1
|
Kim Y, Kwon D, Kim SS, Lee K, Yoon H. Echocardiographic changes in the progress of reverse shunt and improvement to left-to-right shunt after medical treatment in dogs with bidirectional patent ductus arteriosus or ventricular septal defect: A report of two cases. Vet Med Sci 2023; 9:1044-1052. [PMID: 36716384 DOI: 10.1002/vms3.1081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Two Maltese dogs were referred for evaluation of a congenital heart disease: one was diagnosed with patent ductus arteriosus and the other was diagnosed with a ventricular septal defect. The PDA patient was diagnosed with congenital heart disease 2 weeks ago and the VSD patient about 11 months ago at another hospital. Echocardiographic findings revealed a bidirectional shunt condition, and the dogs were treated with medical management using sildenafil and oxygen inhalation. After medical management, the dogs returned to clinically normal conditions, and echocardiographic findings revealed a return to left-to-right shunt tendency. These dogs had no clinical signs associated with heart disease 3 years after treatment. This case report describes changes in echocardiography findings according to the progression of the reverse shunt and the possibility of improvement to a left-to-right shunt after medical treatment.
Collapse
Affiliation(s)
- Yein Kim
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Danbee Kwon
- Bundang Leaders Animal Medical Center, Seongnam, South Korea
| | | | - Kichang Lee
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Hakyoung Yoon
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
2
|
Mulvaney EP, Renzo F, Adão R, Dupre E, Bialesova L, Salvatore V, Reid HM, Conceição G, Grynblat J, Llucià-Valldeperas A, Michel JB, Brás-Silva C, Laurent CE, Howard LS, Montani D, Humbert M, Vonk Noordegraaf A, Perros F, Mendes-Ferreira P, Kinsella BT. The thromboxane receptor antagonist NTP42 promotes beneficial adaptation and preserves cardiac function in experimental models of right heart overload. Front Cardiovasc Med 2022; 9:1063967. [PMID: 36588576 PMCID: PMC9794752 DOI: 10.3389/fcvm.2022.1063967] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a progressive disease characterized by increased pulmonary artery pressure leading to right ventricular (RV) failure. While current PAH therapies improve patient outlook, they show limited benefit in attenuating RV dysfunction. Recent investigations demonstrated that the thromboxane (TX) A2 receptor (TP) antagonist NTP42 attenuates experimental PAH across key hemodynamic parameters in the lungs and heart. This study aimed to validate the efficacy of NTP42:KVA4, a novel oral formulation of NTP42 in clinical development, in preclinical models of PAH while also, critically, investigating its direct effects on RV dysfunction. Methods The effects of NTP42:KVA4 were evaluated in the monocrotaline (MCT) and pulmonary artery banding (PAB) models of PAH and RV dysfunction, respectively, and when compared with leading standard-of-care (SOC) PAH drugs. In addition, the expression of the TP, the target for NTP42, was investigated in cardiac tissue from several other related disease models, and from subjects with PAH and dilated cardiomyopathy (DCM). Results In the MCT-PAH model, NTP42:KVA4 alleviated disease-induced changes in cardiopulmonary hemodynamics, pulmonary vascular remodeling, inflammation, and fibrosis, to a similar or greater extent than the PAH SOCs tested. In the PAB model, NTP42:KVA4 improved RV geometries and contractility, normalized RV stiffness, and significantly increased RV ejection fraction. In both models, NTP42:KVA4 promoted beneficial RV adaptation, decreasing cellular hypertrophy, and increasing vascularization. Notably, elevated expression of the TP target was observed both in RV tissue from these and related disease models, and in clinical RV specimens of PAH and DCM. Conclusion This study shows that, through antagonism of TP signaling, NTP42:KVA4 attenuates experimental PAH pathophysiology, not only alleviating pulmonary pathologies but also reducing RV remodeling, promoting beneficial hypertrophy, and improving cardiac function. The findings suggest a direct cardioprotective effect for NTP42:KVA4, and its potential to be a disease-modifying therapy in PAH and other cardiac conditions.
Collapse
Affiliation(s)
- Eamon P. Mulvaney
- ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Fabiana Renzo
- ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Rui Adão
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine of the University of Porto, Porto, Portugal
| | | | - Lucia Bialesova
- ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Viviana Salvatore
- ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Helen M. Reid
- ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Glória Conceição
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Julien Grynblat
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France,INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Aida Llucià-Valldeperas
- PHEniX Laboratory, Department of Pulmonary Medicine, Amsterdam UMC (Location VUMC), Amsterdam Cardiovascular Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands,Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, Netherlands
| | | | - Carmen Brás-Silva
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Charles E. Laurent
- IPS Therapeutique Inc., Sherbrooke, QC, Canada,ToxiPharm Laboratories Inc., Ste-Catherine-de-Hatley, QC, Canada
| | - Luke S. Howard
- Imperial College London, National Heart and Lung Institute, London, United Kingdom
| | - David Montani
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France,INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France,AP-HP, Dept of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France,INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France,AP-HP, Dept of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Anton Vonk Noordegraaf
- PHEniX Laboratory, Department of Pulmonary Medicine, Amsterdam UMC (Location VUMC), Amsterdam Cardiovascular Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Frédéric Perros
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France,INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France,Paris-Porto Pulmonary Hypertension Collaborative Laboratory (3PH), INSERM UMR_S 999, Université Paris-Saclay, Le Kremlin-Bicêtre, France,INSERM, INRAE, CarMeN Laboratory and Centre de Recherche en Nutrition Humaine Rhône-Alpes (CRNH-RA), Claude Bernard University Lyon 1, University of Lyon, Lyon, France
| | - Pedro Mendes-Ferreira
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine of the University of Porto, Porto, Portugal,Paris-Porto Pulmonary Hypertension Collaborative Laboratory (3PH), INSERM UMR_S 999, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - B. Therese Kinsella
- ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland,UCD School of Biomolecular and Biomedical Research, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland,*Correspondence: B. Therese Kinsella,
| |
Collapse
|
3
|
Kv7 Channels in Cyclic-Nucleotide Dependent Relaxation of Rat Intra-Pulmonary Artery. Biomolecules 2022; 12:biom12030429. [PMID: 35327621 PMCID: PMC8946781 DOI: 10.3390/biom12030429] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension is treated with drugs that stimulate cGMP or cAMP signalling. Both nucleotides can activate Kv7 channels, leading to smooth muscle hyperpolarisation, reduced Ca2+ influx and relaxation. Kv7 activation by cGMP contributes to the pulmonary vasodilator action of nitric oxide, but its contribution when dilation is evoked by the atrial natriuretic peptide (ANP) sensitive guanylate cyclase, or cAMP, is unknown. Small vessel myography was used to investigate the ability of Kv7 channel blockers to interfere with pulmonary artery relaxation when cyclic nucleotide pathways were stimulated in different ways. The pan-Kv7 blockers, linopirdine and XE991, caused substantial inhibition of relaxation evoked by NO donors and ANP, as well as endothelium-dependent dilators, the guanylate cyclase stimulator, riociguat, and the phosphodiesterase-5 inhibitor, sildenafil. Maximum relaxation was reduced without a change in sensitivity. The blockers had relatively little effect on cAMP-mediated relaxation evoked by forskolin, isoprenaline or treprostinil. The Kv7.1-selective blocker, HMR1556, had no effect on cGMP or cAMP-dependent relaxation. Western blot analysis demonstrated the presence of Kv7.1 and Kv7.4 proteins, while selective activators of Kv7.1 and Kv7.4 homomeric channels, but not Kv7.5, caused pulmonary artery relaxation. It is concluded that Kv7.4 channels contribute to endothelium-dependent dilation and the effects of drugs that act by stimulating cGMP, but not cAMP, signalling.
Collapse
|
4
|
Tomita N, Hotta Y, Naiki-Ito A, Sanagawa A, Kataoka T, Furukawa-Hibi Y, Takahashi S, Kimura K. Protective effects of tadalafil on damaged podocytes in an adriamycin-induced nephrotic syndrome model. J Pharmacol Sci 2022; 149:53-59. [DOI: 10.1016/j.jphs.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022] Open
|
5
|
PDE-Mediated Cyclic Nucleotide Compartmentation in Vascular Smooth Muscle Cells: From Basic to a Clinical Perspective. J Cardiovasc Dev Dis 2021; 9:jcdd9010004. [PMID: 35050214 PMCID: PMC8777754 DOI: 10.3390/jcdd9010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases are important causes of mortality and morbidity worldwide. Vascular smooth muscle cells (SMCs) are major components of blood vessels and are involved in physiologic and pathophysiologic conditions. In healthy vessels, vascular SMCs contribute to vasotone and regulate blood flow by cyclic nucleotide intracellular pathways. However, vascular SMCs lose their contractile phenotype under pathological conditions and alter contractility or signalling mechanisms, including cyclic nucleotide compartmentation. In the present review, we focus on compartmentalized signaling of cyclic nucleotides in vascular smooth muscle. A deeper understanding of these mechanisms clarifies the most relevant axes for the regulation of vascular tone. Furthermore, this allows the detection of possible changes associated with pathological processes, which may be of help for the discovery of novel drugs.
Collapse
|
6
|
Al-Kuraishy HM, Al-Gareeb AI, Al-Niemi MS, Al-Buhadily AK, Al-Harchan NA, Lugnier C. COVID-19 and Phosphodiesterase Enzyme Type 5 Inhibitors. J Microsc Ultrastruct 2020; 8:141-145. [PMID: 33623736 PMCID: PMC7883493 DOI: 10.4103/jmau.jmau_63_20] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/25/2020] [Accepted: 08/06/2020] [Indexed: 01/14/2023] Open
Abstract
COVID-19 pathology is mainly associated to a pulmonary disease which sometimes might result in an uncontrollable storm related to inflammatory diseases which could be fatal. It is well known that phosphodiesterase enzyme type 5 inhibitors (PDE5Is), such as sildenafil, have been successfully developed for the treatment of pulmonary arterial hypertension; interestingly, more recently, it was shown that PDE5Is might be also anti-inflammatory. Therefore, it would be of interest to question about the use of PDE5Is to overcome the COVID-19 storm, as much as PDE5 is mainly present in the lung tissues and vessels.
