associated shock supports efforts to develop adjunctive therapies that target both toxins in patients with progressive shock.
Collapse
Affiliation(s)
- Caitlin W. Hicks
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA;
- Howard Hughes Medical Institute-National Institutes of Health Research Scholar, National Institutes of Health, Bethesda, MD 20814, USA
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (X.C.); (Y.L.); (A.B.)
| | - Daniel A. Sweeney
- Medical Intensivist Program, Washington Hospital, Fremont, CA 94538, USA;
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (X.C.); (Y.L.); (A.B.)
| | - Amisha Barochia
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (X.C.); (Y.L.); (A.B.)
| | - Peter Q. Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (X.C.); (Y.L.); (A.B.)
- Author to whom correspondence should be addressed; ; Tel.: +1-301-496-9320; Fax: +1-301-402-1213
| |
Collapse
|
11
|
Combined ligand based pharmacophore modeling, virtual screening methods to identify critical chemical features of novel potential inhibitors for phosphodiesterase-5. J Taiwan Inst Chem Eng 2011. [DOI: 10.1016/j.jtice.2011.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
12
|
Abstract
Bacillus anthracis infection is rare in developed countries. However, recent outbreaks in the United States and Europe and the potential use of the bacteria for bioterrorism have focused interest on it. Furthermore, although anthrax was known to typically occur as one of three syndromes related to entry site of (i.e., cutaneous, gastrointestinal, or inhalational), a fourth syndrome including severe soft tissue infection in injectional drug users is emerging. Although shock has been described with cutaneous anthrax, it appears much more common with gastrointestinal, inhalational (5 of 11 patients in the 2001 outbreak in the United States), and injectional anthrax. Based in part on case series, the estimated mortalities of cutaneous, gastrointestinal, inhalational, and injectional anthrax are 1%, 25 to 60%, 46%, and 33%, respectively. Nonspecific early symptomatology makes initial identification of anthrax cases difficult. Clues to anthrax infection include history of exposure to herbivore animal products, heroin use, or clustering of patients with similar respiratory symptoms concerning for a bioterrorist event. Once anthrax is suspected, the diagnosis can usually be made with Gram stain and culture from blood or surgical specimens followed by confirmatory testing (e.g., PCR or immunohistochemistry). Although antibiotic therapy (largely quinolone-based) is the mainstay of anthrax treatment, the use of adjunctive therapies such as anthrax toxin antagonists is a consideration.
Collapse
Affiliation(s)
- Daniel A Sweeney
- Medical Intensivist Program, Washington Hospital, Fremont, California, USA
| | | | | | | | | |
Collapse
|
13
|
Francis SH, Blount MA, Corbin JD. Mammalian Cyclic Nucleotide Phosphodiesterases: Molecular Mechanisms and Physiological Functions. Physiol Rev 2011; 91:651-90. [DOI: 10.1152/physrev.00030.2010] [Citation(s) in RCA: 451] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The superfamily of cyclic nucleotide (cN) phosphodiesterases (PDEs) is comprised of 11 families of enzymes. PDEs break down cAMP and/or cGMP and are major determinants of cellular cN levels and, consequently, the actions of cN-signaling pathways. PDEs exhibit a range of catalytic efficiencies for breakdown of cAMP and/or cGMP and are regulated by myriad processes including phosphorylation, cN binding to allosteric GAF domains, changes in expression levels, interaction with regulatory or anchoring proteins, and reversible translocation among subcellular compartments. Selective PDE inhibitors are currently in clinical use for treatment of erectile dysfunction, pulmonary hypertension, intermittent claudication, and chronic pulmonary obstructive disease; many new inhibitors are being developed for treatment of these and other maladies. Recently reported x-ray crystallographic structures have defined features that provide for specificity for cAMP or cGMP in PDE catalytic sites or their GAF domains, as well as mechanisms involved in catalysis, oligomerization, autoinhibition, and interactions with inhibitors. In addition, major advances have been made in understanding the physiological impact and the biochemical basis for selective localization and/or recruitment of specific PDE isoenzymes to particular subcellular compartments. The many recent advances in understanding PDE structures, functions, and physiological actions are discussed in this review.
