101
|
Santi D, Granata ARM, Guidi A, Pignatti E, Trenti T, Roli L, Bozic R, Zaza S, Pacchioni C, Romano S, Nofer JR, Rochira V, Carani C, Simoni M. Six months of daily treatment with vardenafil improves parameters of endothelial inflammation and of hypogonadism in male patients with type 2 diabetes and erectile dysfunction: a randomized, double-blind, prospective trial. Eur J Endocrinol 2016; 174:513-22. [PMID: 26792933 DOI: 10.1530/eje-15-1100] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/19/2016] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is associated with endothelial dysfunction, characterized by a reduction of nitric oxide (NO)-mediated relaxation. Phosphodiesterase type 5 inhibitors (PDE5i) improve NO levels. The aim of the study was to investigate whether long-term, chronic treatment with the PDE5i vardenafil improves systemic endothelial function in diabetic men. DESIGN A prospective, investigator-initiated, randomized, placebo-controlled, double-blind, clinical trial was conducted. METHODS In total, 54 male patients affected by T2DM, diagnosed within the last 5 years, and erectile dysfunction were enrolled, regardless of testosterone levels. In all, 26 and 28 patients were assigned to verum and placebo groups respectively. The study consisted of an enrollment phase, a treatment phase (24 weeks) (vardenafil/placebo 10 mg twice in a day) and a follow-up phase (24 weeks). Parameters evaluated were as follows: International Index of Erectile Function 15 (IIEF-15), flow-mediated dilation (FMD), serum interleukin 6 (IL6), endothelin 1 (ET-1), gonadotropins and testosterone (measured by liquid chromatography/tandem mass spectrometry). RESULTS IIEF-15 erectile function improved during the treatment (P<0.001). At the end of the treatment both FMD (P=0.040) and IL6 (P=0.019) significantly improved. FMD correlated with serum testosterone levels (R(2)=0.299; P<0.001). Testosterone increased significantly under vardenafil treatment and returned in the eugonadal range only in hypogonadal men (n=13), without changes in gonadotropins. Chronic vardenafil treatment did not result in relevant side effects. CONCLUSION This is the first double-blind, placebo-controlled clinical trial designed to evaluate the effects of chronic treatment of vardenafil on endothelial health-related parameters and sexual hormones in patients affected by a chronic disease. Chronically administered vardenafil is effective and improves endothelial parameters in T2DM patient. Moreover, chronic vardenafil therapy improves hypogonadism in diabetic, hypogonadal men.
Collapse
Affiliation(s)
- Daniele Santi
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Antonio R M Granata
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Alessandro Guidi
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Elisa Pignatti
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Tommaso Trenti
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Laura Roli
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Roberto Bozic
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Stefano Zaza
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Chiara Pacchioni
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Stefania Romano
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Jerzy Roch Nofer
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Vincenzo Rochira
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Cesare Carani
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| | - Manuela Simoni
- Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany Unit of Endocrinology & MetabolismDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, ItalyDepartment of MedicineEndocrinology, Metabolism and Geriatrics, Azienda USL of Modena, Modena, ItalyCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Modena, ItalyDepartment of Laboratory Medicine and Pathological AnatomyAzienda USL of Modena, Modena, ItalyPerkinElmer20126 Milan, ItalySHIMADZU ItaliaMilan, ItalyDiabetic ServiceAzienda USL of Modena, Modena, ItalyCenter for Laboratory MedicineUniversity Hospital Münster, Münster, Germany
| |
Collapse
|
102
|
The hypertension of Cushing's syndrome: controversies in the pathophysiology and focus on cardiovascular complications. J Hypertens 2016; 33:44-60. [PMID: 25415766 PMCID: PMC4342316 DOI: 10.1097/hjh.0000000000000415] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cushing's syndrome is associated with increased mortality, mainly due to cardiovascular complications, which are sustained by the common development of systemic arterial hypertension and metabolic syndrome, which partially persist after the disease remission. Cardiovascular diseases and hypertension associated with endogenous hypercortisolism reveal underexplored peculiarities. The use of exogenous corticosteroids also impacts on hypertension and cardiovascular system, especially after prolonged treatment. The mechanisms involved in the development of hypertension differ, whether glucocorticoid excess is acute or chronic, and the source endogenous or exogenous, introducing inconsistencies among published studies. The pleiotropic effects of glucocorticoids and the overlap of the several regulatory mechanisms controlling blood pressure suggest that a rigorous comparison of in-vivo and in-vitro studies is necessary to draw reliable conclusions. This review, developed during the first ‘Altogether to Beat Cushing's syndrome’ workshop held in Capri in 2012, evaluates the most important peculiarities of hypertension associated with CS, with a particular focus on its pathophysiology. A critical appraisal of most significant animal and human studies is compared with a systematic review of the few available clinical trials. A special attention is dedicated to the description of the clinical features and cardiovascular damage secondary to glucocorticoid excess. On the basis of the consensus reached during the workshop, a pathophysiology-oriented therapeutic algorithm has been developed and it could serve as a first attempt to rationalize the treatment of hypertension in Cushing's syndrome.
Collapse
|
103
|
Pofi R, Gianfrilli D, Badagliacca R, Di Dato C, Venneri MA, Giannetta E. Everything you ever wanted to know about phosphodiesterase 5 inhibitors and the heart (but never dared ask): How do they work? J Endocrinol Invest 2016; 39:131-42. [PMID: 26142740 DOI: 10.1007/s40618-015-0339-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 06/11/2015] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Phosphodiesterase 5 inhibitors (PDE5i) were developed while investigating novel treatments for coronary artery disease, but their andrological side effects shifted their indication toward the management of erectile dysfunction. Although PDE5i are now also indicated for pulmonary arterial hypertension and there are mounting preclinical and clinical evidences about their potentially beneficial cardiac effects, their use remains controversial and the involved mechanisms remain unclear. MATERIALS AND METHODS This review aimed to analyze the effects of PDE5i administration in various animal and humans models of cardiovascular diseases. RESULTS Animal studies have shown that PDE5i have protective effects in several models of cardiac disease. In humans, some studies showed that PDE5i improves microvascular and endothelial dysfunction and exerts positive effects in different samples of cardiovascular (CV) impairment. In contrast, other studies found no benefit (and no harm) in heart failure with preserved ejection fraction. The discrepancies in these findings are likely related to the fact that the mechanisms targeted by PDE5i in human disease are still poorly understood and the target population not yet identified. The mechanisms of actions herein reviewed suggest that hypertrophy, microvascular impairment, and inflammation, should be variably present for PDE5i to work. All these conditions frequently coexist in diabetes. A gender responsiveness has also been recently proposed. CONCLUSIONS Continuous PDE5 inhibition may exert cardioprotective effects, improving endothelial function and counteracting cardiac remodeling in some but not all conditions. A better patient selection could help to clarify the controversies on PDE5i use for CV disorders.
Collapse
Affiliation(s)
- R Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - R Badagliacca
- Department of Cardiovascular and Respiratory Science, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - C Di Dato
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - M A Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - E Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| |
Collapse
|
104
|
Corinaldesi C, Di Luigi L, Lenzi A, Crescioli C. Phosphodiesterase type 5 inhibitors: back and forward from cardiac indications. J Endocrinol Invest 2016; 39:143-51. [PMID: 26122487 PMCID: PMC4712255 DOI: 10.1007/s40618-015-0340-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/11/2015] [Indexed: 12/24/2022]
Abstract
PDE5 inhibitors (PDE5i) are widely known as treatment for erectile dysfunction (ED). This favorable action has emerged as a "side effect" from pioneering studies when PDE5i have been originally proposed as treatment for coronary artery disease (CAD). PDE5i showed marginal benefits for CAD treatment; although disappointing for that indication, they improved systemic and pulmonary vasodilation and ameliorated general endothelial function. Therefore, PDE5i have been approved and licensed also for pulmonary artery hypertension (PAH), besides ED. Nowadays, fine-tuned biomolecular mechanisms of PDE5i are well recognized to be beneficial onto myocardial contractility and geometry, to reduce tissue fibrosis, hypertrophy and apoptosis. PDE5i consistently exert benefits on heart failure, infarct, cardiomyopathy. The concept that PDE5i likely blunt Th1-driven inflammatory processes, which shift the homeostatic balance from health to disease, has emerged; PDE5i seem to decrease the release of active biomolecules from cells to tissues interested by inflammation. In this view, following clinical and basic research progresses, PDE5i can be undoubtedly "re-allocated" for cardiac indications and, hopefully, they could be approved as therapeutic tools to treat and prevent heart disease. This review aims to summarize PDE5i different clinical applications, from past to present and future, focusing on their potential power as treatment for cardiac diseases.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cardiovascular Agents/pharmacokinetics
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/immunology
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/physiopathology
- Coronary Artery Disease/drug therapy
- Coronary Artery Disease/immunology
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/physiopathology
- Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Evidence-Based Medicine
- Heart/drug effects
- Heart/physiopathology
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Myocardium/enzymology
- Myocardium/immunology
- Myocardium/metabolism
- Phosphodiesterase 5 Inhibitors/pharmacokinetics
- Phosphodiesterase 5 Inhibitors/pharmacology
- Phosphodiesterase 5 Inhibitors/therapeutic use
- Vasodilator Agents/pharmacokinetics
- Vasodilator Agents/pharmacology
- Vasodilator Agents/therapeutic use
Collapse
Affiliation(s)
- C Corinaldesi
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy
| | - L Di Luigi
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - C Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy.