Collapse
Affiliation(s)
- Hayder M. Al-Kuraishy
- Department of Clinical Pharmacology, Medicine and Therapeutic, Medical Faculty, College of Medicine, Al-Mustansiriyia University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology, Medicine and Therapeutic, Medical Faculty, College of Medicine, Al-Mustansiriyia University, Baghdad, Iraq
| | - Marwa S. Al-Niemi
- Department of Clinical Pharmacology, Medicine and Therapeutic, Medical Faculty, College of Medicine, Al-Farahedi University, Baghdad, Iraq
| | - Ali K. Al-Buhadily
- Department of Clinical Pharmacology, Medicine and Therapeutic, Medical Faculty, College of Medicine, Al-Mustansiriyia University, Baghdad, Iraq
| | - Nasser A. Al-Harchan
- Department of Clinical Pharmacology, College of Dentistry, Al-Rasheed University, Baghdad, Iraq
| | - Claire Lugnier
- Department EA 3072 “Mitochondria, Oxidative Stress and Muscular Protection”, Institute of Physiology, Faculty of Medicine, Strasbourg Cedex, France
| |
Collapse
|
7
|
Perros F, de Man FS, Bogaard HJ, Antigny F, Simonneau G, Bonnet S, Provencher S, Galiè N, Humbert M. Use of β-Blockers in Pulmonary Hypertension. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003703. [DOI: 10.1161/circheartfailure.116.003703] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/24/2017] [Indexed: 12/13/2022]
Abstract
Contrasting with the major attention that left heart failure has received, right heart failure remains understudied both at the preclinical and clinical levels. However, right ventricle failure is a major predictor of outcomes in patients with precapillary pulmonary hypertension because of pulmonary arterial hypertension, and in patients with postcapillary pulmonary hypertension because of left heart disease. In pulmonary hypertension, the status of the right ventricle is one of the most important predictors of both morbidity and mortality. Paradoxically, there are currently no approved therapies targeting the right ventricle in pulmonary hypertension. By analogy with the key role of β-blockers in the management of left heart failure, some authors have proposed to use these agents to support the right ventricle function in pulmonary hypertension. In this review, we summarize the current knowledge on the use of β-blockers in pulmonary hypertension.
Collapse
Affiliation(s)
- Frédéric Perros
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Frances S. de Man
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Harm J. Bogaard
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Fabrice Antigny
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Gérald Simonneau
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Sébastien Bonnet
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Steeve Provencher
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Nazzareno Galiè
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| | - Marc Humbert
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (F.P., F.A., G.S., M.H.); Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (F.P., F.A., G.S., M.H.); Department of Pulmonology, VU University Medical Centre, Amsterdam, The Netherlands (F.S.d.M., H.J.B.); Pulmonary Hypertension Research Group, Centre de Recherche de
| |
Collapse
|
8
|
Thakkar N, Gonzalez D, Cohen-Wolkowiez M, Massaro M, Bernhardt J, Zane NR, Laughon MM. An opportunistic study evaluating pharmacokinetics of sildenafil for the treatment of pulmonary hypertension in infants. J Perinatol 2016; 36:744-7. [PMID: 27171763 PMCID: PMC5002251 DOI: 10.1038/jp.2016.79] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/23/2016] [Accepted: 04/01/2016] [Indexed: 12/02/2022]
Abstract
OBJECTIVE The objective of this study is to assess sildenafil and N-desmethyl sildenafil (DMS) exposure in infants receiving sildenafil for the treatment of pulmonary hypertension (PH). STUDY DESIGN Data were collected from six infants receiving sildenafil for the treatment of PH and plasma samples were collected at the time of routine laboratory blood draws. The echocardiography results were assessed for improvement in right ventricular (RV) hypertension following sildenafil treatment. RESULT The median (range) sildenafil and DMS concentrations were 27.4 ng ml(-1) (2.6 to 434.0) and 105.5 ng ml(-1) (3.6 to 314.0), respectively. The median metabolite-to-parent ratio was higher in infants receiving co-medications that can induce cytochrome P450 (CYP) enzymes (5.2 vs 0.7). The echocardiography results showed improvement in RV hypertension for the majority of infants (5/6). CONCLUSION The concentrations of sildenafil and DMS were within the previously observed ranges. Our results suggest that caution may be warranted when CYP-related co-medications are administered during sildenafil treatment for PH.
Collapse
Affiliation(s)
- Nilay Thakkar
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthew Massaro
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice Bernhardt
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicole R. Zane
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew M. Laughon
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
9
|
Sisignano M, Parnham MJ, Geisslinger G. Drug Repurposing for the Development of Novel Analgesics. Trends Pharmacol Sci 2015; 37:172-183. [PMID: 26706620 DOI: 10.1016/j.tips.2015.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 01/12/2023]
Abstract
Drug development consumes huge amounts of time and money and the search for novel analgesics, which are urgently required, is particularly difficult, having resulted in many setbacks in the past. Drug repurposing - the identification of new uses for existing drugs - is an alternative approach, which bypasses most of the time- and cost-consuming components of drug development. Recent, unexpected findings suggest a role for several existing drugs, such as minocycline, ceftriaxone, sivelestat, and pioglitazone, as novel analgesics in chronic and neuropathic pain states. Here, we discuss these findings as well as their proposed antihyperalgesic mechanisms and outline the merits of pathway-based repurposing screens, in combination with bioinformatics and novel cellular reprogramming techniques, for the identification of novel analgesics.
Collapse
Affiliation(s)
- Marco Sisignano
- Institute of Clinical Pharmacology, pharmazentrum Frankfurt/ZAFES, University Hospital of Goethe-University, 60590 Frankfurt am Main, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project Group Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, pharmazentrum Frankfurt/ZAFES, University Hospital of Goethe-University, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project Group Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
10
|
Yang JY, Yang G, Ren J, Zhao J, Li S. Caffeine Suppresses GABA Receptor-Mediated Current in Rat Primary Sensory Neurons via Inhibition of Intracellular Phosphodiesterase. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9506-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
11
|
Isidori AM, Cornacchione M, Barbagallo F, Di Grazia A, Barrios F, Fassina L, Monaco L, Giannetta E, Gianfrilli D, Garofalo S, Zhang X, Chen X, Xiang YK, Lenzi A, Pellegrini M, Naro F. Inhibition of type 5 phosphodiesterase counteracts β2-adrenergic signalling in beating cardiomyocytes. Cardiovasc Res 2015; 106:408-20. [PMID: 25852085 DOI: 10.1093/cvr/cvv123] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 03/20/2015] [Indexed: 12/31/2022] Open
Abstract
AIMS Compartmentalization of cAMP and PKA activity in cardiac muscle cells plays a key role in maintaining basal and enhanced contractility stimulated by sympathetic nerve activity. In cardiomyocytes, activation of adrenergic receptor increases cAMP production, which is countered by the hydrolytic activity of selective phosphodiesterases (PDEs). The intracellular regional dynamics of cAMP production and hydrolysis modulate downstream signals resulting in different biological responses. The interplay between beta receptors (βARs) signalling and phosphodiesterase 5 (PDE5) activity remains to be addressed. METHODS AND RESULTS Using combined strategies with pharmacological inhibitors and genetic deletion of PDEs and βAR isoforms, we revealed a specific pool of cAMP that is under dual regulation by PDE2 and, indirectly, PDE5 activity. Inhibition of PDE5 with sildenafil produces a cGMP-dependent activation of PDE2 that attenuates cAMP generation induced by βAR agonists, with concomitant modulation of stimulated contraction rate and calcium transients. PDE2 haploinsufficiency abolished the effects of sildenafil. The negative chronotropic effect of PDE5 inhibition through PDE2 activation was also observed in sinoatrial node tissue from adult mice. PDE5 inhibition selectively lowered contraction rate stimulated by β2AR, but not β1AR activation, supporting a compartmentalization of the cGMP-modulated pool of cAMP. CONCLUSION These data identify a new effect of PDE5 inhibitors on the modulation of cardiomyocyte response to adrenergic stimulation via PDE5-PDE2-mediated cross-talk.
Collapse
Affiliation(s)
- Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marisa Cornacchione
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Di Grazia
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University, Rome, Italy
| | - Florencia Barrios
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University, Rome, Italy
| | - Lorenzo Fassina
- Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvio Garofalo
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Xiaoxiao Zhang
- Department of Physiology, Temple University, Philadelphia, PA, USA
| | - Xiongwen Chen
- Department of Physiology, Temple University, Philadelphia, PA, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Manuela Pellegrini
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University, Rome, Italy
| |
Collapse
|
12
|
Kruangtip O, Chootip K, Temkitthawon P, Changwichit K, Chuprajob T, Changtam C, Suksamrarn A, Khorana N, Scholfield CN, Ingkaninan K. Curcumin analogues inhibit phosphodiesterase-5 and dilate rat pulmonary arteries. J Pharm Pharmacol 2014; 67:87-95. [DOI: 10.1111/jphp.12302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 07/04/2014] [Indexed: 12/13/2022]
Abstract
Abstract
Objectives
Phosphodiesterase (PDE)-5 inhibitors are useful as vasodilators for the treatment of pulmonary arterial hypertension. We aimed to study curcumin analogues for PDE5 inhibitory activity and vasorelaxation of rat pulmonary arteries.
Methods
Three natural curcuminoids (1–3) and six synthetic analogues (4–9) were tested for PDE5 and PDE6 inhibitory activities using enzymatic radioassay. Their vasorelaxation was measured using freshly isolated segments of rat pulmonary artery and aorta.
Key findings
Curcuminoids (1–3) mildly inhibited PDE5 (half maximal inhibitory concentration (IC50) = 18 µm): the metamethoxyl of curcumin was important for PDE5 inhibition. But hydroxyl rearrangements, removing both methoxyls and one ketomethylene, yielded the potent 7 and 9 (IC50 = 4 µm) (compared with sildenafil, IC50 = 0.03 µm). Only 1, 3 and 4 were PDE5 selective over PDE6. Triazole-carboxylic addition provided water-solubility while preserving potency. All analogues possessed concentration-dependent vasorelaxant activity on pulmonary arteries (40% of maximal effective concentration (EC40) = 29–90 µm, maximum response = 60–90% at 300 µm), while compounds (1–8) were weakly acting in aorta (maximum response <40%). Only demethoxycurcumin (2) and analogues 5, 8, 9 had endothelium-dependent actions. Sildenafil was highly potent (EC40 = 0.04 µm) and highly endothelium dependent in pulmonary artery but weak on intact aorta (EC40 = 1.8 µm). Activity profiles suggest actions through additional cell pathways for promoting vasorelaxation.
Conclusions
Curcumin analogues are potential leads for developing efficacious and selective PDE5 inhibitors and other pathologies of pulmonary hypertension.