Collapse
Affiliation(s)
- Sharron H. Francis
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Medicine-Renal Division, Emory University School of Medicine, Atlanta, Georgia
| | - Mitsi A. Blount
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Medicine-Renal Division, Emory University School of Medicine, Atlanta, Georgia
| | - Jackie D. Corbin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Medicine-Renal Division, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
14
|
Sweeney DA, Cui X, Solomon SB, Vitberg DA, Migone TS, Scher D, Danner RL, Natanson C, Subramanian GM, Eichacker PQ. Anthrax lethal and edema toxins produce different patterns of cardiovascular and renal dysfunction and synergistically decrease survival in canines. J Infect Dis 2010; 202:1885-96. [PMID: 21067373 PMCID: PMC3061475 DOI: 10.1086/657408] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 07/14/2010] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND High mortality in the 2001 US and recent European anthrax outbreaks suggests that better understanding of the effects of the toxins produced by this bacterium is needed to improve treatment. METHODS AND RESULTS Here, 24-h edema (ETx) and lethal (LeTx) toxin infusions were investigated for 96 hin sedated canines receiving mechanical ventilation. The initial study compared similarly lethal doses of ETx (n=8) or LeTx (n=15) alone. ETx was 24 times less lethal than LeTx, and the median time to death in nonsurvivors (n=6 and n=9, respectively) was shorter with ETx (42 vs 67 h; P=.04). Compared with controls(n=9), both toxins decreased arterial and central venous pressures and systemic vascular resistance and increased heart rate, cardiac index, blood urea nitrogen (BUN) level, creatinine (Cr) concentration, BUN:Cr ratio, and hepatic transaminase levels (P ≤ .05 for toxin effect or time interaction). However, ETx stimulated early diuresis,reduced serum sodium levels, and had more pronounced vasodilatory effects, compared with LeTx, as reflected by greater or earlier central venous pressures, systemic vascular resistance, and changes in the BUN:Cr ratio(P ≤ .01). LeTx progressively decreased the left ventricular ejection fraction (P ≤ .002). In a subsequent study, a lethal dose of LeTx with an equimolar nonlethal ETx dose (n=8) increased mortality, compared with LeTx alone (n=8; P= .05). CONCLUSION Shock with ETx or LeTx may require differing supportive therapies, whereas toxin antagonists should likely target both toxins.
Collapse
Affiliation(s)
- Daniel A. Sweeney
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - David A. Vitberg
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | | | - Dara Scher
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Robert L. Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | | | - Peter Q. Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| |
Collapse
|
15
|
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: 736] [Impact Index Per Article: 49.1] [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
|
16
|
Pelletier AM, Venkataramana S, Miller KG, Bennett BM, Nair DG, Lourenssen S, Blennerhassett MG. Neuronal nitric oxide inhibits intestinal smooth muscle growth. Am J Physiol Gastrointest Liver Physiol 2010; 298:G896-907. [PMID: 20338922 DOI: 10.1152/ajpgi.00259.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hyperplasia of smooth muscle contributes to the thickening of the intestinal wall that is characteristic of inflammation, but the mechanisms of growth control are unknown. Nitric oxide (NO) from enteric neurons expressing neuronal NO synthase (nNOS) might normally inhibit intestinal smooth muscle cell (ISMC) growth, and this was tested in vitro. In ISMC from the circular smooth muscle of the adult rat colon, chemical NO donors inhibited [(3)H]thymidine uptake in response to FCS, reducing this to baseline without toxicity. This effect was inhibited by the guanylyl cyclase inhibitor ODQ and potentiated by the phosphodiesterase-5 inhibitor zaprinast. Inhibition was mimicked by 8-bromo (8-Br)-cGMP, and ELISA measurements showed increased levels of cGMP but not cAMP in response to sodium nitroprusside. However, 8-Br-cAMP and cilostamide also showed inhibitory actions, suggesting an additional role for cAMP. Via a coculture model of ISMC and myenteric neurons, immunocytochemistry and image analysis showed that innervation reduced bromodeoxyuridine uptake by ISMC. Specific blockers of nNOS (7-NI, NAAN) significantly increased [(3)H]thymidine uptake in response to a standard stimulus, showing that nNOS activity normally inhibits ISMC growth. In vivo, nNOS axon number was reduced threefold by day 1 of trinitrobenzene sulfonic acid-induced rat colitis, preceding the hyperplasia of ISMC described earlier in this model. We conclude that NO can inhibit ISMC growth primarily via a cGMP-dependent mechanism. Functional evidence that NO derived from nNOS causes inhibition of ISMC growth in vitro predicts that the loss of nNOS expression in colitis contributes to ISMC hyperplasia in vivo.