| |
Collapse
|
105
|
Corona G, Rastrelli G, Burri A, Jannini EA, Maggi M. The safety and efficacy of Avanafil, a new 2nd generation PDE5i: comprehensive review and meta-analysis. Expert Opin Drug Saf 2016; 15:237-47. [DOI: 10.1517/14740338.2016.1130126] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Giovanni Corona
- Endocrinology Unit, Maggiore-Bellaria Hospital, Medical Department, Azienda-Usl Bologna, Bologna, Italy
| | - Giulia Rastrelli
- Sexual Medicine and Andrology Unit, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| | - Andrea Burri
- Department of Psychology, University of Zurich, Zurich, Switzerland
| | - Emmanuele A. Jannini
- Endocrinology, Andrology and Medical Sexology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Mario Maggi
- Sexual Medicine and Andrology Unit, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
106
|
Suematsu Y, Miura SI, Goto M, Matsuo Y, Arimura T, Kuwano T, Imaizumi S, Iwata A, Yahiro E, Saku K. LCZ696, an angiotensin receptor-neprilysin inhibitor, improves cardiac function with the attenuation of fibrosis in heart failure with reduced ejection fraction in streptozotocin-induced diabetic mice. Eur J Heart Fail 2016; 18:386-93. [DOI: 10.1002/ejhf.474] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/06/2015] [Accepted: 11/29/2015] [Indexed: 11/09/2022] Open
Affiliation(s)
- Yasunori Suematsu
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Shin-ichiro Miura
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
- Department of Molecular Cardiovascular Therapeutics; Fukuoka University School of Medicine; Fukuoka Japan
| | - Masaki Goto
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Yoshino Matsuo
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Tadaaki Arimura
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Takashi Kuwano
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Satoshi Imaizumi
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Atsushi Iwata
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Eiji Yahiro
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
| | - Keijiro Saku
- Department of Cardiology; Fukuoka University School of Medicine; Fukuoka Japan
- Department of Molecular Cardiovascular Therapeutics; Fukuoka University School of Medicine; Fukuoka Japan
| |
Collapse
|
107
|
Lim SL, Lam CSP. Breakthrough in heart failure with preserved ejection fraction: are we there yet? Korean J Intern Med 2016; 31:1-14. [PMID: 26767852 PMCID: PMC4712413 DOI: 10.3904/kjim.2016.31.1.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/13/2015] [Indexed: 01/09/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFPEF) is a global health problem of considerable socioeconomic burden. It is projected to worsen with the aging population worldwide. The lack of effective therapies underscores our incomplete understanding of this complex heterogeneous syndrome. A novel paradigm has recently emerged, in which central roles are ascribed to systemic inflammation and generalized endothelial dysfunction in the pathophysiology of HFPEF. In this review, we discuss the role of the endothelium in cardiovascular homeostasis and how deranged endothelial-related signaling pathways contribute to the development of HFPEF. We also review the novel therapies in various stages of research and development that target different components of this signaling pathway.
Collapse
Affiliation(s)
- Shir Lynn Lim
- Department of Cardiology, National University Heart Center, Singapore
| | - Carolyn Su Ping Lam
- Department of Cardiology, National Heart Center Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore
| |
Collapse
|
108
|
Wang G, Zhang Q, Yuan W, Wu J, Li C. Sildenafil Protects against Myocardial Ischemia-Reperfusion Injury Following Cardiac Arrest in a Porcine Model: Possible Role of the Renin-Angiotensin System. Int J Mol Sci 2015; 16:27015-31. [PMID: 26569234 PMCID: PMC4661868 DOI: 10.3390/ijms161126010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 10/13/2015] [Accepted: 11/03/2015] [Indexed: 12/28/2022] Open
Abstract
Sildenafil, a phosphodiesterase-5 inhibitor sold as Viagra, is a cardioprotector against myocardial ischemia/reperfusion (I/R) injury. Our study explored whether sildenafil protects against I/R-induced damage in a porcine cardiac arrest and resuscitation (CAR) model via modulating the renin-angiotensin system. Male pigs were randomly divided to three groups: Sham group, Saline group, and sildenafil (0.5 mg/kg) group. Thirty min after drug infusion, ventricular fibrillation (8 min) and cardiopulmonary resuscitation (up to 30 min) was conducted in these animals. We found that sildenafil ameliorated the reduced cardiac function and improved the 24-h survival rate in this model. Sildenafil partly attenuated the increases of plasma angiotensin II (Ang II) and Ang (1–7) levels after CAR. Sildenafil also decreased apoptosis and Ang II expression in myocardium. The increases of expression of angiotensin-converting-enzyme (ACE), ACE2, Ang II type 1 receptor (AT1R), and the Ang (1–7) receptor Mas in myocardial tissue were enhanced after CAR. Sildenafil suppressed AT1R up-regulation, but had no effect on ACE, ACE2, and Mas expression. Sildenafilfurther boosted the upregulation of endothelial nitric oxide synthase (eNOS), cyclic guanosine monophosphate (cGMP) and inducible nitric oxide synthase(iNOS). Collectively, our results suggest that cardioprotection of sildenafil in CAR model is accompanied by an inhibition of Ang II-AT1R axis activation.
Collapse
Affiliation(s)
- Guoxing Wang
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, 8 Worker's Stadium South Road, Chao-Yang District, Beijing 100020, China.
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China.
| | - Qian Zhang
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, 8 Worker's Stadium South Road, Chao-Yang District, Beijing 100020, China.
| | - Wei Yuan
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, 8 Worker's Stadium South Road, Chao-Yang District, Beijing 100020, China.
| | - Junyuan Wu
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, 8 Worker's Stadium South Road, Chao-Yang District, Beijing 100020, China.
| | - Chunsheng Li
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, 8 Worker's Stadium South Road, Chao-Yang District, Beijing 100020, China.
| |
Collapse
|
109
|
Defeudis G, Gianfrilli D, Di Emidio C, Pofi R, Tuccinardi D, Palermo A, Lenzi A, Pozzilli P. Erectile dysfunction and its management in patients with diabetes mellitus. Rev Endocr Metab Disord 2015; 16:10.1007/s11154-015-9321-4. [PMID: 26497842 DOI: 10.1007/s11154-015-9321-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetes can be described as a syndrome of multiple closely related conditions induced by a chronic state of hyperglycaemia resulting from defective insulin secretion, insulin action or both. Chronic complications associated with diabetes (including neuropathy, vascular disease, nephropathy and retinopathy) are common, and of these, erectile dysfunction (ED) deserves special attention. ED and its correlation with cardiovascular disease require careful evaluation and appropriate treatment. PDE5 inhibitors (PDE5is) are an important tool for the treatment of ED, with new drugs coming onto the market since the late 90s. This review offers an overview of PDE5is and their use in treating ED in diabetes. We underline the differences between different types of PDE5i, focusing on available doses, duration of action, T ½, side effects and selectivity profiles in relation to patients with diabetes. We also discuss the link between diabetes and ED in presence of various associated cofactors (obesity, hypertension and its pharmacological treatments, atherosclerosis, hyperhomocysteinaemia, neuropathy, nephropathy, hypogonadism and depression). Finally a number of past and ongoing clinical trials on the use of PDE5is in patients with diabetes are presented to offer an overview of the appropriate treatment of ED in this condition.
Collapse
Affiliation(s)
- Giuseppe Defeudis
- Department of Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy
| | - Chiara Di Emidio
- Department of Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy
| | - Dario Tuccinardi
- Department of Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, Rome, Italy
| | - Andrea Palermo
- Department of Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy
| | - Paolo Pozzilli
- Department of Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, Rome, Italy.
| |
Collapse
|
110
|
Salloum FN, Chau VQ, Hoke NN, Kukreja RC. Tadalafil prevents acute heart failure with reduced ejection fraction in mice. Cardiovasc Drugs Ther 2015; 28:493-500. [PMID: 25322707 DOI: 10.1007/s10557-014-6559-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE Phosphodiesterase-5 (PDE5) inhibitors were shown to exert powerful protection in various animal models of cardiomyopathy. Tadalafil is a long-acting and highly specific PDE5 inhibitor, which makes it the most attractive in its class for long-term management of patients with heart failure. We studied the effects of tadalafil in attenuating ischemic cardiomyopathy in mice. METHODS AND RESULTS Adult male mice underwent myocardial infarction (MI) by permanent left coronary artery ligation and were treated daily with tadalafil (1 mg/kg; ip) or volume-matched 10% DMSO for 4 weeks. Twenty four hours after coronary ligation, infarct size, measured by TTC staining, was reduced from 70.1 ± 3.1% in DMSO-treated group to 49.3 ± 2.6% with tadalafil (P < 0.05). Similarly, tadalafil treatment yielded a smaller fibrotic area (8.8 ± 2.8% of LV), assessed by Masson's trichrome staining, as compared to DMSO group (21.9 ± 3.9%, P < 0.05). Apoptosis, measured by TUNEL assay, also declined with tadalafil (2.1 ± 0.2%) as compared to DMSO (6.7 ± 0.4%, P < 0.05) at 28 days post MI. Tadalafil also attenuated the increase in cardiac hypertrophy and pulmonary edema following infarction. These parameters reflect diminished left ventricular (LV) adverse remodeling and preserved fractional shortening with tadalafil at 7 and 28 days post infarction. CONCLUSIONS Tadalafil attenuates ischemic cardiomyopathy in mice and preserves LV function.