Collapse
Affiliation(s)
- Oraya Kruangtip
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| | - Krongkarn Chootip
- Department of Physiology, Faculty of Medical Sciences, Naresuan University, Phitsanulok, Thailand
| | - Prapapan Temkitthawon
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| | - Kanokwan Changwichit
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| | - Thipphawan Chuprajob
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Chatchawan Changtam
- Division of Physical Sciences, Faculty of Science and Technology, Huachiew Chalermprakiet University, Samutprakarn, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Nantaka Khorana
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| | - C Norman Scholfield
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| | - Kornkanok Ingkaninan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
13
|
Vasodilator compounds derived from plants and their mechanisms of action. Molecules 2013; 18:5814-57. [PMID: 23685938 PMCID: PMC6270466 DOI: 10.3390/molecules18055814] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 04/24/2013] [Accepted: 05/07/2013] [Indexed: 12/31/2022] Open
Abstract
The present paper reviews vasodilator compounds isolated from plants that were reported in the past 22 years (1990 to 2012) and the different mechanisms of action involved in their vasodilator effects. The search for reports was conducted in a comprehensive manner, intending to encompass those metabolites with a vasodilator effect whose mechanism of action involved both vascular endothelium and arterial smooth muscle. The results obtained from our bibliographic search showed that over half of the isolated compounds have a mechanism of action involving the endothelium. Most of these bioactive metabolites cause vasodilation either by activating the nitric oxide/cGMP pathway or by blocking voltage-dependent calcium channels. Moreover, it was found that many compounds induced vasodilation by more than one mechanism. This review confirms that secondary metabolites, which include a significant group of compounds with extensive chemical diversity, are a valuable source of new pharmaceuticals useful for the treatment and prevention of cardiovascular diseases.
Collapse
|
14
|
Freund-Michel V, Guibert C, Dubois M, Courtois A, Marthan R, Savineau JP, Muller B. Reactive oxygen species as therapeutic targets in pulmonary hypertension. Ther Adv Respir Dis 2013; 7:175-200. [PMID: 23328248 DOI: 10.1177/1753465812472940] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH) is characterized by a progressive elevation of pulmonary arterial pressure due to alterations of both pulmonary vascular structure and function. This disease is rare but life-threatening, leading to the development of right heart failure. Current PH treatments, designed to target altered pulmonary vascular reactivity, include vasodilating prostanoids, phosphodiesterase-5 inhibitors and endothelin-1 receptor antagonists. Although managing to slow the progression of the disease, these molecules still do not cure PH. More effective treatments need to be developed, and novel therapeutic strategies, targeting in particular vascular remodelling, are currently under investigation. Reactive oxygen species (ROS) are important physiological messengers in vascular cells. In addition to atherosclerosis and other systemic vascular diseases, emerging evidence also support a role of ROS in PH pathogenesis. ROS production is increased in animal models of PH, associated with NADPH oxidases increased expression, in particular of several Nox enzymes thought to be the major source of ROS in the pulmonary vasculature. These increases have also been observed in vitro and in vivo in humans. Moreover, several studies have shown either the deleterious effect of agents promoting ROS generation on pulmonary vasculature or, conversely, the beneficial effect of antioxidant agents in animal models of PH. In these studies, ROS production has been directly linked to pulmonary vascular remodelling, endothelial dysfunction, altered vasoconstrictive responses, inflammation and modifications of the extracellular matrix, all important features of PH pathophysiology. Altogether, these findings indicate that ROS are interesting therapeutic targets in PH. Blockade of ROS-dependent signalling pathways, or disruption of sources of ROS in the pulmonary vasculature, targeting in particular Nox enzymes, represent promising new therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Véronique Freund-Michel
- Laboratoire de Pharmacologie-INSERM U1045, UFR des Sciences Pharmaceutiques, Université Bordeaux Segalen, Case 83, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
15
|
Han BH, Vellimana AK, Zhou ML, Milner E, Zipfel GJ. Phosphodiesterase 5 inhibition attenuates cerebral vasospasm and improves functional recovery after experimental subarachnoid hemorrhage. Neurosurgery 2012; 70:178-86; discussion 186-7. [PMID: 21796010 DOI: 10.1227/neu.0b013e31822ec2b0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cerebral vasospasm is an independent predictor of poor outcome after subarachnoid hemorrhage (SAH). The nitric oxide-cyclic guanosine monophosphate (NO-cGMP) vasodilatory pathway is strongly implicated in its pathophysiology. Preliminary studies suggest that phosphodiesterase 5 (PDE5), an enzyme that degrades cGMP, may play a role because the PDE5 inhibitor sildenafil was found to reduce vasospasm after SAH. However, several questions that are critical when considering translational studies remain unanswered. OBJECTIVE To elucidate the mechanism of action of sildenafil against vasospasm and to assess whether sildenafil attenuates SAH-induced neuronal cell death, improves functional outcome after SAH, or causes significant physiological side effects when administered at therapeutically relevant doses. METHODS SAH was induced via endovascular perforation in male C57BL6 mice. Beginning 2 hours later, mice received sildenafil citrate (0.7, 2 or 5 mg/kg orally twice daily) or vehicle. Neurological outcome was assessed daily. Vasospasm was determined on post-SAH day 3. Brain PDE5 expression and activity, cGMP content, neuronal cell death, arterial blood pressure, and intracranial pressure were examined. RESULTS We found that PDE5 activity (but not expression) is increased after SAH, leading to decreased cGMP levels. Sildenafil attenuates this increase in PDE5 activity and restores cGMP levels after SAH. Post-SAH initiation of sildenafil was found to decrease vasospasm and neuronal cell death and markedly improve neurological outcome without causing significant physiological side effects. CONCLUSION Sildenafil, a US Food and Drug Administration-approved drug with a proven track record of safety in humans, is a promising new therapy for vasospasm and neurological deficits after SAH.
Collapse
Affiliation(s)
- Byung Hee Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
16
|
Gebska MA, Stevenson BK, Hemnes AR, Bivalacqua TJ, Haile A, Hesketh GG, Murray CI, Zaiman AL, Halushka MK, Krongkaew N, Strong TD, Cooke CA, El-Haddad H, Tuder RM, Berkowitz DE, Champion HC. Phosphodiesterase-5A (PDE5A) is localized to the endothelial caveolae and modulates NOS3 activity. Cardiovasc Res 2011; 90:353-63. [PMID: 21421555 DOI: 10.1093/cvr/cvq410] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS It has been well demonstrated that phosphodiesterase-5A (PDE5A) is expressed in smooth muscle cells and plays an important role in regulation of vascular tone. The role of endothelial PDE5A, however, has not been yet characterized. The present study was undertaken to determine the presence, localization, and potential physiologic significance of PDE5A within vascular endothelial cells. METHODS AND RESULTS We demonstrate primary location of human, mouse, and bovine endothelial PDE5A at or near caveolae. We found that the spatial localization of PDE5A at the level of caveolin-rich lipid rafts allows for a feedback loop between endothelial PDE5A and nitric oxide synthase (NOS3). Treatment of human endothelium with PDE5A inhibitors resulted in a significant increase in NOS3 activity, whereas overexpression of PDE5A using an adenoviral vector, both in vivo and in cell culture, resulted in decreased NOS3 activity and endothelium-dependent vasodilation. The molecular mechanism responsible for these interactions is primarily regulated by cGMP-dependent second messenger. PDE5A overexpression also resulted in a significant decrease in protein kinase 1 (PKG1) activity. Overexpression of PKG1 rapidly activated NOS3, whereas silencing of the PKG1 gene with siRNA inhibited both NOS3 phosphorylation (S1179) and activity, indicating a novel role for PKG1 in direct regulation of NOS3. CONCLUSION Our data collectively suggest another target for PDE5A inhibition in endothelial dysfunction and provide another physiologic significance for PDE5A in the modulation of endothelial-dependent flow-mediated vasodilation. Using both in vitro and in vivo models, as well as human data, we show that inhibition of endothelial PDE5A improves endothelial function.
Collapse
Affiliation(s)
- Milena A Gebska
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yin J, Kukucka M, Hoffmann J, Sterner-Kock A, Burhenne J, Haefeli WE, Kuppe H, Kuebler WM. Sildenafil preserves lung endothelial function and prevents pulmonary vascular remodeling in a rat model of diastolic heart failure. Circ Heart Fail 2011; 4:198-206. [PMID: 21216837 DOI: 10.1161/circheartfailure.110.957050] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pulmonary hypertension as a frequent complication of left heart disease (PH-LHD) is characterized by lung endothelial dysfunction and vascular remodeling. Although PH-LHD contributes to morbidity and mortality in heart failure, established therapies for PH-LHD are lacking. We tested the effect of chronic sildenafil treatment in an experimental model of PH-LHD. METHODS AND RESULTS In Sprague-Dawley rats, PH-LHD was induced by supracoronary aortic banding. Oral sildenafil treatment (60 mg/kg daily) was initiated after 7 days, and lung endothelial function (n=5), vascular remodeling, and right ventricular function (n=11 each) were analyzed 9 weeks after banding. As compared with sham-operated controls, aortic banding induced pulmonary hypertension and lung endothelial dysfunction evident as lack of endothelial nitric oxide production and endothelium-dependent vasodilation. These changes were associated with an increased pulmonary vascular resistance, medial thickening, and biventricular cardiac hypertrophy. Sildenafil treatment largely attenuated these pathological changes and was not associated with detectable adverse effects pertinent to lung vascular barrier function, edema formation, or systemic hemodynamics. CONCLUSIONS Our data identify sildenafil as a promising therapy for PH-LHD. In light of its documented protective effects at the myocardial level in heart failure, sildenafil presents a particularly attractive strategy in that it simultaneously targets cardiac remodeling and secondary PH-LHD.