Collapse
Affiliation(s)
- Anne-Marie Pelletier
- Gastrointestinal Diseases Research Unit, Queen's Univ., 76 Stuart St., Kingston, Ontario K7L 2V6
| | | | | | | | | | | | | |
Collapse
|
17
|
Novel 6-N-arylcarboxamidopyrazolo[4,3-d]pyrimidin-7-one derivatives as potential anti-cancer agents. Bioorg Med Chem Lett 2010; 20:1630-3. [DOI: 10.1016/j.bmcl.2010.01.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 12/08/2009] [Accepted: 01/13/2010] [Indexed: 11/19/2022]
|
18
|
Houslay MD. Underpinning compartmentalised cAMP signalling through targeted cAMP breakdown. Trends Biochem Sci 2010; 35:91-100. [DOI: 10.1016/j.tibs.2009.09.007] [Citation(s) in RCA: 341] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 09/28/2009] [Accepted: 09/29/2009] [Indexed: 01/14/2023]
|
19
|
Van Tassell BW, Radwanski P, Movsesian M, Munger MA. Combination therapy with beta-adrenergic receptor antagonists and phosphodiesterase inhibitors for chronic heart failure. Pharmacotherapy 2009; 28:1523-30. [PMID: 19025433 DOI: 10.1592/phco.28.12.1523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abstract Rational use of phosphodiesterase inhibitors represents an ongoing controversy in contemporary pharmacotherapy for heart failure. In randomized clinical trials, phosphodiesterase inhibitors increased cardiac output at the expense of worsening the rates of sudden cardiac death and cardiovascular mortality. Preliminary findings from ongoing clinical and preclinical investigations of phosphodiesterase activity suggest that combined use of phosphodiesterase inhibitors with beta-adrenergic antagonists may prevent these adverse outcomes. Compartmentation of cyclic adenosine 3',5'-monophosphate signaling may prove critical in determining myocardial response to combination therapy.
Collapse
Affiliation(s)
- Benjamin W Van Tassell
- Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA.