Collapse
Affiliation(s)
- Fadi N Salloum
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020A, Richmond, VA, 23298-0204, USA,
| | | | | | | |
Collapse
|
111
|
Mandosi E, Giannetta E, Filardi T, Lococo M, Bertolini C, Fallarino M, Gianfrilli D, Venneri MA, Lenti L, Lenzi A, Morano S. Endothelial dysfunction markers as a therapeutic target for Sildenafil treatment and effects on metabolic control in type 2 diabetes. Expert Opin Ther Targets 2015; 19:1617-22. [DOI: 10.1517/14728222.2015.1066337] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
112
|
Venneri MA, Giannetta E, Panio G, De Gaetano R, Gianfrilli D, Pofi R, Masciarelli S, Fazi F, Pellegrini M, Lenzi A, Naro F, Isidori AM. Chronic Inhibition of PDE5 Limits Pro-Inflammatory Monocyte-Macrophage Polarization in Streptozotocin-Induced Diabetic Mice. PLoS One 2015; 10:e0126580. [PMID: 25961566 PMCID: PMC4427327 DOI: 10.1371/journal.pone.0126580] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/06/2015] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is characterized by changes in endothelial cells that alter monocyte recruitment, increase classic (M1-type) tissue macrophage infiltration and lead to self-sustained inflammation. Our and other groups recently showed that chronic inhibition of phosphodiesterase-5 (PDE5i) affects circulating cytokine levels in patients with diabetes; whether PDE5i also affects circulating monocytes and tissue inflammatory cell infiltration remains to be established. Using murine streptozotocin (STZ)-induced diabetes and in human vitro cell-cell adhesion models we show that chronic hyperglycemia induces changes in myeloid and endothelial cells that alter monocyte recruitment and lead to self-sustained inflammation. Continuous PDE5i with sildenafil (SILD) expanded tissue anti-inflammatory TIE2-expressing monocytes (TEMs), which are known to limit inflammation and promote tissue repair. Specifically, SILD: 1) normalizes the frequency of circulating pro-inflammatory monocytes triggered by hyperglycemia (53.7 ± 7.9% of CD11b+Gr-1+ cells in STZ vs. 30.4 ± 8.3% in STZ+SILD and 27.1 ± 1.6% in CTRL, P<0.01); 2) prevents STZ-induced tissue inflammatory infiltration (4-fold increase in F4/80+ macrophages in diabetic vs. control mice) by increasing renal and heart anti-inflammatory TEMs (30.9 ± 3.6% in STZ+SILD vs. 6.9 ± 2.7% in STZ, P <0.01, and 11.6 ± 2.9% in CTRL mice); 3) reduces vascular inflammatory proteins (iNOS, COX2, VCAM-1) promoting tissue protection; 4) lowers monocyte adhesion to human endothelial cells in vitro through the TIE2 receptor. All these changes occurred independently from changes of glycemic status. In summary, we demonstrate that circulating renal and cardiac TEMs are defective in chronic hyperglycemia and that SILD normalizes their levels by facilitating the shift from classic (M1-like) to alternative (M2-like)/TEM macrophage polarization. Restoration of tissue TEMs with PDE5i could represent an additional pharmacological tool to prevent end-organ diabetic complications.
Collapse
Affiliation(s)
- Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- * E-mail: (AMI); (MAV)
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Panio
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rita De Gaetano
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvia Masciarelli
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - Manuela Pellegrini
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - Andrea M. Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- * E-mail: (AMI); (MAV)
| |
Collapse
|
113
|
Goulopoulou S, Hannan JL, Matsumoto T, Ogbi S, Ergul A, Webb RC. Reduced vascular responses to soluble guanylyl cyclase but increased sensitivity to sildenafil in female rats with type 2 diabetes. Am J Physiol Heart Circ Physiol 2015; 309:H297-304. [PMID: 25957216 DOI: 10.1152/ajpheart.00079.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/04/2015] [Indexed: 01/25/2023]
Abstract
Impaired nitric oxide (NO), soluble guanylyl cyclase (sGC), and cyclic guanosine monophosphate (cGMP) signaling (NO-sGC-cGMP) has been implicated in the pathogenesis of diabetic vascular dysfunction. Efforts to directly target this signaling have led to the development of sGC agonists that activate the heme group of sGC (stimulators) or preferentially activate sGC when the heme is oxidized (activators). In this study, we hypothesized that resistance arteries from female rats with spontaneous type 2 diabetes (Goto-Kakizaki rats, GK) would have reduced vasodilatory responses to heme-dependent sGC activation and increased responses to heme-independent sGC activation compared with control rats (Wistar). Endothelium-dependent and -independent relaxation was assessed in isolated segments from mesenteric resistance arteries (MA) mounted in a wire myograph. GK MA had reduced responses to acetylcholine (pEC50: 7.96 ± 0.06 vs. 7.66 ± 0.05, P < 0.05) and sodium nitroprusside (pEC50: 8.34 ± 0.05 vs. 7.77 ± 0.04, P < 0.05). There were no group differences in 8-bromoguanosine cGMP-induced relaxation and protein kinase G1 expression (P > 0.05). GK MA had attenuated responses to BAY 41-2272 (heme-dependent sGC stimulator; pEC50: 7.56 ± 0.05 vs. 6.93 ± 0.06, P < 0.05) and BAY 58-2667 (heme-independent sGC activator; pEC50: 10.82 ± 0.07 vs. 10.27 ± 0.08, P < 0.05) and increased sensitivity to sildenafil [phosphodiesterase 5 (PDE5) inhibitor; pEC50: 7.89 ± 0.14 vs. 8.25 ± 0.13, P < 0.05]. Isolated resistance arteries from female rats of reproductive age that spontaneously develop type 2 diabetes have increased sensitivity to PDE5 inhibition and reduced responsiveness to sGC activators and stimulators.
Collapse
Affiliation(s)
| | - Johanna L Hannan
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Safia Ogbi
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Adviye Ergul
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - R Clinton Webb
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
114
|
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: 3.6] [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
|
115
|
|
116
|
Degen CV, Bishu K, Zakeri R, Ogut O, Redfield MM, Brozovich FV. The emperor's new clothes: PDE5 and the heart. PLoS One 2015; 10:e0118664. [PMID: 25747598 PMCID: PMC4351884 DOI: 10.1371/journal.pone.0118664] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
Phosphodiesterase-5 (PDE5) is highly expressed in the pulmonary vasculature, but its expression in the myocardium is controversial. Cyclic guanosine monophosphate (cGMP) activates protein kinase G (PKG), which has been hypothesized to blunt cardiac hypertrophy and negative remodeling in heart failure. Although PDE5 has been suggested to play a significant role in the breakdown of cGMP in cardiomyocytes and hence PKG regulation in the myocardium, the RELAX trial, which tested effect of PDE5 inhibition on exercise capacity in patients with heart failure with preserved ejection fraction (HFpEF) failed to show a beneficial effect. These results highlight the controversy regarding the role and expression of PDE5 in the healthy and failing heart. This study used one- and two-dimensional electrophoresis and Western blotting to examine PDE5 expression in mouse (before and after trans-aortic constriction), dog (control and HFpEF) as well as human (healthy and failing) heart. We were unable to detect PDE5 in any cardiac tissue lysate, whereas PDE5 was present in the murine and bovine lung samples used as positive controls. These results indicate that if PDE5 is expressed in cardiac tissue, it is present in very low quantities, as PDE5 was not detected in either humans or any model of heart failure examined. Therefore in cardiac muscle, it is unlikely that PDE5 is involved the regulation of cGMP-PKG signaling, and hence PDE5 does not represent a suitable drug target for the treatment of cardiac hypertrophy. These results highlight the importance of rigorous investigation prior to clinical trial design.
Collapse
Affiliation(s)
- Chantal V. Degen
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Kalkidan Bishu
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Rosita Zakeri
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Ozgur Ogut
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Margaret M. Redfield
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Frank V. Brozovich
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
- * E-mail:
| |
Collapse
|
117
|
Santi D, Giannetta E, Isidori AM, Vitale C, Aversa A, Simoni M. Therapy of endocrine disease. Effects of chronic use of phosphodiesterase inhibitors on endothelial markers in type 2 diabetes mellitus: a meta-analysis. Eur J Endocrinol 2015; 172:R103-14. [PMID: 25277671 DOI: 10.1530/eje-14-0700] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Diabetes mellitus (DM) is associated with endothelial dysfunction, reducing nitric oxide-dependent vasodilation, and increasing production of pro-inflammatory factors, leading to an increased risk of long-term cardiovascular disease. As the effects of phosphodiesterase 5 inhibitors (PDE5i) on endothelial function have not been systematically investigated, we conducted a meta-analysis of available randomized clinical trials (RCTs). DESIGN A thorough search of the literature was carried out. Relevant studies were considered according to RCT study design, enrollment of men with type 2 DM, chronic administration of PDE5i, and evaluation of endothelial function through both hemodynamic and endothelial inflammation-related parameters. RESULTS Fifteen studies fulfilled the eligibility criteria but only six RCTs met the inclusion criteria and were analyzed for 476 diabetic men, 239 randomized to Sildenafil, and 237 to placebo respectively. Four RCTs evaluated flow-mediated dilation (FMD), demonstrating a weighted mean increase of 2.19% (95% CI 0.48 to 3.90). This result showed a high heterogeneity (I(2): 98%). Thus, a further sub-group meta-analysis was performed and this analysis confirmed a significant, Sildenafil-related FMD improvement. Sildenafil improved endothelin 1 and high sensitivity C-reactive protein by ∼-0.94 pg/ml and -0.36 mg/l, respectively, not reaching statistical significance (P=0.69 and P=0.22 respectively). Finally, Sildenafil administration significantly reduced serum levels of interleukin 6 (IL6, -0.82 pg/ml; 95% CI -1.58 to -0.07). CONCLUSION This meta-analysis suggests a beneficial effect of chronic PDE5i administration on endothelial function. Chronic Sildenafil administration seems to improve hemodynamic (FMD) and serum pro-inflammatory makers (IL6) in diabetic men. Larger studies are needed to confirm the effects of chronic PDE5i on endothelial function.
Collapse
Affiliation(s)
- Daniele Santi
- Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy
| | - Elisa Giannetta
- Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy
| | - Andrea M Isidori
- Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy
| | - Cristiana Vitale
- Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy
| | - Antonio Aversa
- Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy
| | - Manuela Simoni
- Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy Unit of EndocrinologyDepartment of Biomedical Metabolic and Neural Sciences. University of Modena and Reggio Emilia, ItalyAzienda USL of ModenaItalySapienza University of RomeDepartment of Experimental MedicineDepartment of Medical SciencesIRCSS San Raffaele, Rome, Italy
| |
Collapse
|
118
|
Tham YK, Bernardo BC, Ooi JYY, Weeks KL, McMullen JR. Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets. Arch Toxicol 2015; 89:1401-38. [DOI: 10.1007/s00204-015-1477-x] [Citation(s) in RCA: 371] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 12/18/2022]
|
119
|
Rowther FB, Wei W, Dawson TP, Ashton K, Singh A, Madiesse-Timchou MP, Thomas DGT, Darling JL, Warr T. Cyclic nucleotide phosphodiesterase-1C (PDE1C) drives cell proliferation, migration and invasion in glioblastoma multiforme cells in vitro. Mol Carcinog 2015; 55:268-79. [PMID: 25620587 DOI: 10.1002/mc.22276] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/10/2014] [Accepted: 12/01/2014] [Indexed: 12/17/2022]
Abstract
Cyclic nucleotides (cAMP & cGMP) are critical intracellular second messengers involved in the transduction of a diverse array of stimuli and their catabolism is mediated by phosphodiesterases (PDEs). We previously detected focal genomic amplification of PDE1C in >90 glioblastoma multiforme (GBM) cells suggesting a potential as a novel therapeutic target in these cells. In this report, we show that genomic gain of PDE1C was associated with increased expression in low passage GBM-derived cell cultures. We demonstrate that PDE1C is essential in driving cell proliferation, migration and invasion in GBM cultures since silencing of this gene significantly mitigates these functions. We also define the mechanistic basis of this functional effect through whole genome expression analysis by identifying down-stream gene effectors of PDE1C which are involved in cell cycle and cell adhesion regulation. In addition, we also demonstrate that Vinpocetine, a general PDE1 inhibitor, can also attenuate proliferation with no effect on invasion/migration. Up-regulation of at least one of this gene set (IL8, CXCL2, FOSB, NFE2L3, SUB1, SORBS2, WNT5A, and MMP1) in TCGA GBM cohorts is associated with worse outcome and PDE1C silencing down-regulated their expression, thus also indicating potential to influence patient survival. Therefore we conclude that proliferation, migration, and invasion of GBM cells could also be regulated downstream of PDE1C.