Collapse
Affiliation(s)
- Jun Yin
- Keenan Research Centre at the Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Pulmonary hemodynamic response to acute combination and monotherapy with sildenafil and brain natriuretic peptide in rats with monocrotaline-induced pulmonary hypertension. Am J Med Sci 2010; 339:55-9. [PMID: 19996941 DOI: 10.1097/maj.0b013e3181c078d7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The lungs of patients with pulmonary arterial hypertension (PAH) exhibit decreased bioavailability of nitric oxide and downstream signaling through cyclic guanosine monophosphate (cGMP). Therapies that enhance cGMP-mediated vasodilation have shown efficacy in treating PAH. We tested the hypothesis that combination therapy with sildenafil, a cGMP phosphodiesterase type 5 inhibitor, and brain natriuretic peptide (BNP), a receptor-mediated guanosine cyclase stimulator, synergistically attenuates monocrotaline-induced PAH in rats compared with either monotherapy. METHODS Adult male Sprague-Dawley rats were subcutaneously injected with monocrotaline (n = 41, 50 mg/kg). After approximately 4 weeks, the rats were infused intravenously with vehicle solution, sildenafil (42 and 85 microg/kg/min), or BNP (50 and 100 ng/kg/min), alone and in varied combination. The primary endpoint was the relative change in right ventricular systolic pressure (RVSP) and mean arterial systemic pressure (MAP). Secondary endpoints included heart rate and dP/dt. RESULTS Vehicle infusions did not alter hemodynamic variables. Sildenafil85 (85 microg/kg/min) alone decreased RVSP (-16.6 +/- 5.6%) and decreased MAP (-4.0 +/- 4.7%). BNP50 (50 ng/kg/min) and BNP100 (100 ng/kg/min) decreased RVSP (-23.3 +/- 5.7% and -27.1 +/- 2.9%, respectively) and MAP (-6.4 +/- 5.8% and -14.3 +/- 4.1%, respectively). Combination therapy with sildenafil42 and BNP50 decreased RVSP (-20.7 +/- 5.6%) and showed a lessened systemic effect (MAP = -11.6 +/- 5.9%). Combination therapy with sildenafil85 and BNP100 decreased RVSP (-27.6 +/- 3.2%, P = NS) and showed increased systemic effect (MAP = -20.7 +/- 3.1%, P < 0.05) in comparison with sildenafil85. CONCLUSIONS This study suggests that intravenous administration of both sildenafil and BNP monotherapy produces significant improvement in RVSP, making them potentially viable options for the treatment of PAH, whereas combination therapy produces no additional improvement in pulmonary hemodynamics.
Collapse
|
19
|
Lu W, Ran P, Zhang D, Peng G, Li B, Zhong N, Wang J. Sildenafil inhibits chronically hypoxic upregulation of canonical transient receptor potential expression in rat pulmonary arterial smooth muscle. Am J Physiol Cell Physiol 2009; 298:C114-23. [PMID: 19889962 DOI: 10.1152/ajpcell.00629.2008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In pulmonary arterial smooth muscle cells (PASMCs), Ca2+ influx through store-operated Ca2+ channels thought to be composed of canonical transient receptor potential (TRPC) proteins is an important determinant of intracellular free calcium concentration ([Ca2+](i)) and pulmonary vascular tone. Sildenafil, a type V phosphodiesterase inhibitor that increases cellular cGMP, is recently identified as a promising agent for treatment of pulmonary hypertension. We previously demonstrated that chronic hypoxia elevated basal [Ca2+](i) in PASMCs due in large part to enhanced store-operated Ca2+ entry (SOCE); moreover, ex vivo exposure to prolonged hypoxia (4% O2 for 60 h) upregulated TRPC1 and TRPC6 expression in PASMCs. We examined the effect of sildenafil on basal [Ca2+](i), SOCE, and the expression of TRPC in PASMCs under prolonged hypoxia exposure. We also examined the effect of sildenafil on TRPC1 and TRPC6 expression in pulmonary arterial smooth muscle (PA) from rats that developed chronically hypoxic pulmonary hypertension (CHPH). Compared with vehicle control, treatment with sildenafil (300 nM) inhibited prolonged hypoxia induced increases of 1) basal [Ca2+](i), 2) SOCE, and 3) mRNA and protein expression of TRPC in PASMCs. Moreover, sildenafil (50 mg . kg(-1) . day(-1)) inhibited mRNA and protein expression of TRPC1 and TRPC6 in PA from chronically hypoxic (10% O2 for 21 days) rats, which was associated with decreased right ventricular pressure and right ventricular hypertrophy. Furthermore, we found, in PASMCs exposed to prolonged hypoxia, that knockdown of TRPC1 or TRPC6 by their specific small interference RNA attenuated the hypoxic increases of SOCE and basal [Ca2+]i, suggesting a cause and effect link between increases of TRPC1 and TRPC6 expression and the hypoxic increases of SOCE and basal [Ca2+]i. These results suggest that sildenafil may alter basal [Ca2+](i) in PASMCs by decreasing SOCE through downregulation of TRPC1 and TRPC6 expression, thereby contributing to decreased vascular tone of pulmonary arteries during the development of CHPH.
Collapse
Affiliation(s)
- Wenju Lu
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical Univ., 151 Yanjiang Rd., Guangzhou, Guangdong, 510120 People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
20
|
Tahseldar-Roumieh R, Keravis T, Maarouf S, Justiniano H, Sabra R, Lugnier C. PDEs1-5 activity and expression in tissues of cirrhotic rats reveal a role for aortic PDE3 in NO desensitization. Int J Exp Pathol 2009; 90:605-14. [PMID: 19758418 DOI: 10.1111/j.1365-2613.2009.00678.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Liver cirrhosis is associated with increased nitric oxide (NO) production in the vasculature. We have previously demonstrated that aorta from rats with liver cirrhosis have a reduced relaxant response to NO donors that is corrected by DMPPO, a PDE5-specific inhibitor. Vasodilator responses to DMPPO itself were also reduced in rings from cirrhotic rats. These results supported previous suggestions that upregulation of PDE5 in liver cirrhosis might contribute to renal sodium retention, and consequently modulate vascular reactivity in the context of increased NO production (Tahseldar-Roumieh et al. in Am. J. Physiol. Heart Circ. Physiol. 290, H481-H488, 2006). Here, we investigated the possible alteration in activity and expression of cyclic nucleotide phosphodiesterase PDE1-PDE5 in kidney and vascular tissues in rats 4 weeks after bile duct ligation. The kidney of rats with cirrhosis had increased activity of PDE1 and PDE4 but not PDE5, and increased expression of PDE1A. Unexpectedly and interestingly, there was no change in cirrhotic aorta PDE5, but an increase in PDE3 and PDE4 activity associated with increased expression of PDE3A and PDE3B. Cilostamide, a specific PDE3 inhibitor, corrected the decreased response to an NO donor in isolated aorta from cirrhotic rats, suggesting that the difference in response to NO donors was due to differences in PDE3-induced hydrolysis of cGMP or to cGMP-induced inhibition of PDE3, rather than to differences in PDE5 contribution. In conclusion, these changes in PDE isozymes could greatly contribute to NO desensitization and to the regulation of vascular and renal function in liver cirrhosis.
Collapse
Affiliation(s)
- Rima Tahseldar-Roumieh
- Biophotonique et Pharmacologie, CNRS UMR 7213, Université de Strasbourg, 74 route du Rhin, Illkirch, France
| | | | | | | | | | | |
Collapse
|
21
|
Gonçalves RL, Lugnier C, Keravis T, Lopes MJ, Fantini FA, Schmitt M, Cortes SF, Lemos VS. The flavonoid dioclein is a selective inhibitor of cyclic nucleotide phosphodiesterase type 1 (PDE1) and a cGMP-dependent protein kinase (PKG) vasorelaxant in human vascular tissue. Eur J Pharmacol 2009; 620:78-83. [PMID: 19686719 DOI: 10.1016/j.ejphar.2009.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 07/19/2009] [Accepted: 08/04/2009] [Indexed: 11/30/2022]
Abstract
The inhibitory effect of the flavonoid dioclein was assessed on purified vascular cyclic nucleotide phosphodiesterase isoforms (EC 3.1.4.17, PDE1-5) in comparison with 8-methoxymethyl-isobutylmethylxanthine (8-MM-IBMX) and vinpocetine which are currently used as PDE1 inhibitors. The mechanism underlying the vasorelaxant effect of dioclein was investigated in human saphenous vein. Dioclein inhibited PDE1 more selectively than vinpocetine and 8-MM-IBMX, with IC(50) values of 2.47+/-0.26 and 1.44+/-0.35 microM, respectively in basal- and calmodulin-activated states. Dioclein behaved as a competitive inhibitor for cGMP hydrolysis by PDE1 in basal- and calmodulin-activated states (K(i)=0.62+/-0.14 and 0.55+/-0.07 microM, respectively), indicating this inhibitory effect to be independent of calmodulin interactions. In addition, dioclein induced a concentration-dependent relaxation of human saphenous vein which was independent on the presence of functional endothelium (EC(50) values of 7.3+/-3.1 and 11+/-2.7 microM, respectively with and without endothelium). 8-MM-IBMX relaxed human saphenous vein with an EC(50)=31+/-16 microM, whereas vinpocetine did not cause any vasorelaxation at concentrations up to 100 microM. Rp-8-pCPT-cGMPS, which inhibits cGMP-dependent protein kinase (PKG), blocked the vasodilator effect of dioclein, whereas H-89, which is a cAMP-dependent protein kinase (PKA) inhibitor, had a minor inhibitory effect. Our data show that dioclein is a potent calmodulin-independent selective inhibitor of PDE1 and that inhibition of PDE1 is involved in the PKG-mediated vasorelaxant effect of dioclein in human saphenous vein. Furthermore, dioclein may represent a new archetype to develop more specific PDE1 inhibitors.
Collapse
Affiliation(s)
- Roberta L Gonçalves
- Department of Physiology and Biophysics, ICB, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Bender G, Gosset J, Florian J, Tan K, Field M, Marshall S, DeJongh J, Bies R, Danhof M. Population pharmacokinetic model of the pregabalin-sildenafil interaction in rats: application of simulation to preclinical PK-PD study design. Pharm Res 2009; 26:2259-69. [PMID: 19669867 PMCID: PMC2737110 DOI: 10.1007/s11095-009-9942-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 07/08/2009] [Indexed: 10/30/2022]
Abstract
PURPOSE Preliminary evidence has suggested a synergistic interaction between pregabalin and sildenafil for the treatment of neuropathic pain. The focus of this study was to determine the influence of sildenafil on the pharmacokinetics (PK) of pregabalin with the objective of informing the design of a quantitative pharmacodynamic (PD) study. METHODS The pharmacokinetics were determined in rats following 2-hr intravenous infusions of pregabalin at doses of 4 mg/kg/hr and 10 mg/kg/hr with and without a sildenafil bolus (2.2 mg) and steady state infusion (12 mg/kg/hr for 6 h). This PK model was utilized in a preclinical trial simulation with the aim of selecting the optimal sampling strategy to characterize the PK-PD profile in a future study. Eight logistically feasible PK sampling strategies were simulated in NONMEM and examined through trial simulation techniques. RESULTS A two-compartment population PK model best described pregabalin pharmacokinetics. Significant model covariates included either a binary effect of sildenafil administration (30.2% decrease in clearance) or a concentration-dependent effect due to sildenafil's active metabolite. CONCLUSIONS Analysis of simulations indicated that three post-PD samples had the best cost/benefit ratio by providing a significant increase in the precision (and minor improvement in bias) of both PK and PD parameters compared with no PK sampling.