| | | | | | | |
Collapse
|
20
|
Control of pulmonary vascular tone during exercise in health and pulmonary hypertension. Pharmacol Ther 2008; 119:242-63. [PMID: 18586325 DOI: 10.1016/j.pharmthera.2008.04.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 04/29/2008] [Indexed: 11/24/2022]
Abstract
Despite the importance of the pulmonary circulation as a determinant of exercise capacity in health and disease, studies into the regulation of pulmonary vascular tone in the healthy lung during exercise are scarce. This review describes the current knowledge of the role of various endogenous vasoactive mechanisms in the control of pulmonary vascular tone at rest and during exercise. Recent studies demonstrate an important role for endothelial factors (NO and endothelin) and neurohumoral factors (noradrenaline, acetylcholine). Moreover, there is evidence that natriuretic peptides, reactive oxygen species and phosphodiesterase activity can influence resting pulmonary vascular tone, but their role in the control of pulmonary vascular tone during exercise remains to be determined. K-channels are purported end-effectors in control of pulmonary vascular tone. However, K(ATP) channels do not contribute to regulation of pulmonary vascular tone, while the role of K(V) and K(Ca) channels at rest and during exercise remains to be determined. Pulmonary hypertension is associated with alterations in pulmonary vascular function and structure, resulting in blunted pulmonary vasodilatation during exercise and impaired exercise capacity. Although there is a paucity of studies pertaining to the regulation of pulmonary vascular tone during exercise in idiopathic pulmonary hypertension, the few studies that have been performed in models of pulmonary hypertension secondary to left ventricular dysfunction suggest altered control of pulmonary vascular tone during exercise. Since the increased pulmonary vascular tone during exercise limits exercise capacity, future studies are needed to investigate the vasomotor mechanisms that are responsible for the blunted exercise-induced pulmonary vasodilatation in pulmonary hypertension.
Collapse
|
21
|
Chen G, Wang H, Robinson H, Cai J, Wan Y, Ke H. An insight into the pharmacophores of phosphodiesterase-5 inhibitors from synthetic and crystal structural studies. Biochem Pharmacol 2008; 75:1717-28. [PMID: 18346713 PMCID: PMC2409583 DOI: 10.1016/j.bcp.2008.01.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 01/23/2008] [Accepted: 01/24/2008] [Indexed: 10/22/2022]
Abstract
Selective inhibitors of cyclic nucleotide phosphodiesterase-5 (PDE5) have been used as drugs for treatment of male erectile dysfunction and pulmonary hypertension. An insight into the pharmacophores of PDE5 inhibitors is essential for development of second generation of PDE5 inhibitors, but has not been completely illustrated. Here we report the synthesis of a new class of the sildenafil derivatives and a crystal structure of the PDE5 catalytic domain in complex with 5-(2-ethoxy-5-(sulfamoyl)-3-thienyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (12). Inhibitor 12 induces conformational change of the H-loop (residues 660-683), which is different from any of the known PDE5 structures. The pyrazolopyrimidinone groups of 12 and sildenafil are well superimposed, but their sulfonamide groups show a positional difference of as much as 1.5A. The structure-activity analysis suggests that a small hydrophobic pocket and the H-loop of PDE5 are important for the inhibitor affinity, in addition to two common elements for binding of almost all the PDE inhibitors: the stack against the phenylalanine and the hydrogen bond with the invariant glutamine. However, the PDE5-12 structure does not provide a full explanation to affinity changes of the inhibitors. Thus alternatives such as conformational change of the M-loop are open and further structural study is required.
Collapse
Affiliation(s)
- Gong Chen
- School of Chemistry and Chemical Engineering, and Center of Structure Biology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Huanchen Wang
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Howard Robinson
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Jiwen Cai
- School of Chemistry and Chemical Engineering, and Center of Structure Biology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Yiqian Wan
- School of Chemistry and Chemical Engineering, and Center of Structure Biology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| |
Collapse
|
22
|
Illi B, Russo CD, Colussi C, Rosati J, Pallaoro M, Spallotta F, Rotili D, Valente S, Ragone G, Martelli F, Biglioli P, Steinkuhler C, Gallinari P, Mai A, Capogrossi MC, Gaetano C. Nitric Oxide Modulates Chromatin Folding in Human Endothelial Cells via Protein Phosphatase 2A Activation and Class II Histone Deacetylases Nuclear Shuttling. Circ Res 2008; 102:51-8. [DOI: 10.1161/circresaha.107.157305] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nitric oxide (NO) modulates important endothelial cell (EC) functions and gene expression by a molecular mechanism which is still poorly characterized. Here we show that in human umbilical vein ECs (HUVECs) NO inhibited serum-induced histone acetylation and enhanced histone deacetylase (HDAC) activity. By immunofluorescence and Western blot analyses it was found that NO induced class II HDAC4 and 5 nuclear shuttling and that class II HDACs selective inhibitor MC1568 rescued serum-dependent histone acetylation above control level in NO-treated HUVECs. In contrast, class I HDACs inhibitor MS27–275 had no effect, indicating a specific role for class II HDACs in NO-dependent histone deacetylation. In addition, it was found that NO ability to induce HDAC4 and HDAC5 nuclear shuttling involved the activation of the protein phosphatase 2A (PP2A). In fact, HDAC4 nuclear translocation was impaired in ECs expressing small-t antigen and exposed to NO. Finally, in cells engineered to express a HDAC4-Flag fusion protein, NO induced the formation of a macromolecular complex including HDAC4, HDAC3, HDAC5, and an active PP2A. The present results show that NO-dependent PP2A activation plays a key role in class II HDACs nuclear translocation.
