1
|
Abstract
Heart failure (HF) is a common consequence of several cardiovascular diseases and is understood as a vicious cycle of cardiac and hemodynamic decline. The current inventory of treatments either alleviates the pathophysiological features (eg, cardiac dysfunction, neurohumoral activation, and ventricular remodeling) and/or targets any underlying pathologies (eg, hypertension and myocardial infarction). Yet, since these do not provide a cure, the morbidity and mortality associated with HF remains high. Therefore, the disease constitutes an unmet medical need, and novel therapies are desperately needed. Cyclic guanosine-3',5'-monophosphate (cGMP), synthesized by nitric oxide (NO)- and natriuretic peptide (NP)-responsive guanylyl cyclase (GC) enzymes, exerts numerous protective effects on cardiac contractility, hypertrophy, fibrosis, and apoptosis. Impaired cGMP signaling, which can occur after GC deactivation and the upregulation of cyclic nucleotide-hydrolyzing phosphodiesterases (PDEs), promotes cardiac dysfunction. In this study, we review the role that NO/cGMP and NP/cGMP signaling plays in HF. After considering disease etiology, the physiological effects of cGMP in the heart are discussed. We then assess the evidence from preclinical models and patients that compromised cGMP signaling contributes to the HF phenotype. Finally, the potential of pharmacologically harnessing cardioprotective cGMP to rectify the present paucity of effective HF treatments is examined.
Collapse
|
2
|
Premer C, Kanelidis AJ, Hare JM, Schulman IH. Rethinking Endothelial Dysfunction as a Crucial Target in Fighting Heart Failure. Mayo Clin Proc Innov Qual Outcomes 2019; 3:1-13. [PMID: 30899903 PMCID: PMC6408687 DOI: 10.1016/j.mayocpiqo.2018.12.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is characterized by nitric oxide dysregulation and an altered redox state. Oxidative stress and inflammatory markers prevail, thus promoting atherogenesis and hypertension, important risk factors for the development and progression of heart failure. There has been a reemerging interest in the role that endothelial dysfunction plays in the failing circulation. Accordingly, patients with heart failure are being clinically assessed for endothelial dysfunction via various methods, including flow-mediated vasodilation, peripheral arterial tonometry, quantification of circulating endothelial progenitor cells, and early and late endothelial progenitor cell outgrowth measurements. Although the mechanisms underlying endothelial dysfunction are intimately related to cardiovascular disease and heart failure, it remains unclear whether targeting endothelial dysfunction is a feasible strategy for ameliorating heart failure progression. This review focuses on the pathophysiology of endothelial dysfunction, the mechanisms linking endothelial dysfunction and heart failure, and the various diagnostic methods currently used to measure endothelial function, ultimately highlighting the therapeutic implications of targeting endothelial dysfunction for the treatment of heart failure.
Collapse
Key Words
- Ach, acetylcholine
- CAD, coronary artery disease
- CVD, cardiovascular disease
- ECFC, endothelial colony-forming cell
- EDHF, endothelium-derived hyperpolarizing factor
- EPC, endothelial progenitor cell
- EPC-CFU, EPC–colony-forming unit
- FMD, flow-mediated vasodilation
- H2O2, hydrogen peroxide
- HF, heart failure
- HFpEF, HF with preserved ejection fraction
- HFrEF, HF with reduced ejection fraction
- IVUS, intravascular ultrasound
- LVEF, left ventricular ejection fraction
- NO, nitric oxide
- NOS, NO synthase
- PAT, peripheral arterial tonometry
- QCA, quantitative coronary angiography
- ROS, reactive oxygen species
- cGMP, cyclic guanosine monophosphate
- eNOS, endothelial nitric oxide synthase
Collapse
Affiliation(s)
- Courtney Premer
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL.,Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
3
|
Gulati G, Udelson JE. Heart Failure With Improved Ejection Fraction: Is it Possible to Escape One's Past? JACC-HEART FAILURE 2018; 6:725-733. [PMID: 30098965 DOI: 10.1016/j.jchf.2018.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022]
Abstract
Among patients with heart failure with reduced ejection fraction, investigators have repeatedly identified a subgroup whose left ventricular ejection fraction and structural remodeling can improve to normal or nearly normal levels with or without medical therapy. This subgroup of patients with "heart failure with improved ejection fraction" has distinct clinical characteristics and a more favorable prognosis compared with patients who continue to have reduced ejection fraction. However, many of these patients also manifest clinical and biochemical signs of incomplete resolution of heart failure pathophysiology and remain at some risk of adverse outcomes, thus indicating that they may not have completely recovered. Although rigorous evidence on managing these patients is sparse, there are several reasons to recommend continuation of heart failure therapies, including device therapies, to prevent clinical deterioration. Notable exceptions to this recommendation may include patients who recover from peripartum cardiomyopathy, fulminant myocarditis, or stress cardiomyopathy, whose excellent long-term prognoses may imply true myocardial recovery. More research on these patients is needed to better understand the mechanisms that lead to improvement in ejection fraction and to guide their clinical management.