Collapse
Affiliation(s)
- Gregor Bender
- Leiden-Amsterdam Center for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
|
24
|
Schäfer S, Ellinghaus P, Janssen W, Kramer F, Lustig K, Milting H, Kast R, Klein M. Chronic inhibition of phosphodiesterase 5 does not prevent pressure-overload-induced right-ventricular remodelling. Cardiovasc Res 2009; 82:30-9. [PMID: 19131365 DOI: 10.1093/cvr/cvp002] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Inhibition of phosphodiesterase 5 (PDE5) decreases pulmonary pressure and improves symptoms in patients with pulmonary arterial hypertension. It is unclear however, whether inhibition of PDE5 can prevent myocardial remodelling during right-ventricular pressure overload. METHODS AND RESULTS Right-ventricular pressure overload was produced in male rats in a pulmonary hypertension model (monocrotaline 60 mg/kg s.c.) or by surgical pulmonary artery banding. PDE5 inhibition using oral sildenafil (50 mg/kg/day in drinking water) or placebo was initiated 14 days after monocrotaline treatment and continued for 14 days until final examination. In the pulmonary artery banding groups, rats were treated with sildenafil (50 mg/kg/day) or placebo for 21 days following surgical pulmonary artery banding. At the final experiments, right-ventricular haemodynamics were measured and remodelling was analysed using histological, biochemical, and gene expression markers. Both monocrotaline and pulmonary artery banding increased right-ventricular systolic pressure to approximately 80 mmHg. In parallel, both interventions induced markers of hypertrophy (upregulation of natriuretic peptides, increase in myocyte diameter) and fibrosis (upregulation of collagen types 1A2 and 3A1) as well as mRNA expression of the tissue inhibitor of matrix metalloproteases 1 and osteopontin in the right ventricle. In monocrotaline model, sildenafil decreased pulmonary pressure, reduced right-ventricular hypertrophy, and prevented fibrosis marker gene upregulation. After pulmonary artery banding, in contrast, sildenafil increased markers of myocardial remodelling and right-ventricular myocyte diameter. CONCLUSION Sildenafil prevents myocardial remodelling in pulmonary hypertension through an indirect action via right-ventricular unloading.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Blood Pressure/drug effects
- Cyclic GMP/blood
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Disease Models, Animal
- Fibrillar Collagens/metabolism
- Fibrosis
- Hemodynamics/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/enzymology
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/prevention & control
- Male
- Monocrotaline
- Myocardium/enzymology
- Myocardium/pathology
- Natriuretic Peptides/metabolism
- Osteopontin/metabolism
- Phosphodiesterase 5 Inhibitors
- Phosphodiesterase Inhibitors/administration & dosage
- Phosphodiesterase Inhibitors/pharmacokinetics
- Phosphodiesterase Inhibitors/pharmacology
- Piperazines/administration & dosage
- Piperazines/pharmacokinetics
- Piperazines/pharmacology
- Pulmonary Artery/surgery
- Purines/administration & dosage
- Purines/pharmacokinetics
- Purines/pharmacology
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Rats, Wistar
- Sildenafil Citrate
- Stroke Volume/drug effects
- Sulfones/administration & dosage
- Sulfones/pharmacokinetics
- Sulfones/pharmacology
- Time Factors
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Ventricular Pressure/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Stefan Schäfer
- Cardiology Research, Bayer Schering Pharma, Aprather Weg 18, 42096 Wuppertal, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Caffeine inhibits InsP3 responses and capacitative calcium entry in canine pulmonary arterial smooth muscle cells. Vascul Pharmacol 2008; 50:89-97. [PMID: 19084078 DOI: 10.1016/j.vph.2008.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 10/11/2008] [Accepted: 11/09/2008] [Indexed: 11/24/2022]
Abstract
Caffeine is a well described and characterized ryanodine receptor (RyR) activator. Previous evidence from independent research studies also indicate caffeine inhibits InsP3 receptor functionality, which is important to activation of capacitative Ca2+ entry (CCE) in some cell types. In addition, RyR activation elicits excitatory-coupled Ca2+ entry (ECCE) in skeletal muscle myotubes. Recent studies by our group show that canine pulmonary arterial smooth muscle cells (PASMCs) have functional InsP3 receptors as well as RyRs, and that CCE is dependent on InsP3 receptor activity. The potential for caffeine to activate ECCE as well as inhibit InsP3 receptor function and CCE was examined using fura-2 fluorescent imaging in canine PASMCs. The data show caffeine causes transient as well as sustained cytosolic Ca2+ increases, though this is not due to CCE or ECCE activity as evidenced by a lack of an increase in Mn2+ quench of fura-2. The experiments also show caffeine reversibly inhibits 5-HT elicited-InsP3 mediated Ca2+ responses with an IC50 of 6.87x10(-4) M and 10 mM caffeine fully inhibits CCE. These studies provide the first evidence that caffeine is an inhibitor of InsP3 generated Ca2+ signals and CCE in PASMCs.
Collapse
|
26
|
Fujikawa H, Kanno T, Nagata T, Nishizaki T. The phosphodiesterase III inhibitor olprinone inhibits hippocampal glutamate release via a cGMP/PKG pathway. Neurosci Lett 2008; 448:208-11. [PMID: 18983890 DOI: 10.1016/j.neulet.2008.10.079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 10/16/2008] [Accepted: 10/21/2008] [Indexed: 11/19/2022]
Abstract
Olprinone, an inhibitor of cyclic nucleotide phosphodiesterase III, inhibited an increase in intracellular Ca(2+) concentrations for acutely dissociated rat hippocampal pyramidal neurons induced by extracellular high K(+) (35 mM) depolarization. Olprinone (100 microM) significantly reduced spontaneous glutamate release from rat hippocampal slices. Furthermore, olprinone significantly decreased the rate of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor-mediated miniature excitatory postsynaptic currents (AMPA-mEPSCs) monitored from CA1 pyramidal neurons of rat hippocampal slices, and the effect was blocked by KT5823, an inhibitor of protein kinase G (PKG), but not by H-89, an inhibitor of protein kinase A (PKA). In the PKA assay using PC-12 cells, olprinone did not activate PKA. Taken together, the results of the present study show that olprinone attenuates intracellular Ca(2+) rise through voltage-sensitive Ca(2+) channels and inhibits presynaptic glutamate release via a cGMP/PKG pathway.
Collapse
Affiliation(s)
- Hirokazu Fujikawa
- Division of Bioinformation, Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan
| | | | | | | |
Collapse
|
27
|
Morin C, Guibert C, Sirois M, Echave V, Gomes MM, Rousseau E. Effects of omega-hydroxylase product on distal human pulmonary arteries. Am J Physiol Heart Circ Physiol 2008; 294:H1435-43. [PMID: 18203846 DOI: 10.1152/ajpheart.01115.2007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to provide a mechanistic insight into how 20-hydroxyeicosatetraenoic acid (20-HETE) relaxes distal human pulmonary arteries (HPAs). This compound is produced by omega-hydroxylase from free arachidonic acid. Tension measurements, performed on either fresh or 1 day-cultured pulmonary arteries, revealed that the contractile responses to 1 microM 5-hydroxytryptamine were largely relaxed by 20-HETE in a concentration-dependent manner (0.01-10 microM). Iberiotoxin pretreatments (10 nM) partially decreased 20-HETE-induced relaxations. However, 10 microM indomethacin and 3 microM SC-560 pretreatments significantly reduced the relaxations to 20-HETE in these tissues. The relaxing responses induced by the eicosanoid were likely related to a reduced Ca2+ sensitivity of the myofilaments since free Ca2+ concentration ([Ca2+])-response curves performed on beta-escin-permeabilized cultured explants were shifted toward higher [Ca2+]. 20-HETE also abolished the tonic responses induced by phorbol-ester-dibutyrate (a PKC-sensitizing agent). Western blot analyses, using two specific primary antibodies against the PKC-potentiated inhibitory protein CPI-17 and its PKC-dependent phosphorylated isoform pCPI-17, confirmed that 20-HETE interferes with this intracellular process. We also investigated the effect of 20-HETE on the activation of Rho-kinase pathway-induced Ca2+ sensitivity. The data demonstrated that 20-HETE decreased U-46619-induced Ca2+ sensitivity on arteries. Hence, this observation was correlated with an increased staining of p116(Rip), a RhoA-binding protein. Together, these results strongly suggest that the 20-hydroxyarachidonic acid derivative is a potent modulator of tone in HPAs in vitro.
Collapse
Affiliation(s)
- Caroline Morin
- Le Bilarium, Department of Physiology and Biophysics, Université de Sherbrooke, 3001 12th Ave. N, Sherbrooke, J1H 5N4, QC, Canada
| | | | | | | | | | | |
Collapse
|
28
|
Salom JB, Castelló-Ruiz M, Burguete MC, Guzmán C, Jover-Mengual T, Torregrosa G, Jover R, Lizasoain I, Alborch E. Role of K+ and Ca2+ fluxes in the cerebroarterial vasoactive effects of sildenafil. Eur J Pharmacol 2007; 581:138-47. [PMID: 18155692 DOI: 10.1016/j.ejphar.2007.11.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 11/05/2007] [Accepted: 11/15/2007] [Indexed: 11/28/2022]
Abstract
The aim of this study was to assess the role of K(+) and Ca(2+) fluxes in the cerebroarterial vasoactive effects of the phosphodiesterase-5 inhibitor sildenafil. We used isolated rabbit basilar arteries to assess the effects of extracellular K(+) raising on sildenafil-induced vasodilatation, and studied the pharmacological interaction of sildenafil with selective modulators of membrane K(+) and Ca(2+) channels. Expression of Kv1 subunits of K(+) channels was assessed at messenger and protein levels. Parallel experiments were carried out with zaprinast for comparison. Sildenafil (10 nM-0.1 mM) induced concentration-dependent relaxation of endothelin-1 (10 nM)-precontracted arteries, which was partially inhibited by depolarization with KCl (50 mM), 3 mM tetraethylammonium (non-selective K(+) channel blocker) or 1 mM aminopyridine (inhibitor of K(v) channels), but not by 1 microM glibenclamide (inhibitor of K(ATP) channels) or 50 nM iberiotoxin (inhibitor of K(Ca) channels). Arterial smooth muscle expressed messengers for Kv1.2, Kv1.3, Kv1.4, Kv1.5 and Kv1.6, and proteins of Kv1.1, Kv1.2 and Kv1.4. CaCl(2) (10 microM- 10 mM) induced concentration-dependent contraction in Ca(2+)-free, depolarizing (50 mM KCl) medium. Sildenafil (0.1-100 microM) produced reversible concentration-dependent inhibition of the response to CaCl(2), which was completely abolished by the highest sildenafil concentration. By contrast, only 100 microM zaprinast inhibited the response to CaCl(2). The L-type Ca(2+) channel activator Bay K 8644 (0.1 nM-1 microM) induced concentration-dependent potentiation of the response to CaCl(2) inhibited by 100 microM sildenafil. Moreover, Bay K 8644 (0.1 nM-1 microM) induced concentration-dependent contraction in slightly depolarizing (15 mM) medium, which was inhibited to the same extent and in a concentration-dependent way by sildenafil (0.1-100 microM) and zaprinast (1 or 100 microM). These results show that sildenafil relaxes the rabbit basilar artery by increasing K(+) efflux through K(v) channels, which in turn may affect Ca(2+) signalling. Expression of Kv1 subunits involved in this pharmacological effect occurs at the messenger and, in some cases, at the protein level. In addition to this phosphodiesterase-5-related effect, sildenafil and zaprinast inhibit cerebroarterial vasoconstriction at least in part by directly blocking L-type Ca(2+) channels, although a decrease in the sensitivity of the contractile apparatus to Ca(2+) can not be discarded.