Collapse
Affiliation(s)
- Barbara Illi
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Claudio Dello Russo
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Claudia Colussi
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Jessica Rosati
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Michele Pallaoro
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Francesco Spallotta
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Dante Rotili
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Sergio Valente
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Gianluca Ragone
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Fabio Martelli
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Paolo Biglioli
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Christian Steinkuhler
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Paola Gallinari
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Antonello Mai
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Maurizio C. Capogrossi
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| | - Carlo Gaetano
- From the Laboratorio di Biologia Vascolare e Terapia Genica (B.I., F.S.), Centro Cardiologico Fondazione “I. Monzino”, IRCCS, Milan; Istituto di Ricerche di Biologia Molecolare I.R.B.M. P. Angeletti (C.D.R., C.S., P.G.), Via Pontina km 30 600, Pomezia, Rome; Laboratorio di Patologia Vascolare (C.C., J.R., G.R., F.M., M.C.C.), Istituto Dermopatico dell’ Immacolata-IRCCS, Rome; Università di Siena (M.P.), Siena; Dipartimento di Cardiochirurgia (P.B.), Centro Cardiologico Fondazione “I. Monzino”,
| |
Collapse
|
23
|
Wang H, Ye M, Robinson H, Francis SH, Ke H. Conformational variations of both phosphodiesterase-5 and inhibitors provide the structural basis for the physiological effects of vardenafil and sildenafil. Mol Pharmacol 2008; 73:104-10. [PMID: 17959709 PMCID: PMC2950070 DOI: 10.1124/mol.107.040212] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Vardenafil has higher affinity to phosphodiesterase-5 (PDE5) than sildenafil and lower administered dosage for the treatment of erectile dysfunction. However, the molecular basis for these differences is puzzling because two drugs have similar chemical structures. Reported here is a crystal structure of the fully active and nonmutated PDE5A1 catalytic domain in complex with vardenafil. The structure shows that the conformation of the H-loop in the PDE5A1-vardenafil complex is different from those of any known structures of the unliganded PDE5 and its complexes with the inhibitors. In addition, the molecular configuration of vardenafil differs from that of sildenafil when bound to PDE5. It is noteworthy that the binding of vardenafil causes loss of the divalent metal ions that have been observed in all the previously published PDE structures. The conformational variation of both PDE5 and the inhibitors provides structural insight into the different potencies of the drugs.
Collapse
Affiliation(s)
- Huanchen Wang
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | | | | | | | | |
Collapse
|
24
|
McCahill AC, Huston E, Li X, Houslay MD. PDE4 associates with different scaffolding proteins: modulating interactions as treatment for certain diseases. Handb Exp Pharmacol 2008:125-66. [PMID: 18491051 DOI: 10.1007/978-3-540-72843-6_6] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
cAMP is an ubiquitous second messenger that is crucial to many cellular processes. The sole means of terminating the cAMP signal is degradation by cAMP phosphodiesterases (PDEs). The PDE4 family is of particular interest because PDE4 inhibitors have therapeutic potential for the treatment of various inflammatory and auto-immune diseases and also have anti-depressant and memory-enhancing effects. The subcellular targeting of PDE4 isoforms is fundamental to the compartmentalization of cAMP signaling pathways and is largely achieved via proteinprotein interactions. Increased knowledge of these protein-protein interactions and their regulatory properties could aid in the design of novel isoform-specific inhibitors with improved efficacy and fewer prohibitive side effects.