Collapse
Affiliation(s)
- Gaurav Gulati
- Division of Cardiology and the CardioVascular Center, Tufts Medical Center, Boston, Massachusetts
| | - James E Udelson
- Division of Cardiology and the CardioVascular Center, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
4
|
Bae H, Choi J, Kim YW, Lee D, Kim JH, Ko JH, Bang H, Kim T, Lim I. Effects of Nitric Oxide on Voltage-Gated K⁺ Currents in Human Cardiac Fibroblasts through the Protein Kinase G and Protein Kinase A Pathways but Not through S-Nitrosylation. Int J Mol Sci 2018. [PMID: 29534509 PMCID: PMC5877675 DOI: 10.3390/ijms19030814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study investigated the expression of voltage-gated K+ (KV) channels in human cardiac fibroblasts (HCFs), and the effect of nitric oxide (NO) on the KV currents, and the underlying phosphorylation mechanisms. In reverse transcription polymerase chain reaction, two types of KV channels were detected in HCFs: delayed rectifier K+ channel and transient outward K+ channel. In whole-cell patch-clamp technique, delayed rectifier K+ current (IK) exhibited fast activation and slow inactivation, while transient outward K+ current (Ito) showed fast activation and inactivation kinetics. Both currents were blocked by 4-aminopyridine. An NO donor, S-nitroso-N-acetylpenicillamine (SNAP), increased the amplitude of IK in a concentration-dependent manner with an EC50 value of 26.4 µM, but did not affect Ito. The stimulating effect of SNAP on IK was blocked by pretreatment with 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or by KT5823. 8-bromo-cyclic GMP stimulated the IK. The stimulating effect of SNAP on IK was also blocked by pretreatment with KT5720 or by SQ22536. Forskolin and 8-bromo-cyclic AMP each stimulated IK. On the other hand, the stimulating effect of SNAP on IK was not blocked by pretreatment of N-ethylmaleimide or by DL-dithiothreitol. Our data suggest that NO enhances IK, but not Ito, among KV currents of HCFs, and the stimulating effect of NO on IK is through the PKG and PKA pathways, not through S-nitrosylation.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Jeongyoon Choi
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Young-Won Kim
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Donghee Lee
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Jung-Ha Kim
- Department of Family Medicine, College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Seoul 06973, Korea.
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Hyoweon Bang
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Seoul 06973, Korea.
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| |
Collapse
|
5
|
Bonafede R, Manucha W. Óxido nítrico y factores relacionados a oxidación e inflamación como posibles biomarcadores de insuficiencia cardíaca. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2018; 30:84-94. [PMID: 29477484 DOI: 10.1016/j.arteri.2017.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/13/2017] [Indexed: 11/25/2022]
|
6
|
Li H, Wang C, Sun J, Liu C, Li N, Chen J. Pravastatin Decreases Infarct Size Induced by Coronary Artery Ischemia/Reperfusion with Elevated eNOS Expression in Rats. Int Heart J 2018; 59:154-160. [PMID: 29375110 DOI: 10.1536/ihj.16-607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Our previous study showed that pravastatin prevents ischemia and reperfusion-induced lethal ventricular fibrillation in rats. This study explored whether pravastatin decreases myocardial infarct size and this effect is associated with endothelial nitric oxide synthase (eNOS) expression in myocardium. Rats were treated with ischemia (30 minutes) and reperfusion (60 minutes) after chronic oral administration of pravastatin, fluvastatin, or vehicle once daily for 22 days. Electrocardiograms and blood pressure were continuously recorded, myocardial infarct size was measured by TTC-staining, and eNOS expression was measured by western blot. The results showed that pravastatin and fluvastatin significantly reduced myocardial infarct size. No statistical differences were found in the areas at risk among all groups. However, a significant reduction in infarct size was observed in three pravastatin groups and one fluvastatin group compared to control. Both pravastatin and fluvastatin significantly increased eNOS protein expression in ischemic and non-ischemic tissues compared to control. Our results suggest that pravastatin decreases cardiovascular mortality beyond its cholesterol-lowering effect. Pravastatin is more potent than fluvastatin in reducing infarct size. These effects may be associated with elevation of eNOS expression.