Collapse
Affiliation(s)
- Farjana B Rowther
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| | - Weinbin Wei
- School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | - Timothy P Dawson
- Lancashire Teaching Hospitals, Royal Preston Hospital, Preston, UK
| | - Katherine Ashton
- Lancashire Teaching Hospitals, Royal Preston Hospital, Preston, UK
| | - Anushree Singh
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| | | | - D G T Thomas
- National Hospital for Neurology and Neurosurgery, London
| | - John L Darling
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| | - Tracy Warr
- Brain Tumour Research Centre, University of Wolverhampton, Wolverhampton, UK
| |
Collapse
|
120
|
Papapetropoulos A, Hobbs AJ, Topouzis S. Extending the translational potential of targeting NO/cGMP-regulated pathways in the CVS. Br J Pharmacol 2015; 172:1397-414. [PMID: 25302549 DOI: 10.1111/bph.12980] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 09/08/2014] [Accepted: 10/05/2014] [Indexed: 02/06/2023] Open
Abstract
The discovery of NO as both an endogenous signalling molecule and as a mediator of the cardiovascular effects of organic nitrates was acknowledged in 1998 by the Nobel Prize in Physiology/Medicine. The characterization of its downstream signalling, mediated through stimulation of soluble GC (sGC) and cGMP generation, initiated significant translational interest, but until recently this was almost exclusively embodied by the use of PDE5 inhibitors in erectile dysfunction. Since then, research progress in two areas has contributed to an impressive expansion of the therapeutic targeting of the NO-sGC-cGMP axis: first, an increased understanding of the molecular events operating within this complex pathway and second, a better insight into its dys-regulation and uncoupling in human disease. Already-approved PDE5 inhibitors and novel, first-in-class molecules, which up-regulate the activity of sGC independently of NO and/or of the enzyme's haem prosthetic group, are undergoing clinical evaluation to treat pulmonary hypertension and myocardial failure. These molecules, as well as combinations or second-generation compounds, are also being assessed in additional experimental disease models and in patients in a wide spectrum of novel indications, such as endotoxic shock, diabetic cardiomyopathy and Becker's muscular dystrophy. There is well-founded optimism that the modulation of the NO-sGC-cGMP pathway will sustain the development of an increasing number of successful clinical candidates for years to come.
Collapse
|
121
|
Di Luigi L, Botrè F, Sabatini S, Sansone M, Mazzarino M, Guidetti L, Baldari C, Lenzi A, Caporossi D, Romanelli F, Sgrò P. Acute effects of physical exercise and phosphodiesterase's type 5 inhibition on serum 11β-hydroxysteroid dehydrogenases related glucocorticoids metabolites: a pilot study. Endocrine 2014; 47:952-8. [PMID: 24532078 DOI: 10.1007/s12020-014-0185-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 01/25/2014] [Indexed: 12/11/2022]
Abstract
Endogenous glucocorticoids (GC) rapidly increase after acute exercise, and the phosphodiesterase's type 5 inhibitor (PDE5i) tadalafil influences this physiological adaptation. No data exist on acute effects of both acute exercise and PDE5i administration on 11β-hydroxysteroid dehydrogenases (11β-HSDs)-related GC metabolites. We aimed to investigate the rapid effects of exercise on serum GC metabolites, with and without tadalafil administration. A double blind crossover study was performed in eleven healthy male volunteers. After the volunteers randomly received a short-term administration of placebo or tadalafil (20 mg/die for 2 days), a maximal exercise test to exhaustion on cycle ergometer was performed. Then, after a 2-week washout period, the volunteers were crossed over. Blood samples were collected before starting exercise and at 5 and 30 min of recovery (+5-Rec, +30-Rec). Serum ACTH, corticosterone (Cn), cortisol (F), cortisone (E), tetrahydrocortisol (THF), tetrahydrocortisone (THE), cortols, cortolones and respective ratios were evaluated. Pre-Ex THF was higher after tadalafil. Exercise increased ACTH, Cn, F, E, THE, cortols and cortolones after both placebo and tadalafil, and THF after placebo. The F/E ratio increased at +5-Rec and decreased at +30-Rec after placebo. Compared to placebo, after tadalafil lower ACTH, F and Cn, higher THF/F and THE/E, and not E (at +5-Rec) and F/E modifications were observed. Acute exercise rapidly influences serum GC metabolites concentrations. Tadalafil influences both GC adaptation and 11β-HSDs activity during acute exercise. Additional researches on the effects of both exercise and PDE5i on tissue-specific 11β-HSDs activity at rest and during physiological adaptation are warranted.
Collapse
Affiliation(s)
- Luigi Di Luigi
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro de Bosis, 15, 00135, Rome, Italy,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Das A, Durrant D, Salloum FN, Xi L, Kukreja RC. PDE5 inhibitors as therapeutics for heart disease, diabetes and cancer. Pharmacol Ther 2014; 147:12-21. [PMID: 25444755 DOI: 10.1016/j.pharmthera.2014.10.003] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
Abstract
The phosphodiesterase 5 (PDE5) inhibitors, including sildenafil (Viagra™), vardenafil (Levitra™), and tadalafil (Cialis™) have been developed for treatment of erectile dysfunction. Moreover, sildenafil and tadalafil are used for the management of pulmonary arterial hypertension in patients. Since our first report showing the cardioprotective effect of sildenafil in 2002, there has been tremendous growth of preclinical and clinical studies on the use of PDE5 inhibitors for cardiovascular diseases and cancer. Numerous animal studies have demonstrated that PDE5 inhibitors have powerful protective effect against myocardial ischemia/reperfusion (I/R) injury, doxorubicin cardiotoxicity, ischemic and diabetic cardiomyopathy, cardiac hypertrophy, Duchenne muscular dystrophy and the improvement of stem cell efficacy for myocardial repair. Mechanistically, PDE5 inhibitors protect the heart against I/R injury through increased expression of nitric oxide synthases, activation of protein kinase G (PKG), PKG-dependent hydrogen sulfide generation, and phosphorylation of glycogen synthase kinase-3β - a master switch immediately proximal to mitochondrial permeability transition pore and the end effector of cardioprotection. In addition, PDE5 inhibitors enhance the sensitivity of certain types of cancer to standard chemotherapeutic drugs, including doxorubicin. Many clinical trials with PDE5 inhibitors have focused on the potential cardiovascular and anti-cancer benefits. Despite mixed results of these clinical trials, there is a continuing strong interest by basic scientists and clinical investigators in exploring their new clinical uses. It is our hope that future new mechanistic investigations and carefully designed clinical trials would help in reaping additional benefits of PDE5 inhibitors for cardiovascular disease and cancer in patients.
Collapse
Affiliation(s)
- Anindita Das
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - David Durrant
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Fadi N Salloum
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Rakesh C Kukreja
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|
123
|
Giannetta E, Feola T, Gianfrilli D, Pofi R, Dall'Armi V, Badagliacca R, Barbagallo F, Lenzi A, Isidori AM. Is chronic inhibition of phosphodiesterase type 5 cardioprotective and safe? A meta-analysis of randomized controlled trials. BMC Med 2014; 12:185. [PMID: 25330139 PMCID: PMC4201993 DOI: 10.1186/s12916-014-0185-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/17/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The myocardial effects of phosphodiesterase type 5 inhibitors (PDE5i) have recently received consideration in several preclinical studies. The risk/benefit ratio in humans remains unclear. METHODS We performed a meta-analysis of randomized, placebo-controlled trials (RCTs) to evaluate the efficacy and safety of PDE5i on cardiac morphology and function. From March 2012 to December 2013 (update: May 2014), we searched English-language studies from MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials and SCOPUS-selecting RCTs of continuous PDE5i administration that reported cardiovascular outcomes: cardiac geometry and performance, afterload, endothelial function and safety. The pooled estimate of a weighted mean difference between treatment and placebo was obtained for all outcomes using a random effects model. A test for heterogeneity was performed and the I2 statistic calculated. RESULTS Overall, 1,622 subjects were treated, with 954 randomized to PDE5i and 772 to placebo in 24 RCTs. According to our analysis, sustained PDE5 inhibition produced: (1) an anti-remodeling effect by reducing cardiac mass (-12.21 g/m2, 95% confidence interval (CI): -18.85; -5.57) in subjects with left ventricular hypertrophy (LVH) and by increasing end-diastolic volume (5.00 mL/m2; 95% CI: 3.29; 6.71) in non-LVH patients; (2) an improvement in cardiac performance by increasing cardiac index (0.30 L/min/m2, 95% CI: 0.202; 0.406) and ejection fraction (3.56%, 95% CI: 1.79; 5.33). These effects are parallel to a decline of N-terminal-pro brain natriuretic peptide (NT-proBNP) in subjects with severe LVH (-486.7 pg/ml, 95% CI: -712; -261). PDE5i administration also produced: (3) no changes in afterload parameters and (4) an improvement in flow-mediated vasodilation (3.31%, 95% CI: 0.53; 6.08). Flushing, headache, epistaxis and gastric symptoms were the commonest side effects. CONCLUSIONS This meta-analysis suggests for the first time that PDE5i have anti-remodeling properties and improve cardiac inotropism, independently of afterload changes, with a good safety profile. Given the reproducibility of the findings and tolerability across different populations, PDE5i could be reasonably offered to men with cardiac hypertrophy and early stage heart failure. Given the limited gender data, a larger trial on the sex-specific response to long-term PDE5i treatment is required.