Collapse
Affiliation(s)
- Juan B Salom
- Centro de Investigación, Hospital Universitario La Fe, Valencia, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ballester C, Sarriá B, García-Granero E, Morcillo EJ, Lledó S, Cortijo J. Relaxation of the isolated human internal anal sphincter by sildenafil. Br J Surg 2007; 94:894-902. [PMID: 17335124 DOI: 10.1002/bjs.5724] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Hypertonicity of the internal anal sphincter (IAS) appears to be involved in the pathogenesis of anal fissure. The relaxant effects of sildenafil, a selective phosphodiesterase 5 (PDE5) inhibitor, on isolated human IAS were investigated. METHODS The efficacy (maximal effect, E(max)) and potency (-log IC(50), where IC(50) is half-maximal inhibitory concentration) of the PDE5 inhibitors, sildenafil and zaprinast, and of nitric oxide donors, sodium nitroprusside and glyceryl trinitrate, as relaxants of histamine (0.1 mmol/l)-induced tone were examined in IAS strips under isometric contraction. The presence of PDE5 isoenzymes and changes in intracellular calcium and cyclic nucleotide levels in IAS muscle were tested by real-time reverse transcriptase-polymerase chain reaction, epifluorescence microscopy and enzyme immunoassay respectively. RESULTS Sildenafil produced a concentration-related inhibition of the mean(s.e.m.) histamine-induced tone (E(max) 83(2) per cent, - log IC(50) 7.04(0.05); n = 12). Zaprinast produced relaxation to similar degree, but with lower potency. Nitric oxide donors also relaxed IAS. Sildenafil (1 micromol/l) produced a 1.8-fold increase in guanosine 3',5'-cyclic monophosphate content, with no change in adenosine 3',5'-cyclic monophosphate levels. Sildenafil markedly depressed the peak intracellular calcium increase evoked by histamine. PDE5A1, PDE5A2 and PDE5A3 transcripts were expressed in IAS muscle. CONCLUSION Sildenafil relaxes the augmented tone of human IAS in vitro. These results support the potential use of this PDE5 inhibitor in the treatment of chronic anal fissure.
Collapse
Affiliation(s)
- C Ballester
- Department of Surgery, University of Valencia, Valencia, Spain
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
The therapy of pulmonary hypertension has evolved rapidly in the last 10 years from the use of non-selective vasodilators to drugs that specifically target pulmonary vasodilation, endothelial function, and vascular remodeling. Sildenafil is a phosphodiesterase type 5 inhibitor that has an expanding role in the treatment of pulmonary hypertension. Case series and small studies, as well as the first large randomized controlled trial, have demonstrated the safety and efficacy of sildenafil in improving mean pulmonary artery pressure, pulmonary vascular resistance, cardiac index, and exercise tolerance in pulmonary arterial hypertension. It may be useful in adults, children, and neonates after cardiac surgery, with left heart failure, in fibrotic pulmonary disease, high altitude exposure, and thromboembolic disease, and in combination with other therapies for pulmonary hypertension, such as inhaled iloprost. The oral formulation and favorable adverse effect profile make sildenafil an attractive alternative in the treatment of selected patients with pulmonary hypertension.
Collapse
Affiliation(s)
- Christopher F Barnett
- Critical Care Medicine Department, Clinical Center, National Institutes of HealthBethesda, MD, USA
- Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of HealthBethesda, MD, USA
| | - Roberto F Machado
- Critical Care Medicine Department, Clinical Center, National Institutes of HealthBethesda, MD, USA
- Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of HealthBethesda, MD, USA
| |
Collapse
|
31
|
Orhan I, Onur R, Taşdemir C, Ayar A, Kadioğlu A. Sildenafil Citrate Inhibits Agonist Induced Contractions in Isolated Rat Seminal Vesicles. J Urol 2006; 175:2350-3. [PMID: 16697872 DOI: 10.1016/s0022-5347(06)00280-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Indexed: 11/25/2022]
Abstract
PURPOSE Sildenafil is reported to regulate smooth muscle contractility through nitric oxide-cyclic guanosine monophosphate, not only in the corpus cavernosum. Its possible effects on seminal vesicle contractility might be of importance with respect to premature ejaculation. We investigated the effects of sildenafil citrate (Pfizer, New York, New York) on agonist induced isometric contractions of the rat seminal vesicle in vitro. MATERIALS AND METHODS Seminal vesicles isolated from adult male Wistar rats were suspended in an organ bath and contracted by NE (10 microM), ACh (10 microM) or KCl (60 mM) (Sigma, Deisenhofen, Germany). The effects of sildenafil citrate (100 to 300 microM) were evaluated in terms of mean contraction amplitude, the area under force-time curves and isometric contractility indexes. RESULTS Sildenafil citrate (300 microM) significantly inhibited the mean amplitude +/- SEM of contractile responses induced by NE (1,061 +/- 153 vs 271 +/- 65 mg, p <0.0001), ACh (475 +/- 51 vs 68 +/- 17 mg, p <0.0001) and KCl (546 +/- 71 vs 59 +/- 18 mg, p <0.0002). It also caused dose dependent concomitant decreases in the area under force-time curves. Additionally, pretreatment with sildenafil citrate markedly prevented the contractile response to NE, ACh and KCl. CONCLUSIONS Our results indicate that sildenafil citrate inhibits the contractions of isolated rat seminal vesicle that are induced by NE, ACh or KCl. Future studies may support an in vivo effect of sildenafil for delaying or inhibiting seminal vesicle emission, thereby, promoting improvement in patients with premature ejaculation.
Collapse
Affiliation(s)
- Irfan Orhan
- Department of Urology, Firat University, Elazig, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
32
|
Prieto D, Rivera L, Recio P, Rubio JLR, Hernández M, García-Sacristán A. Role of nitric oxide in the relaxation elicited by sildenafil in penile resistance arteries. J Urol 2006; 175:1164-70. [PMID: 16469645 DOI: 10.1016/s0022-5347(05)00320-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Indexed: 12/26/2022]
Abstract
PURPOSE We investigated the role of the vascular endothelium and the L-arginine/nitric oxide pathway in the vasorelaxant effect of the phosphodiesterase type 5 inhibitor sildenafil in penile resistance arteries. MATERIALS AND METHODS Second or third order branches of the horse deep intracavernous penile artery were mounted in microvascular myographs. The vasodilator effects of sildenafil and the NO donor SNAP (S-nitrosoacetyl-D,L-penicillamine) were evaluated in the absence and presence of the endothelium and inhibitors of the NO/cGMP (cyclic guanosine 3',5'-monophosphate) pathway. RESULTS In phenylephrine precontracted, endothelium intact arteries sildenafil elicited potent relaxations that were markedly decreased by the blockade of soluble guanylate cyclase with ODQ (1H-[1,2,4]oxadiazolo[4-3a]quinoxalin-1-one). Endothelium removal and the inhibition of NO synthase with N(G)-nitro-L-arginine (L-NOARG) caused pronounced inhibition of sildenafil elicited relaxations but not of SNAP induced responses. Combined treatment with the cyclooxygenase blocker indomethacin and L-NOARG caused significantly greater inhibition of sildenafil relaxations than that produced by L-NOARG alone. Inhibitors of the cGMP (PKG) and the cAMP (cyclic adenosine 3',5'-monophosphate) dependent protein kinases Rp-8-Br-PET-cGMPS (beta-phenyl-1, N2-etheno-8-bromoguanosine-3',5'-cyclic monophosphorothioate, Rp-isomer) and Rp-8-CPT-cAMPS (Rp-8-CPT-cAMPS (8-(4-chlorophenylthio) adenosine-3',5'-cyclic monophosphorothioate, Rp-isomer), respectively, inhibited the sildenafil concentration-relaxation curves. The relaxant responses of SNAP were markedly decreased by PKG inhibitor and to a lesser extent by cAMP dependent protein kinase inhibitor. CONCLUSIONS The current results demonstrate a potent relaxant effect of sildenafil in penile resistance arteries due in part to cGMP accumulation and to the enhanced effects of basal released, endothelial derived NO acting through PKG activation. Cross-activation of the cAMP signaling pathway by sildenafil is also suggested.
Collapse
Affiliation(s)
- Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid and Servicio de Urología, Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
33
|
Wu BN, Chen IC, Lin RJ, Chiu CC, An LM, Chen IJ. Aortic smooth muscle relaxants KMUP-3 and KMUP-4, two nitrophenylpiperazine derivatives of xanthine, display cGMP-enhancing activity: roles of endothelium, phosphodiesterase, and K+ channel. J Cardiovasc Pharmacol 2006; 46:600-8. [PMID: 16220066 DOI: 10.1097/01.fjc.0000180900.32489.f9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The cellular mechanisms of vasorelaxant effects of newly synthesized KMUP-3 and KMUP-4 were investigated in rat aortic smooth muscle (RASM). KMUP-3 (7-[2-[4-(4-nitrobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) and KMUP-4 (7-[2-[4-(2-nitrobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) elicited concentration-dependent relaxation of endothelium-intact and denuded RASM precontracted with phenylephrine. Relaxant responses were also produced by the PDE inhibitors theophylline, milrinone, rolipram, and zaprinast (1 nM-100 microM). The relaxant responses of KMUP-3 and KMUP-4 were reduced by endothelium removal and by the presence of the NOS inhibitor L-NAME (100 microM), the sGC inhibitor ODQ (1 microM), the adenylyl cyclase (AC) inhibitor SQ 22536 (100 microM), and the prostaglandin inhibitor indomethacin (10 microM). Additionally, the vasorelaxations of both agents were also attenuated by pretreatment with the nonselective K+ channel blocker TEA (10 mM), the KATP channel blocker glibenclamide (1 microM), the voltage-dependent K+ (KV) channel blocker 4-AP (100 microM), and Ca(2+)-dependent K+ (KCa) channel blockers apamin (1 microM) and charybdotoxin (ChTX, 0.1 microM). In addition, elevated extracellular K+ (80 mM) interferes with KMUP-3- and KMUP-4-induced vasorelaxations. Preincubation with both agents (1 microM) significantly enhanced the dilator responses of isoproterenol and SNP. KMUP-3 and KMUP-4 inhibited PDE activities and increased cAMP and cGMP levels in primary culture of RASM that were inhibited by SQ 22536 and ODQ, respectively. In cultured HUVECs, KMUP-3 and KMUP-4 (0.1 microM), more potent than YC-1, significantly increased the expression of eNOS protein. In summary, KMUP-3 and KMUP-4 induce aortic relaxations through both endothelium-dependent and -independent mechanisms. Mechanisms of vasorelaxation induced by both compounds involve multiple processes, such as accumulation of cyclic nucleotides partly as a result of PDE inhibition, K-channel activation, and indomethacin-sensitive endothelium function.