Collapse
Affiliation(s)
- A C McCahill
- Molecular Pharmacology Group, Division of Biochemistry and Molecular Biology, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow, G128QQ, UK
| | | | | | | |
Collapse
|
25
|
PDE inhibitors in psychiatry--future options for dementia, depression and schizophrenia? Drug Discov Today 2007; 12:870-8. [PMID: 17933689 DOI: 10.1016/j.drudis.2007.07.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 07/21/2007] [Accepted: 07/23/2007] [Indexed: 02/07/2023]
Abstract
Phosphodiesterases are key enzymes in cellular signalling pathways. They degrade cyclic nucleotides and their inhibition via specific inhibitors offers unique 'receptor-independent' opportunities to modify cellular function. An increasing number of in vitro and animal model studies point to innovative treatment options in neurology and psychiatry. This review critiques a selection of recent studies and developments with a focus on dementia/neuroprotection, depression and schizophrenia. Despite increased interest among the clinical neurosciences, there are still no approved PDE inhibitors for clinical use in neurology or psychiatry. Adverse effects are a major impediment for clinical approval. It is therefore necessary to search for more specific inhibitors at the level of different PDE sub-families and isoforms.
Collapse
|
26
|
Vandeput F, Wolda SL, Krall J, Hambleton R, Uher L, McCaw KN, Radwanski PB, Florio V, Movsesian MA. Cyclic nucleotide phosphodiesterase PDE1C1 in human cardiac myocytes. J Biol Chem 2007; 282:32749-57. [PMID: 17726023 DOI: 10.1074/jbc.m703173200] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Isoforms in the PDE1 family of cyclic nucleotide phosphodiesterases were recently found to comprise a significant portion of the cGMP-inhibited cAMP hydrolytic activity in human hearts. We examined the expression of PDE1 isoforms in human myocardium, characterized their catalytic activity, and quantified their contribution to cAMP hydrolytic and cGMP hydrolytic activity in subcellular fractions of this tissue. Western blotting with isoform-selective anti-PDE1 monoclonal antibodies showed PDE1C1 to be the principal isoform expressed in human myocardium. Immunohistochemical analysis showed that PDE1C1 is distributed along the Z-lines and M-lines of cardiac myocytes in a striated pattern that differs from that of the other major dual-specificity cyclic nucleotide phosphodiesterase in human myocardium, PDE3A. Most of the PDE1C1 activity was recovered in soluble fractions of human myocardium. It binds both cAMP and cGMP with K(m) values of approximately 1 microm and hydrolyzes both substrates with similar catalytic rates. PDE1C1 activity in subcellular fractions was quantified using a new PDE1-selective inhibitor, IC295. At substrate concentrations of 0.1 microm, PDE1C1 constitutes the great majority of cAMP hydrolytic and cGMP hydrolytic activity in soluble fractions and the majority of cGMP hydrolytic activity in microsomal fractions, whereas PDE3 constitutes the majority of cAMP hydrolytic activity in microsomal fractions. These results indicate that PDE1C1 is expressed at high levels in human cardiac myocytes with an intracellular distribution distinct from that of PDE3A and that it may have a role in the integration of cGMP-, cAMP- and Ca(2+)-mediated signaling in these cells.
Collapse
Affiliation(s)
- Fabrice Vandeput
- Cardiology Section, Veterans Affairs Salt Lake City Health Care System, 500 Foothill Boulevard, Salt Lake City, UT 84148, USA
| | | | | | | | | | | | | | | | | |
Collapse
|