Collapse
Affiliation(s)
- Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| | - Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| | - Valentina Dall'Armi
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana of Rome, Via della Pisana 235, Rome, 00163, Italy.
| | - Roberto Badagliacca
- Department of Cardiovascular and Respiratory Science, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy.
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, 00161, Italy.
| |
Collapse
|
124
|
|
125
|
Corona G, Maseroli E, Rastrelli G, Isidori AM, Sforza A, Mannucci E, Maggi M. Cardiovascular risk associated with testosterone-boosting medications: a systematic review and meta-analysis. Expert Opin Drug Saf 2014; 13:1327-51. [PMID: 25139126 DOI: 10.1517/14740338.2014.950653] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Recent reports have significantly halted the enthusiasm regarding androgen-boosting; suggesting that testosterone supplementation (TS) increases cardiovascular (CV) events. AREAS COVERED In order to overcome some of the limitations of the current evidence, the authors performed an updated systematic review and meta-analysis of all placebo-controlled randomized clinical trials (RCTs) on the effect of TS on CV-related problems. Out of 2747 retrieved articles, 75 were analyzed, including 3016 and 2448 patients in TS and placebo groups, respectively, and a mean duration of 34 weeks. Our analyses, performed on the largest number of studies collected so far, indicate that TS is not related to any increase in CV risk, even when composite or single adverse events were considered. In RCTs performed in subjects with metabolic derangements a protective effect of TS on CV risk was observed. EXPERT OPINION The present systematic review and meta-analysis does not support a causal role between TS and adverse CV events. Our results are in agreement with a large body of literature from the last 20 years supporting TS of hypogonadal men as a valuable strategy in improving a patient's metabolic profile, reducing body fat and increasing lean muscle mass, which would ultimately reduce the risk of heart disease.
Collapse
Affiliation(s)
- Giovanni Corona
- Azienda-Usl Bologna, Maggiore-Bellaria Hospital, Medical Department, Endocrinology Unit , Bologna , Italy
| | | | | | | | | | | | | |
Collapse
|
126
|
Abstract
Diabetes and insulin resistance have a variety of detrimental effects on cardiovascular health and outcomes. Cardiac magnetic resonance offers a non-invasive means to obtain many layers of information at a tissue level, including fibrosis, edema, intramyocardial motion, triglyceride content, and myocardial energetics. The role of cardiovascular magnetic resonance is particularly important in the evaluation of recognized and unrecognized coronary artery disease. In this review, we address the current state-of-the-art in cardiac magnetic resonance imaging - for both clinical and investigational use - as it applies to diabetic cardiovascular disease.
Collapse
Affiliation(s)
- Ravi V Shah
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | | | | |
Collapse
|
127
|
Larghat AM, Swoboda PP, Biglands JD, Kearney MT, Greenwood JP, Plein S. The microvascular effects of insulin resistance and diabetes on cardiac structure, function, and perfusion: a cardiovascular magnetic resonance study. Eur Heart J Cardiovasc Imaging 2014; 15:1368-76. [PMID: 25117473 PMCID: PMC4240406 DOI: 10.1093/ehjci/jeu142] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aims Type 2 diabetes mellitus is an independent risk factor for the development of heart failure. To better understand the mechanism by which this occurs, we investigated cardiac structure, function, and perfusion in patients with and without diabetes. Methods and results Sixty-five patients with no stenosis >30% on invasive coronary angiography were categorized into diabetes (19) and non-diabetes (46) which was further categorized into prediabetes (30) and controls (16) according to the American Diabetes Association guidelines. Each patient underwent comprehensive cardiovascular magnetic resonance assessment. Left-ventricular (LV) mass, relative wall mass (RWM), Lagrangian circumferential strain, LV torsion, and myocardial perfusion reserve (MPR) were calculated. LV mass was higher in diabetics than non-diabetics (112.8 ± 39.7 vs. 91.5 ± 21.3 g, P = 0.01) and in diabetics than prediabetics (112.8 ± 39.7 vs. 90.3 ± 18.7 g, P = 0.02). LV torsion angle was higher in diabetics than non-diabetics (9.65 ± 1.90 vs. 8.59 ± 1.91°, P = 0.047), and MPR was lower in diabetics than non-diabetics (2.10 ± 0.76 vs. 2.84 ± 1.25 mL/g/min, P = 0.01). There was significant correlation between MPR and early diastolic strain rate (r = −0.310, P = 0.01) and LV torsion (r = −0.306, P = 0.01). In multivariable linear regression analysis, non-diabetics waist–hip ratio, but not body mass index, had a significant association with RWM (Beta = 0.34, P = 0.02). Conclusion Patients with diabetes have increased LV mass, LV torsion, and decreased MPR. There is a significant association between decreased MPR and increased LV torsion suggesting a possible mechanistic link between microvascular disease and cardiac dysfunction in diabetes.
Collapse
Affiliation(s)
- Abdulghani M Larghat
- Multidisciplinary Cardiovascular Research Centre & Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds LS2 9JT, UK Department of Cardiology and Internal Medicine, Tripoli Medical Centre, Faculty of Medicine, University of Tripoli, Tripoli, Libya
| | - Peter P Swoboda
- Multidisciplinary Cardiovascular Research Centre & Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | | | - Mark T Kearney
- Multidisciplinary Cardiovascular Research Centre & Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | - John P Greenwood
- Multidisciplinary Cardiovascular Research Centre & Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre & Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
128
|
Leung DG, Herzka DA, Thompson WR, He B, Bibat G, Tennekoon G, Russell SD, Schuleri KH, Lardo AC, Kass DA, Thompson RE, Judge DP, Wagner KR. Sildenafil does not improve cardiomyopathy in Duchenne/Becker muscular dystrophy. Ann Neurol 2014; 76:541-9. [PMID: 25042693 DOI: 10.1002/ana.24214] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/27/2014] [Accepted: 06/29/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Duchenne and Becker muscular dystrophies (DBMD) are allelic disorders caused by mutations in dystrophin. Adults with DBMD develop life-threatening cardiomyopathy. Inhibition of phosphodiesterase 5 (PDE5) improves cardiac function in mouse models of DBMD. To determine whether the PDE5-inhibitor sildenafil benefits human dystrophinopathy, we conducted a randomized, double-blind, placebo-controlled trial (ClinicalTrials.gov, number NCT01168908). METHODS Adults with DBMD and cardiomyopathy (ejection fraction ≤ 50%) were randomized to receive sildenafil (20mg 3× daily) or placebo for 6 months. All subjects received an additional 6 months of open-label sildenafil. The primary endpoint was change in left ventricular end-systolic volume (LVESV) on cardiac magnetic resonance imaging. Secondary cardiac endpoints, skeletal muscle function, and quality of life were also assessed. RESULTS An interim analysis (performed after 15 subjects completed the blinded phase) revealed that 29% (4 of 14) of subjects had a ≥10% increase in LVESV after 6 months of sildenafil compared to 13% (1 of 8) of subjects receiving placebo. Subjects with LVESV > 120ml at baseline were more likely to worsen at 12 months regardless of treatment assignment (p = 0.035). Due to the higher number of subjects worsening on sildenafil, the data and safety monitoring board recommended early termination of the study. There were no statistically significant differences in outcome measures between treatment arms. INTERPRETATION Due to the small sample size, comparisons between groups must be interpreted with caution. However, this trial suggests that sildenafil is unlikely to improve cardiac function in adults with DBMD.
Collapse
Affiliation(s)
- Doris G Leung
- Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD; Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Wang B, Yang Q, Sun YY, Xing YF, Wang YB, Lu XT, Bai WW, Liu XQ, Zhao YX. Resveratrol-enhanced autophagic flux ameliorates myocardial oxidative stress injury in diabetic mice. J Cell Mol Med 2014; 18:1599-611. [PMID: 24889822 PMCID: PMC4190906 DOI: 10.1111/jcmm.12312] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/31/2014] [Indexed: 01/02/2023] Open
Abstract
Autophagic dysfunction is observed in diabetes mellitus. Resveratrol has a beneficial effect on diabetic cardiomyopathy. Whether the resveratrol-induced improvement in cardiac function in diabetes is via regulating autophagy remains unclear. We investigated the mechanisms underlying resveratrol-mediated protection against heart failure in diabetic mice, with a focus on the role of sirtuin 1 (SIRT1) in regulating autophagic flux. Diabetic cardiomyopathy in mice was induced by streptozotocin (STZ). Long-term resveratrol treatment improved cardiac function, ameliorated oxidative injury and reduced apoptosis in the diabetic mouse heart. Western blot analysis revealed that resveratrol decreased p62 protein expression and promoted SIRT1 activity and Rab7 expression. Inhibiting autophagic flux with bafilomycin A1 increased diabetic mouse mortality and attenuated resveratrol-induced down-regulation of p62, but not SIRT1 activity or Rab7 expression in diabetic mouse hearts. In cultured H9C2 cells, redundant or overactive H2O2 increased p62 and cleaved caspase 3 expression as well as acetylated forkhead box protein O1 (FOXO1) and inhibited SIRT1 expression. Sirtinol, SIRT1 and Rab7 siRNA impaired the resveratrol amelioration of dysfunctional autophagic flux and reduced apoptosis under oxidative conditions. Furthermore, resveratrol enhanced FOXO1 DNA binding at the Rab7 promoter region through a SIRT1-dependent pathway. These results highlight the role of the SIRT1/FOXO1/Rab7 axis in the effect of resveratrol on autophagic flux in vivo and in vitro, which suggests a therapeutic strategy for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Bo Wang
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Carosa E, Castri A, Forcella C, Sebastiani G, Di Sante S, Gravina GL, Ronchi P, Cesarini V, Dolci S, Di Stasi S, Lenzi A, Jannini EA. Platelet-derived growth factor regulation of type-5 phosphodiesterase in human and rat penile smooth muscle cells. J Sex Med 2014; 11:1675-84. [PMID: 24836457 DOI: 10.1111/jsm.12568] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Relaxation of cavernous smooth muscle cells (SMCs) is a key component in the control of the erectile mechanism. SMCs can switch their phenotype from a contractile differentiated state to a proliferative and dedifferentiated state in response to a change of local environmental stimuli. Proliferation and contraction are both regulated by the intracellular second messengers cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), which are degraded by phosphodiesterases (PDEs). The most abundant PDE present in corpora cavernosa is the electrolytic cGMP-specific phosphodiesterase type 5 (PDE5). AIM We investigated the cellular localization of PDE5 in in vitro cultured corpora cavernosa cells and the effect of mitogenic stimulation on PDE5 expression. METHODS Biochemical ad molecular techniques on cultured SMCs from human and rat penis. MAIN OUTCOME MEASURES We studied the ability of the quiescent SMC phenotype vs. the proliferating phenotype in modulation of PDE5 expression. RESULTS We demonstrated that PDE5 is localized in the cytoplasm, in the perinuclear area, and in discrete cytoplasmic foci. As previously demonstrated in human myometrial cells, the cytoplasmic foci may correspond to centrosomes. In corpora cavernosa, PDE5 protein levels are strongly regulated by the mitotic activity of the SMCs, as they were increased in quiescent cultures. In contrast, treatment with platelet-derived grow factor (PDGF), one of the most powerful mitogenic factors for SMCs, reduces the expression of PDE5 after 24 hours of treatment. CONCLUSION We found that PDGF treatment downregulates PDE5 expression in proliferating SMCs, suggesting that PDE5 may represent one of the markers of the contractile phenotype of the SMCs of corpora cavernosa.