Collapse
MESH Headings
- Animals
- Aorta
- Cell Line
- Cyclic AMP/metabolism
- Cyclic GMP/metabolism
- Endothelium, Vascular/physiology
- In Vitro Techniques
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Phosphoric Diester Hydrolases/metabolism
- Piperazines/chemistry
- Piperazines/pharmacology
- Piperidines/chemistry
- Piperidines/pharmacology
- Potassium Channels/metabolism
- Rats
- Rats, Wistar
- Vasoconstrictor Agents/pharmacology
- Vasodilator Agents/chemistry
- Vasodilator Agents/pharmacology
- Xanthines/chemistry
- Xanthines/pharmacology
Collapse
Affiliation(s)
- Bin-Nan Wu
- Department and Graduate Institute of Pharmacology, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung 807, Taiwan
| | | | | | | | | | | |
Collapse
|
34
|
Salom JB, Burguete MC, Pérez-Asensio FJ, Castelló-Ruiz M, Torregrosa G, Alborch E. Relaxant effect of sildenafil in the rabbit basilar artery. Vascul Pharmacol 2005; 44:10-6. [PMID: 16325476 DOI: 10.1016/j.vph.2005.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Revised: 05/01/2005] [Accepted: 07/01/2005] [Indexed: 10/25/2022]
Abstract
We hypothesized that sildenafil, inhibitor of phosphodiesterase-5 (PDE-5), interacts with the nitric oxide (NO)-cGMP pathway in the cerebral arteries and shows vasoactive effects. To prove it in the isolated rabbit basilar artery, we compared the effects of sildenafil with other PDE-5 inhibitors, assessed the endothelial dependence of the vasoactive responses, and used modulators of the cGMP and cAMP signaling processes. Sildenafil (10 nM-0.1 mM) induced concentration-dependent relaxations of endothelin-1 (10 nM)-precontracted basilar artery, which were partially inhibited both in endothelium-denuded arteries and in arteries precontracted by depolarization with KCl (50 mM). Endothelin-1 (1 pM-30 nM) induced concentration-dependent contractions that were inhibited by sildenafil (0.1-100 microM). Zaprinast (10 nM-0.1 mM) and MBCQ (1 nM-0.1 mM), PDE-5 inhibitors, induced concentration-dependent relaxations with lower and higher potency than sildenafil, respectively. Sildenafil-induced relaxation was inhibited in arteries preincubated with the NO synthase inhibitor L-NAME (0.1 mM) or the soluble guanylyl cyclase inhibitor ODQ (10 microM). Preincubation with sildenafil (0.1 microM) enhanced the relaxations induced by acetylcholine (0.1 nM-0.1 mM) and the NO donor sodium nitroprusside (0.1 nM-0.1 mM), but not those induced by the cell-permeable cGMP analogue 8-Br-cGMP (1 nM-0.1 mM) and the adenylyl cyclase activator forskolin (0.1 nM-10 microM). These results show that sildenafil has vasoactive effects in isolated cerebral arteries. By enhancing the NO-cGMP signaling pathway in the cerebrovascular wall, sildenafil induces vasodilation, prevents vasoconstriction, and potentiates the effect of other NO-dependent vasodilators.
Collapse
Affiliation(s)
- Juan B Salom
- Centro de Investigación, Hospital Universitario La Fe, Ave. Campanar 21, 46009 Valencia, Spain.
| | | | | | | | | | | |
Collapse
|
35
|
Teixeira CE, Priviero FBM, Webb RC. Differential effects of the phosphodiesterase type 5 inhibitors sildenafil, vardenafil, and tadalafil in rat aorta. J Pharmacol Exp Ther 2005; 316:654-61. [PMID: 16204472 DOI: 10.1124/jpet.105.092544] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Presumably, the vasorelaxant properties of phosphodiesterase type 5 (PDE5) inhibitors are similar in isolated blood vessels. We aimed to explore the mechanisms underlying the vasorelaxation induced by the selective PDE5 inhibitors sildenafil, vardenafil, and tadalafil in the rat aorta. Aortic rings were mounted in 5-ml organ baths, and concentration-response curves for PDE5 inhibitors (0.0001-10 microM) were constructed in phenylephrine (PE)-precontracted endothelium-intact and -denuded rings. Cyclic nucleotides were measured using enzyme immunoassay kits. Sildenafil, vardenafil, and tadalafil concentration dependently relaxed aortic rings and increased cGMP, but not cAMP, concentrations. Endothelium denudation caused marked rightward shifts in the curves to sildenafil (45-fold), tadalafil (21-fold), and vardenafil (251-fold). Maximal responses to sildenafil and tadalafil were substantially reduced (38 +/- 1% and 53 +/- 2%, respectively), whereas that evoked by vardenafil was not affected. Likewise, inhibition of NO synthase (N(omega)-nitro-L-arginine methyl ester, 100 microM), guanylyl cyclase (1H-[1,2,4]oxadiazolo [4,3,-a]quinoxalin-1-one, 10 microM), or scavenging of NO ([carboxy-PTIO (2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide), 100 microM]) caused similar attenuation of the vasorelaxations evoked by PDE5 inhibitors. Sildenafil, tadalafil, and vardenafil significantly potentiated relaxations mediated by glyceryl trinitrate (0.0001-3 microM; 8-13-fold) and atrial natriuretic peptide (0.1-100 nM; 2-3-fold). Contractions evoked by CaCl(2) (0.01-5 mM) in PE-treated rings were significantly reduced (26 +/- 4%) by vardenafil, but not sildenafil or tadalafil, whereas phorbol 12,13-dibutyrate-induced contractions were not affected. Ouabain, cyclopiazonic acid, and calyculin A failed to affect vasorelaxations induced by the PDE5 inhibitors. These results suggest that vardenafil, but not sildenafil or tadalafil, affects Ca(2+) handling in the rat aorta in addition to increasing cGMP levels through inhibition of PDE5 to cause relaxation.
Collapse
Affiliation(s)
- Cleber E Teixeira
- Department of Physiology, Medical College of Georgia, Augusta, 30912-3000, USA.
| | | | | |
Collapse
|
36
|
Kruuse C, Khurana TS, Rybalkin SD, Birk S, Engel U, Edvinsson L, Olesen J. Phosphodiesterase 5 and effects of sildenafil on cerebral arteries of man and guinea pig. Eur J Pharmacol 2005; 521:105-14. [PMID: 16182282 DOI: 10.1016/j.ejphar.2005.07.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 07/12/2005] [Accepted: 07/19/2005] [Indexed: 11/25/2022]
Abstract
Sildenafil (Viagra), a selective inhibitor of phosphodiesterase 5 (PDE5), induces headache and migraine. Although previously supposed to be a "vascular" headache, no significant cerebral artery dilatation was found in vivo. Thus, we hypothesised that PDE5 may not be present or that sildenafil is less effective on the cGMP hydrolysis in cerebral arteries, and that sildenafil may not be an effective dilator of cerebral arteries under baseline conditions. We evaluated the presence of PDE5 mRNA and protein in human arteries. Furthermore, the effects of two selective PDE5 inhibitors, sildenafil and UK-114,542, and a PDE1 inhibitor UK-90,234 on cGMP hydrolysis were investigated in human and guinea pig cerebral arteries. The vasoactive responses of the compounds were evaluated in guinea pig basilar arteries in vitro, with concomitant measurements of cAMP and cGMP. PDE5 was found in human middle cerebral arteries. Sildenafil and UK-114,542 inhibited cGMP hydrolysis concentration-dependently in both species. In guinea pig arteries, sildenafil induced an endothelium-dependent vasodilatation only at concentrations above 10 nM, which was augmented by sodium nitroprusside and attenuated by reduction of cGMP, but was cGMP independent at high concentrations. UK-114,542 was more and UK-90,234 was less potent than sildenafil. In conclusion, PDE5 is present in human and guinea pig cerebral arteries, and is inhibited by sildenafil at micromolar levels. Sildenafil in vitro is a poor dilator of guinea pig cerebral arteries unless a nitric oxide donor is co-administered, corresponding to the previous findings in vivo.
Collapse
MESH Headings
- 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors
- 3',5'-Cyclic-GMP Phosphodiesterases/genetics
- 3',5'-Cyclic-GMP Phosphodiesterases/metabolism
- Aged
- Aged, 80 and over
- Animals
- Basilar Artery/drug effects
- Basilar Artery/enzymology
- Basilar Artery/physiology
- Blotting, Western
- Cerebral Arteries/drug effects
- Cerebral Arteries/enzymology
- Cerebral Arteries/physiology
- Cyclic AMP/metabolism
- Cyclic GMP/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 5
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Female
- Guanylate Cyclase/antagonists & inhibitors
- Guinea Pigs
- Humans
- Hydrolysis/drug effects
- In Vitro Techniques
- Infant
- Male
- Morpholines/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Nitroprusside/pharmacology
- Oxadiazoles/pharmacology
- Piperazines/pharmacology
- Purines
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Pyrimidinones/pharmacology
- Quinoxalines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sildenafil Citrate
- Sulfones
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Christina Kruuse
- Department of Clinical Experimental Research, Glostrup University Hospital, University of Copenhagen, Glostrup, Denmark.
| | | | | | | | | | | | | |
Collapse
|
37
|
Sharabi FM, Daabees TT, El-Metwally MA, Senbel AM. Effect of sildenafil on the isolated rat aortic rings. Fundam Clin Pharmacol 2005; 19:449-56. [PMID: 16011732 DOI: 10.1111/j.1472-8206.2005.00345.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Sildenafil, a highly selective inhibitor of PDE 5, is effective in the treatment of erectile dysfunction. Penile erection involves relaxation of smooth muscle of corpus cavernosum and its associated arterioles. The objective of this study was to investigate the effect of sildenafil on nitric oxide/cyclic guanosine monophosphate (NO/cGMP)-dependent relaxation of rat aortic rings. The contribution of sildenafil to the vasorelaxation of rat aortic rings was also investigated. Sildenafil produced significant potentiation of acetylcholine (ACh, 2 x 10(-6) m)-induced relaxation at concentration > or =1 x 10(-8) m. Addition of sildenafil (1 x 10(-7) m) to aortic rings failed to alter the effect of N(G)-nitro-L-arginine (l-NNA, 3 x 10(-5) m) or methylene blue (MB, 3 x 10(-5) m) on ACh response. Similarly, sildenafil (1 x 10(-7) m) augmented significantly the vasorelaxation induced by sodium nitroprusside over the range of 1 x 10(-9)-1 x 10(-8) m. When added to phenylephrine (3 x 10(-6) m)-precontracted rat aortic rings, sildenafil (1 x 10(-9)-1 x 10(-4) m) induced concentration-dependent relaxation reaching a maximum of 96.48 +/- 1.44%. These relaxations were not significantly attenuated by previous incubation with L-NNA (3 x 10(-5) m) or MB (3 x 10(-5) m). Denudation did not significantly affect the vasorelaxant effect of sildenafil. Sildenafil may act in the rat aortic rings through the amplification of NO/cGMP pathway. It may augment both basal endothelial NO function and exogenous NO-dependent vasodilatation. However, sildenafil may act by a mechanism independent of NO/cGMP pathway and this mechanism contributes to its smooth muscle relaxant effect.