Collapse
Affiliation(s)
- Eleonora Carosa
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Schlueter N, de Sterke A, Willmes DM, Spranger J, Jordan J, Birkenfeld AL. Metabolic actions of natriuretic peptides and therapeutic potential in the metabolic syndrome. Pharmacol Ther 2014; 144:12-27. [PMID: 24780848 DOI: 10.1016/j.pharmthera.2014.04.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/14/2014] [Indexed: 12/26/2022]
Abstract
Natriuretic peptides (NPs) are a group of peptide-hormones mainly secreted from the heart, signaling via c-GMP coupled receptors. NP are well known for their renal and cardiovascular actions, reducing arterial blood pressure as well as sodium reabsorption. Novel physiological functions have been discovered in recent years, including activation of lipolysis, lipid oxidation, and mitochondrial respiration. Together, these responses promote white adipose tissue browning, increase muscular oxidative capacity, particularly during physical exercise, and protect against diet-induced obesity and insulin resistance. Exaggerated NP release is a common finding in congestive heart failure. In contrast, NP deficiency is observed in obesity and in type-2 diabetes, pointing to an involvement of NP in the pathophysiology of metabolic disease. Based upon these findings, the NP system holds the potential to be amenable to therapeutical intervention against pandemic diseases such as obesity, insulin resistance, and arterial hypertension. Various therapeutic approaches are currently under development. This paper reviews the current knowledge on the metabolic effects of the NP system and discusses potential therapeutic applications.
Collapse
Affiliation(s)
- Nina Schlueter
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Anita de Sterke
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Diana M Willmes
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Jens Jordan
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Andreas L Birkenfeld
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany.
| |
Collapse
|
132
|
Koka S, Aluri HS, Xi L, Lesnefsky EJ, Kukreja RC. Chronic inhibition of phosphodiesterase 5 with tadalafil attenuates mitochondrial dysfunction in type 2 diabetic hearts: potential role of NO/SIRT1/PGC-1α signaling. Am J Physiol Heart Circ Physiol 2014; 306:H1558-68. [PMID: 24727492 DOI: 10.1152/ajpheart.00865.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Enhanced nitric oxide (NO) production is known to activate silent information regulator 1 (SIRT1), which is a histone deacetylase that regulates PGC-1α, a regulator of mitochondrial biogenesis and coactivator of transcription factors impacting energy homeostasis. Since phosphodiesterase-5 inhibitors potentiate NO signaling, we hypothesized that chronic treatment with phosphodiesterase-5 inhibitor tadalafil would activate SIRT1-PGC-1α signaling and protect against metabolic stress-induced mitochondrial dysfunction in diabetic hearts. Diabetic db/db mice (n = 32/group; 40 wk old) were randomized to receive DMSO (10%, 0.2 ml ip) or tadalafil (1 mg/kg ip in 10% DMSO) for 8 wk. Wild-type C57BL mice served as nondiabetic controls. The hearts were excised and homogenized to study SIRT1 activity and downstream protein targets. Mitochondrial function was determined by measuring oxidative phosphorylation (OXPHOS), and reactive oxygen species generation was studied in isolated mitochondria. Tadalafil-treated diabetic mice demonstrated significantly improved left ventricular function, which is associated with increased cardiac SIRT1 activity. Tadalafil also enhanced plasma NO oxidation levels, myocardial SIRT1, PGC-1α expression, and phosphorylation of eNOS, Akt, and AMPK in the diabetic hearts. OXPHOS with the complex I substrate glutamate was decreased by 50% in diabetic hearts compared with the nondiabetic controls. Tadalafil protected OXPHOS with an improved glutamate state 3 respiration rates. The increased reactive oxygen species production from complex I was significantly decreased by tadalafil treatment. In conclusion, chronic treatment with tadalafil activates NO-induced SIRT1-PGC-1α signaling and attenuates mitochondrial dysfunction in type 2 diabetic hearts.
Collapse
Affiliation(s)
- Saisudha Koka
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Hema S Aluri
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Lei Xi
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Cardiology Section, Medical Service, McGuire Veterans Affairs Medical Center, Richmond, Virginia; and Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Rakesh C Kukreja
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
133
|
Therapeutic potential of PDE modulation in treating heart disease. Future Med Chem 2014; 5:1607-20. [PMID: 24047267 DOI: 10.4155/fmc.13.127] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Altered cyclic nucleotide-mediated signaling plays a critical role in the development of cardiovascular pathology. By degrading cAMP/cGMP, the action of cyclic nucleotide PDEs is essential for controlling cyclic nucleotide-mediated signaling intensity, duration, and specificity. Altered expression, localization and action of PDEs have all been implicated in causing changes in cyclic nucleotide signaling in cardiovascular disease. Accordingly, pharmacological inhibition of PDEs has gained interest as a treatment strategy and as an area of drug development. While targeting of certain PDEs has the potential to ameliorate cardiovascular disease, inhibition of others might actually worsen it. This review will highlight recent research on the physiopathological role of cyclic nucleotide signaling, especially with regard to PDEs. While the physiological roles and biochemical properties of cardiovascular PDEs will be summarized, the primary emphasis will be pathological. Research into the potential benefits and hazards of PDE inhibition will also be discussed.
Collapse
|
134
|
Hawi R, Bazi L, Farkouh ME, Aneja A. Imaging in diabetic cardiomyopathy. Expert Rev Cardiovasc Ther 2014. [DOI: 10.1586/14779072.2014.899903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
135
|
Isidori AM, Corona G, Aversa A, Gianfrilli D, Jannini EA, Foresta C, Maggi M, Lenzi A, SIAMS-ED Study Group AndòSebastianoAngellettiGabriellaMatteoBaldiBalerciaGiancarloGiuseppeBellastellaCalogeroAldo E.CanaleDomenicoCaprioMassimilianoCarettaNicolaErennioCiotoliColpiGiovanni MariaFabbriAndreaFornengoRiccardoFrancavillaSandrohttp://orcid.org/0000-0002-6644-2229FrancomanoDavideGavioliSilviaAngeloGiagulli VitoGiannettaElisaGogliaUmbertoNicolaIlacquaLa VigneraSandroLemmaAndreaManciniMarioManieriChiaraMansaniRiccardoTommasoMinistriniMinutoFrancescoAlessandroOppoPaggiFrancescaRosarioPivonelloFiorePelliccioneAnnaPerriPerriniSebastioRiccardoPofihttp://orcid.org/0000-0002-1147-6114PozzaCarlottaSbardellaEmiliaStefanoSerraSinisiAntonio. The SIAMS-ED Trial: A National, Independent, Multicentre Study on Cardiometabolic and Hormonal Impairment of Men with Erectile Dysfunction Treated with Vardenafil. Int J Endocrinol 2014; 2014:858715. [PMID: 24976827 PMCID: PMC4052518 DOI: 10.1155/2014/858715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 12/08/2013] [Accepted: 12/28/2013] [Indexed: 02/01/2023] Open
Abstract
Increased cardiovascular risk has been associated with reduced response to proerectile drugs. The Italian Society of Andrology and Sexual Medicine (SIAMS) promoted an independent, multicenter study performed in 604 men (55 ± 12 yrs) suffering from erectile dysfunction (ED) to assess multiple health outcomes and response to 6-month vardenafil challenge in a real-life setting. Overall, 30.8% men had metabolic syndrome. Cardiovascular risk stratification revealed a greater number of ED subjects with moderate risk of a major adverse cardiovascular event than the general population (P < 0.01). Age-adjusted pulse pressure was positively correlated with ED severity and negatively with androgens and waist circumference (P < 0.01). A decline in total testosterone was observed with increasing arterial pulse pressure (P < 0.05), which was not accompanied by compensatory LH rise. Follow-up on 185 men treated with vardenafil in an nonrandomized, open, single-arm trial documented a significant rise in IIEF-5 (delta = 6.1 ± 4.8) that was maintained in men with high cardiovascular risk. Mild adverse events occurred in <5%, with no differences between cardiovascular risk classes. In summary, ED is a frequent symptom in patients with an elevated, but often unknown, risk of future cardiovascular events. Androgens predict vascular resistance in ED patients. Vardenafil's response and safety profile were preserved in subjects with higher cardiovascular risk.
Collapse
Affiliation(s)
- Andrea M. Isidori
- Department Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | - Giovanni Corona
- Endocrinology Unit, Maggiore-Bellaria Hospital, 40133 Bologna, Italy
| | - Antonio Aversa
- Department Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | - Daniele Gianfrilli
- Department Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | | | - Carlo Foresta
- Centre Cryopreservation of Male Gamete, University of Padova, 35122 Padua, Italy
| | - Mario Maggi
- Biomedicine, University of Florence, 50121 Florence, Italy
| | - Andrea Lenzi
- Department Experimental Medicine, Sapienza University, 00161 Rome, Italy
- *Andrea Lenzi:
| | | |
Collapse
|
136
|
Roubille F, Busseuil D, Merlet N, Kritikou EA, Rhéaume E, Tardif JC. Investigational drugs targeting cardiac fibrosis. Expert Rev Cardiovasc Ther 2013; 12:111-25. [DOI: 10.1586/14779072.2013.839942] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
137
|
Crescioli C, Sturli N, Sottili M, Bonini P, Lenzi A, Di Luigi L. Insulin-like effect of the phosphodiesterase type 5 inhibitor tadalafil onto male human skeletal muscle cells. J Endocrinol Invest 2013; 36:1020-6. [PMID: 23873283 DOI: 10.3275/9034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Phosphodiesterase type 5 inhibitors (PDE5i), widely used to treat male erectile dysfunction, seem to counteract insulin resistance (IR) in animals and humans. IR, primarily manifest in peripheral tissues and particularly in skeletal muscle, is due to impaired insulin signal transduction. Investigators have been focusing onto intracellular defects responsible for IR to identify suitable pharmacological tools targeted toward the specific defects. Albeit some effects of PDE5i have been reported onto animal muscular tissues or cells, whether and how they might affect metabolic processes directly in human skeletal muscle still remains unclear. AIM We aimed to investigate in human fetal skeletal muscle cells (Hfsmc) the effect of tadalafil, one of PDE5i, onto some intracellular factors involved in response to insulin, such as ras-raf mitogen activated protein kinase (MAPK), phosphatidylinositol 3-kinase/protein kinase B (PKB/Akt), glycogen synthase kinase 3β (GSK-3β), and the transcriptional factor c-Myc; proliferation rate; lactate (lact) and free fatty acid (ffa) release; activity of citrate synthase (CS) and succinate dehydrogenase (SDH), both enzymes of Kreb's cycle; PDE5 gene expression. MATERIALS AND METHODS Western blot analysis, enzyme-linked immunosorbent assay, enzymatic assays, cell count, MTT assay and Real Time PCR were performed in Hfsmc with and without tadalafil. RESULTS In Hfsmc tadalafil affected the insulin-related intracellular cascade, by increasing MAPK, PKB/Akt, GSK-3β phosphorylation and c-Myc expression. ffa release and CS activity also significantly increased, with no changes in SDH activity and lact release. CONCLUSIONS Tadalafil, like insulin, targeted part of the machinery dedicated to energy management and metabolic control in human skeletal muscle cells.
Collapse
Affiliation(s)
- C Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", 00135 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
138
|
Gelzinis TA. New Insights Into Diastolic Dysfunction and Heart Failure With Preserved Ejection Fraction. Semin Cardiothorac Vasc Anesth 2013; 18:208-17. [DOI: 10.1177/1089253213510748] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
As the population ages, the incidence of patients presenting for surgical procedures with diastolic dysfunction and heart failure with preserved ejection fraction will rise. This review will discuss the most current and relevant information on the pathophysiology, treatment, and perioperative management of these patients.
Collapse
|
139
|
Circulation: Cardiovascular Imaging
Editors’ Picks. Circ Cardiovasc Imaging 2013. [DOI: 10.1161/circimaging.113.001335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
140
|
Pappachan JM, Varughese GI, Sriraman R, Arunagirinathan G. Diabetic cardiomyopathy: Pathophysiology, diagnostic evaluation and management. World J Diabetes 2013; 4:177-189. [PMID: 24147202 PMCID: PMC3797883 DOI: 10.4239/wjd.v4.i5.177] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/02/2013] [Accepted: 08/17/2013] [Indexed: 02/05/2023] Open
Abstract
Diabetes affects every organ in the body and cardiovascular disease accounts for two-thirds of the mortality in the diabetic population. Diabetes-related heart disease occurs in the form of coronary artery disease (CAD), cardiac autonomic neuropathy or diabetic cardiomyopathy (DbCM). The prevalence of cardiac failure is high in the diabetic population and DbCM is a common but underestimated cause of heart failure in diabetes. The pathogenesis of diabetic cardiomyopathy is yet to be clearly defined. Hyperglycemia, dyslipidemia and inflammation are thought to play key roles in the generation of reactive oxygen or nitrogen species which are in turn implicated. The myocardial interstitium undergoes alterations resulting in abnormal contractile function noted in DbCM. In the early stages of the disease diastolic dysfunction is the only abnormality, but systolic dysfunction supervenes in the later stages with impaired left ventricular ejection fraction. Transmitral Doppler echocardiography is usually used to assess diastolic dysfunction, but tissue Doppler Imaging and Cardiac Magnetic Resonance Imaging are being increasingly used recently for early detection of DbCM. The management of DbCM involves improvement in lifestyle, control of glucose and lipid abnormalities, and treatment of hypertension and CAD, if present. The role of vasoactive drugs and antioxidants is being explored. This review discusses the pathophysiology, diagnostic evaluation and management options of DbCM.
Collapse
|
141
|
Hamdani N, Franssen C, Lourenço A, Falcão-Pires I, Fontoura D, Leite S, Plettig L, López B, Ottenheijm CA, Becher PM, González A, Tschöpe C, Díez J, Linke WA, Leite-Moreira AF, Paulus WJ. Myocardial titin hypophosphorylation importantly contributes to heart failure with preserved ejection fraction in a rat metabolic risk model. Circ Heart Fail 2013; 6:1239-49. [PMID: 24014826 DOI: 10.1161/circheartfailure.113.000539] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Obesity and diabetes mellitus are important metabolic risk factors and frequent comorbidities in heart failure with preserved ejection fraction. They contribute to myocardial diastolic dysfunction (DD) through collagen deposition or titin modification. The relative importance for myocardial DD of collagen deposition and titin modification was investigated in obese, diabetic ZSF1 rats after heart failure with preserved ejection fraction development at 20 weeks. METHODS AND RESULTS Four groups of rats (Wistar-Kyoto, n=11; lean ZSF1, n=11; obese ZSF1, n=11, and obese ZSF1 with high-fat diet, n=11) were followed up for 20 weeks with repeat metabolic, renal, and echocardiographic evaluations and hemodynamically assessed at euthanization. Myocardial collagen, collagen cross-linking, titin isoforms, and phosphorylation were also determined. Resting tension (Fpassive)-sarcomere length relations were obtained in small muscle strips before and after KCl-KI treatment, which unanchors titin and allows contributions of titin and extracellular matrix to Fpassive to be discerned. At 20 weeks, the lean ZSF1 group was hypertensive, whereas both obese ZSF1 groups were hypertensive and diabetic. Only the obese ZSF1 groups had developed heart failure with preserved ejection fraction, which was evident from increased lung weight, preserved left ventricular ejection fraction, and left ventricular DD. The underlying myocardial DD was obvious from high muscle strip stiffness, which was largely (±80%) attributable to titin hypophosphorylation. The latter occurred specifically at the S3991 site of the elastic N2Bus segment and at the S12884 site of the PEVK segment. CONCLUSIONS Obese ZSF1 rats developed heart failure with preserved ejection fraction during a 20-week time span. Titin hypophosphorylation importantly contributed to the underlying myocardial DD.
Collapse
Affiliation(s)
- Nazha Hamdani
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Isidori AM, Buvat J, Corona G, Goldstein I, Jannini EA, Lenzi A, Porst H, Salonia A, Traish AM, Maggi M. A critical analysis of the role of testosterone in erectile function: from pathophysiology to treatment-a systematic review. Eur Urol 2013; 65:99-112. [PMID: 24050791 DOI: 10.1016/j.eururo.2013.08.048] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/16/2013] [Indexed: 12/15/2022]
Abstract
CONTEXT Androgen modulation of erectile function (EF) is widely accepted. However, the use of testosterone replacement therapy (TRT) in men with erectile dysfunction (ED) has generated an unprecedented debate. OBJECTIVE To summarize the relevant data on the incidence, diagnosis, and management of ED coexisting with hypogonadism and to develop a pathophysiology-based treatment algorithm. EVIDENCE ACQUISITION We reviewed the relevant medical literature, with a particular emphasis on original molecular studies, prospective observational data, and randomized controlled trials performed in the past 20 yr. EVIDENCE SYNTHESIS Testosterone modulates nearly every component involved in EF, from pelvic ganglions to smooth muscle and the endothelial cells of the corpora cavernosa. It also regulates the timing of the erectile process as a function of sexual desire, coordinating penile erection with sex. Epidemiologic studies confirm the significant overlap of hypogonadism and ED; however, most guidelines do not consider the differential diagnosis of hypogonadism or the relevance of subclinical disease. Various clinical tools can help the physician to assess and restore androgen levels in men with ED. Special attention is given to fertility-sparing treatments, due to the increasing number of older men desiring fatherhood. The simultaneous use of phosphodiesterase type 5 inhibitors (PDE5-Is) and TRT has recently been questioned. Originally proposed as a salvage therapy for nonresponders to PDE5-Is, this approach has been inappropriately transformed into a combination therapy. Clinical data are consistent when reinterpreted in the proper framework, whereas molecular evidence remains controversial. CONCLUSIONS A body of molecular and clinical evidence supports the use of TRT in hypogonadal patients with ED, although the benefit-risk ratio is uncertain in advanced age. Critical appraisal of this evidence enabled the development of a pathophysiology-oriented algorithm designed to avoid inappropriate treatments and support whether to start with TRT, PDE5-I only, or both. Apparently divergent findings are reconciled when TRT is correctly indicated. An improved diagnosis and individualized management is desirable in light of the many available options.