Collapse
Affiliation(s)
- F M Sharabi
- Department of Pharmacology, Faculty of Pharmacy, University of Alexandria, El-Khartoom Square, Alexandria, Egypt.
| | | | | | | |
Collapse
|
38
|
Lugnier C. Cyclic nucleotide phosphodiesterase (PDE) superfamily: a new target for the development of specific therapeutic agents. Pharmacol Ther 2005; 109:366-98. [PMID: 16102838 DOI: 10.1016/j.pharmthera.2005.07.003] [Citation(s) in RCA: 643] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Accepted: 07/12/2005] [Indexed: 01/08/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs), which are ubiquitously distributed in mammalian tissues, play a major role in cell signaling by hydrolyzing cAMP and cGMP. Due to their diversity, which allows specific distribution at cellular and subcellular levels, PDEs can selectively regulate various cellular functions. Their critical role in intracellular signaling has recently designated them as new therapeutic targets for inflammation. The PDE superfamily represents 11 gene families (PDE1 to PDE11). Each family encompasses 1 to 4 distinct genes, to give more than 20 genes in mammals encoding the more than 50 different PDE proteins probably produced in mammalian cells. Although PDE1 to PDE6 were the first well-characterized isoforms because of their predominance in various tissues and cells, their specific contribution to tissue function and their regulation in pathophysiology remain open research fields. This concerns particularly the newly discovered families, PDE7 to PDE11, for which roles are not yet established. In many pathologies, such as inflammation, neurodegeneration, and cancer, alterations in intracellular signaling related to PDE deregulation may explain the difficulties observed in the prevention and treatment of these pathologies. By inhibiting specifically the up-regulated PDE isozyme(s) with newly synthesized potent and isozyme-selective PDE inhibitors, it may be potentially possible to restore normal intracellular signaling selectively, providing therapy with reduced adverse effects.
Collapse
Affiliation(s)
- Claire Lugnier
- CNRS UMR, 7034, Pharmacologie et Physicochimie des Interactions Moléculaires et Cellulaires, Faculté de Pharmacie, Université Louis Pasteur de Strasbourg, 74 route du Rhin, BP 60024, 67401 Illkirch, France.
| |
Collapse
|
39
|
Wharton J, Strange JW, Møller GMO, Growcott EJ, Ren X, Franklyn AP, Phillips SC, Wilkins MR. Antiproliferative effects of phosphodiesterase type 5 inhibition in human pulmonary artery cells. Am J Respir Crit Care Med 2005; 172:105-13. [PMID: 15817798 DOI: 10.1164/rccm.200411-1587oc] [Citation(s) in RCA: 239] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Phosphodiesterase Type 5 (PDE5) inhibition represents a novel strategy for the treatment of pulmonary hypertension. OBJECTIVES Our aim was to establish the distribution of PDE5 in the pulmonary vasculature and effects of PDE5 inhibition on pulmonary artery smooth muscle cells (PASMCs). METHODS AND MEASUREMENTS PDE5 expression was examined by immunohistochemistry and Western blotting, in both normal and hypertensive lung tissues. DNA synthesis, proliferation, PDE activity, and apoptosis were measured in distal human PASMCs treated with soluble guanylyl cyclase activators (nitric oxide donors and BAY41-2272) and sildenafil. MAIN RESULTS Cells containing PDE5 and alpha-smooth muscle actin occurred throughout the pulmonary vasculature, including obstructive intimal lesions. Three molecular forms of PDE5 were identified and protein expression was greater in hypertensive than control lung tissue. Most cyclic guanosine monophosphate hydrolysis (about 80%) in cultured cells was attributed to PDE5. Sildenafil induced a greater elevation of intracellular cyclic guanosine monophosphate levels compared with nitric oxide donors and BAY41-2272 (about 10-fold versus about 2-fold) and cotreatment had a synergistic effect, increasing cyclic nucleotide levels up to 50-fold. Dual stimulation of soluble guanylyl cyclase and inhibition of PDE5 activities also had significant downstream effects, increasing phosphorylation of vasodilator-stimulated phosphoprotein, reducing DNA synthesis and cell proliferation, and stimulating apoptosis, and these effects were mimicked by cyclic guanosine monophosphate analogs. CONCLUSIONS Phosphodiesterase Type 5 is the main factor regulating cyclic guanosine monophosphate hydrolysis and downstream signaling in human PASMCs. The antiproliferative effects of this signaling pathway may be significant in the chronic treatment of pulmonary hypertension with PDE5 inhibitors such as sildenafil.
Collapse
Affiliation(s)
- John Wharton
- Section on Experimental Medicine and Toxicology, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 ONN, UK.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Moreno L, Losada B, Cogolludo AL, Lodi F, Lugnier C, Villamor E, Moro M, Tamargo J, Pérez-Vizcaíno F. Postnatal maturation of phosphodiesterase 5 (PDE5) in piglet pulmonary arteries: activity, expression, effects of PDE5 inhibitors, and role of the nitric oxide/cyclic GMP pathway. Pediatr Res 2004; 56:563-70. [PMID: 15295092 DOI: 10.1203/01.pdr.0000139412.58594.d0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
After birth and during the first days of extrauterine life, pulmonary arterial pressure is progressively reduced to reach the adult values. We hypothesized that changes in PDE5 activity might be involved in the pulmonary postnatal maturation of the nitric oxide (NO)/cGMP pathway. The PDE5 inhibitor sildenafil produced vasorelaxant responses in isolated pulmonary arteries. These effects were similar in newborn (3-18 h) and 2-wk-old piglets, unchanged by endothelium removal, and markedly inhibited by the soluble guanylyl cyclase inhibitor ODQ. The peak of the transient vasorelaxant response to NO gas increased with postnatal age but was unaffected by PDE inhibition. However, the duration of the response to NO was significantly increased. The vasorelaxant response to sodium nitroprusside was potentiated by sildenafil in both age groups. The PDE5 inhibitors dipyridamole and zaprinast, produced qualitatively similar effects but with lower potency. Both total and PDE5-dependent cGMP hydrolytic activity and PDE5 protein expression increased with postnatal age. All these results suggest that PDE5 is a key regulator of NO-induced vasodilation in the postnatal pulmonary arteries. PDE5 inhibition is able to produce pulmonary vasodilation even in the absence of a functional endothelium and potentiates the vasorelaxant response to exogenous NO and nitroprusside. However, PDE5 is not responsible for the maturational increase of NO bioactivity during the first days of extrauterine life.
Collapse
Affiliation(s)
- Laura Moreno
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pauvert O, Bonnet S, Rousseau E, Marthan R, Savineau JP. Sildenafil alters calcium signaling and vascular tone in pulmonary arteries from chronically hypoxic rats. Am J Physiol Lung Cell Mol Physiol 2004; 287:L577-83. [PMID: 15155272 DOI: 10.1152/ajplung.00449.2003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sildenafil, a potent type 5 nucleotide-dependent phosphodiesterase (PDE) inhibitor, has been recently proposed as a therapeutic tool to treat or prevent pulmonary artery hypertension (PAHT). We thus studied the effect of sildenafil on both the calcium signaling of isolated pulmonary artery smooth muscle cells (PASMCs) and the reactivity of pulmonary artery (PA) obtained from chronic hypoxia (CH)-induced pulmonary hypertensive rats compared with control (normoxic) rats. CH rats were maintained in an hypobaric chamber (50.5 kPa) for 3 wk leading to full development of PAHT. Intracellular calcium concentration ([Ca2+]i) was measured in PASMCs loaded with the calcium fluorophore indo 1. Unlike in control rats, sildenafil (10–100 nM) decreased the resting [Ca2+]ivalue in PASMCs obtained from CH rats. In PASMCs from both control and CH rats, sildenafil concentration dependently inhibited the [Ca2+]iresponse induced by G-coupled membrane receptor agonists such as angiotensin II and phenylephrine but had no effect on the amplitude of the [Ca2+]iresponse induced by caffeine. Sildenafil (0.1 nM–1 μM) concentration dependently reduced basal PA tone that is present in CH rats and relaxed PA rings precontracted with phenylephrine in both control and CH rats. These data show that sildenafil is a potent pulmonary artery relaxant in CH rats and that it normalizes CH-induced increases in resting [Ca2+]iand basal tone. Consequently, pharmacological inhibition of sildenafil-sensitive PDE5 downregulates the Ca2+signaling pathway involved in this model of pulmonary hypertension.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Caffeine/pharmacology
- Calcium/metabolism
- Calcium Signaling/drug effects
- Chronic Disease
- Disease Models, Animal
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/physiopathology
- Hypoxia/drug therapy
- Hypoxia/physiopathology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Phenylephrine/pharmacology
- Phosphodiesterase Inhibitors/pharmacology
- Piperazines/pharmacology
- Pulmonary Artery/cytology
- Pulmonary Artery/drug effects
- Pulmonary Artery/physiology
- Purines
- Rats
- Rats, Wistar
- Sildenafil Citrate
- Sulfones
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Olivier Pauvert
- Laboratoire de Physiologie Cellulaire Respiratoire, INSERM (E 356 and IFR 4 Université Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux, France
| | | | | | | | | |
Collapse
|