Collapse
|
143
|
Corona G, Maggi M. Conventional and unconventional cardiovascular risk factors in men with erectile dysfunction. J Sex Med 2013; 10:305-8. [PMID: 23350612 DOI: 10.1111/jsm.12075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
144
|
Konstantinou DM, Chatzizisis YS, Giannoglou GD. Pathophysiology-based novel pharmacotherapy for heart failure with preserved ejection fraction. Pharmacol Ther 2013; 140:156-66. [PMID: 23792088 DOI: 10.1016/j.pharmthera.2013.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 05/30/2013] [Indexed: 02/01/2023]
Abstract
Heart failure has become increasingly prevalent and poses a significant socioeconomic burden in the developed world. Approximately half of heart failure patients have preserved ejection fraction (HFpEF) and experience an increased morbidity and mortality attributed to the lack of effective therapies and to the presence of comorbidities. Suppression of neurohormonal activation by beta-blockers and renin-angiotensin-aldosterone system inhibitors is the cornerstone in the pharmacotherapy of heart failure with reduced ejection fraction (HFrEF). However, these medications are not associated with significant clinical benefit in HFpEF. In this review, we provide an in-depth pathophysiology-based update on novel pharmacotherapies of HFpEF. A deeper insight into the pathophysiologic mechanisms of HFpEF may create opportunities for novel pharmacological interventions.
Collapse
Affiliation(s)
- Dimitrios M Konstantinou
- First Department of Cardiology, AHEPA University Hospital, Aristotle University Medical School, Thessaloniki, Greece; Heart Failure Care Group, The Royal Brompton Hospital, London Imperial College, London, UK
| | | | | |
Collapse
|
145
|
Bruzziches R, Francomano D, Gareri P, Lenzi A, Aversa A. An update on pharmacological treatment of erectile dysfunction with phosphodiesterase type 5 inhibitors. Expert Opin Pharmacother 2013; 14:1333-44. [PMID: 23675780 DOI: 10.1517/14656566.2013.799665] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Phosphodiesterase type 5 inhibitors (PDE5-i) are used for the oral treatment of erectile dysfunction (ED). Since the launch of sildenafil more than 15 years ago, new molecules have become available. At present, in addition to tadalafil and vardenafil, there are three other drugs, udenafil, avanafil and mirodenafil, marketed in some countries which appear to be promising. AREAS COVERED The clinical pharmacological differences in dosage and side effects of all PDE5-i are evaluated. EXPERT OPINION All PDE5-i are equally effective and safe for the treatment of ED. On-demand use of any PDE5-i is also safe for patients with comorbid conditions. Tadalafil seems to be the preferred drug by patients and physicians, probably due to its peculiar pharmacological profile that makes sexual intercourse more spontaneous for the patients. Preliminary data suggest that the use of vardenafil may also be beneficial in cases of ED associated with premature ejaculation. Daily treatment is another option in men with ED and documented vascular or prostate disease. In geriatric or in difficult-to-treat populations, the evaluation of testosterone plasma levels will help to predict the efficacy of any PDE5-i. Remarkably, when such drugs are withdrawn for any reason, ED most often continues to occur because of the presence of an underlying disease.
Collapse
Affiliation(s)
- Roberto Bruzziches
- Sapienza University of Rome, Endocrinology and Food and Science Section, Rome, Italy
| | | | | | | | | |
Collapse
|
146
|
Paulus WJ, Tschöpe C. A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol 2013; 62:263-71. [PMID: 23684677 DOI: 10.1016/j.jacc.2013.02.092] [Citation(s) in RCA: 2545] [Impact Index Per Article: 212.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/07/2013] [Accepted: 02/05/2013] [Indexed: 12/17/2022]
Abstract
Over the past decade, myocardial structure, cardiomyocyte function, and intramyocardial signaling were shown to be specifically altered in heart failure with preserved ejection fraction (HFPEF). A new paradigm for HFPEF development is therefore proposed, which identifies a systemic proinflammatory state induced by comorbidities as the cause of myocardial structural and functional alterations. The new paradigm presumes the following sequence of events in HFPEF: 1) a high prevalence of comorbidities such as overweight/obesity, diabetes mellitus, chronic obstructive pulmonary disease, and salt-sensitive hypertension induce a systemic proinflammatory state; 2) a systemic proinflammatory state causes coronary microvascular endothelial inflammation; 3) coronary microvascular endothelial inflammation reduces nitric oxide bioavailability, cyclic guanosine monophosphate content, and protein kinase G (PKG) activity in adjacent cardiomyocytes; 4) low PKG activity favors hypertrophy development and increases resting tension because of hypophosphorylation of titin; and 5) both stiff cardiomyocytes and interstitial fibrosis contribute to high diastolic left ventricular (LV) stiffness and heart failure development. The new HFPEF paradigm shifts emphasis from LV afterload excess to coronary microvascular inflammation. This shift is supported by a favorable Laplace relationship in concentric LV hypertrophy and by all cardiac chambers showing similar remodeling and dysfunction. Myocardial remodeling in HFPEF differs from heart failure with reduced ejection fraction, in which remodeling is driven by loss of cardiomyocytes. The new HFPEF paradigm proposes comorbidities, plasma markers of inflammation, or vascular hyperemic responses to be included in diagnostic algorithms and aims at restoring myocardial PKG activity.
Collapse
Affiliation(s)
- Walter J Paulus
- Department of Physiology, Institute for Cardiovascular Research VU, VU University Medical Center Amsterdam, Amsterdam, the Netherlands.
| | | |
Collapse
|
147
|
van Heerebeek L, Franssen CPM, Hamdani N, Verheugt FWA, Somsen GA, Paulus WJ. Molecular and cellular basis for diastolic dysfunction. Curr Heart Fail Rep 2013; 9:293-302. [PMID: 22926993 DOI: 10.1007/s11897-012-0109-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is highly prevalent and is frequently associated with metabolic risk factors. Patients with HFpEF have only a slightly lower mortality than patients with HF and reduced EF. The pathophysiology of HFpEF is currently incompletely understood, which precludes specific therapy. Both HF phenotypes demonstrate distinct cardiac remodeling processes at the macroscopic, microscopic, and ultrastructural levels. Increased diastolic left-ventricular (LV) stiffness and impaired LV relaxation are important features of HFpEF, which can be explained by changes in the extracellular matrix and the cardiomyocytes. In HFpEF, elevated intrinsic cardiomyocyte stiffness contributes to high diastolic LV stiffness. Posttranslational changes in the sarcomeric protein titin, affecting titin isoform expression and phosphorylation, contribute to elevated cardiomyocyte stiffness. Increased nitrosative/oxidative stress, impaired nitric oxide bioavailability, and down-regulation of myocardial cyclic guanosine monophosphate and protein kinase G signaling could trigger posttranslational modifications of titin, thereby augmenting cardiomyocyte and LV diastolic stiffness.
Collapse
Affiliation(s)
- Loek van Heerebeek
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
148
|
van Berlo JH, Maillet M, Molkentin JD. Signaling effectors underlying pathologic growth and remodeling of the heart. J Clin Invest 2013; 123:37-45. [PMID: 23281408 DOI: 10.1172/jci62839] [Citation(s) in RCA: 345] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular disease is the number one cause of mortality in the Western world. The heart responds to many cardiopathological conditions with hypertrophic growth by enlarging individual myocytes to augment cardiac pump function and decrease ventricular wall tension. Initially, such cardiac hypertrophic growth is often compensatory, but as time progresses these changes become maladaptive. Cardiac hypertrophy is the strongest predictor for the development of heart failure, arrhythmia, and sudden death. Here we discuss therapeutic avenues emerging from molecular and genetic studies of cardiovascular disease in animal models. The majority of these are based on intracellular signaling pathways considered central to pathologic cardiac remodeling and hypertrophy, which then leads to heart failure. We focus our discussion on selected therapeutic targets that have more recently emerged and have a tangible translational potential given the available pharmacologic agents that could be readily evaluated in human clinical trials.
Collapse
Affiliation(s)
- Jop H van Berlo
- Department of Pediatrics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, Ohio 45229-3039, USA
| | | | | |
Collapse
|
149
|
Lee DI, Kass DA. Phosphodiesterases and cyclic GMP regulation in heart muscle. Physiology (Bethesda) 2012; 27:248-58. [PMID: 22875455 DOI: 10.1152/physiol.00011.2012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The cyclic nucleotide cGMP and its corresponding activated kinase cGK-1 serve as a counterbalance to acute and chronic myocardial stress. cGMP hydrolysis by several members of the phosphodiesterase (PDE) superfamily, PDE1, PDE2, and PDE5, regulate this signaling in the heart. This review details new insights regarding how these PDEs modulate cGMP and cGK-1 to influence heart function and chronic stress responses, and how their inhibition may provide potential therapeutic benefits.
Collapse
Affiliation(s)
- Dong I Lee
- Division of Cardiology, Department of Medicine, The Johns Hopkins University Medical Institutions, Baltimore, Maryland, USA
| | | |
Collapse
|
150
|
Kass DA. Cardiac role of cyclic-GMP hydrolyzing phosphodiesterase type 5: from experimental models to clinical trials. Curr Heart Fail Rep 2012; 9:192-9. [PMID: 22798047 DOI: 10.1007/s11897-012-0101-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cyclic guanosine monophosphate (cGMP) and its primary signaling kinase, protein kinase G, play an important role in counterbalancing stress remodeling in the heart. Growing evidence supports a positive impact on a variety of cardiac disease conditions from the suppression of cGMP hydrolysis. The latter is regulated by members of the phosphodiesterase (PDE) superfamily, of which cGMP-selective PDE5 has been best studied. Inhibitors such as sildenafil and tadalafil ameliorate cardiac pressure and volume overload, ischemic injury, and cardiotoxicity. Clinical trials have begun exploring their potential to benefit dilated cardiomyopathy and heart failure with a preserved ejection fraction. This review discusses recent developments in the field, highlighting basic science and clinical studies.
Collapse
Affiliation(s)
- David A Kass
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Ross Building, Room 858, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| |
Collapse
|