Collapse
Affiliation(s)
- He Li
- Department of Pharmacology, Pharmaceutical College, Beihua University
| | - Chunmei Wang
- Department of Pharmacology, Pharmaceutical College, Beihua University
| | - Jinghui Sun
- Department of Pharmacology, Pharmaceutical College, Beihua University
| | - Cong Liu
- Department of Pharmacology, Pharmaceutical College, Beihua University
| | - Ning Li
- Department of Pharmacology, Pharmaceutical College, Beihua University
| | - Jianguang Chen
- Department of Pharmacology, Pharmaceutical College, Beihua University
| |
Collapse
|
7
|
Park SI, Kim J, Yu KS, Jang IJ, Lee S. Changes in Cardiac Function After a Single Intravenous Administration of CKD-712 in Healthy Male Volunteers. Clin Drug Investig 2017; 37:393-403. [PMID: 28160190 DOI: 10.1007/s40261-017-0494-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVES CKD-712, a candidate treatment for septic shock, acts by increasing cardiac output. This study investigated changes in the pharmacodynamics, pharmacokinetics, and tolerability of CKD-712 after a single intravenous administration. METHODS A dose-block-randomized, double-blind, placebo-controlled, single-dose escalation study was conducted in 44 healthy subjects receiving 20, 40, 80, 160, 240, or 320 μg/kg CKD-712 or placebo. Pharmacodynamics were evaluated using computerized impedance cardiography, vital signs, platelet aggregation, and bleeding time. Serial blood and urine samples for pharmacokinetic analysis were collected up to 12 and 24 h, respectively, after the initiation of intravenous drug infusion. Tolerability assessments were performed throughout the study. RESULTS The area under the effect-time curve of the cardiac index (AUECCI) and systolic blood pressure (AUECSBP) changed significantly with the 160 and 320 µg/kg doses of CKD-712 compared with placebo. Furthermore, the AUECCI and AUECSBP tended to increase as the systemic exposure of CKD-712 increased from 20 to 240 µg/kg. The frequency of drug-related adverse events (AEs), including cardiovascular symptoms, was higher with the 320 µg/kg dose. CONCLUSION The pharmacological effects and on-target AEs of CKD-712 increased relative to the dose increments. The results of this study suggest that potentially therapeutic doses of CKD-712 could range from 160 to 240 μg/kg.
Collapse
Affiliation(s)
- Sang-In Park
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - JaeWoo Kim
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Ledvényiová-Farkašová V, Bernátová I, Balis P, Puzserova A, Barteková M, Gablovsky I, Ravingerová T. Effect of crowding stress on tolerance to ischemia-reperfusion injury in young male and female hypertensive rats: molecular mechanisms. Can J Physiol Pharmacol 2015; 93:793-802. [PMID: 26317433 DOI: 10.1139/cjpp-2015-0026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sex and social stress may represent risk factors in the etiology of hypertension and heart response to ischemia-reperfusion (I/R) injury. Phosphatidylinositol 3-kinase/protein kinase B (Akt) plays an important role in the processes associated with hypertension and myocardial tolerance to I/R, and may be involved in myocardial stress reaction. The impact of chronic stress on the response to I/R was investigated in the hearts of 7-week-old spontaneously hypertensive (SHR) and Wistar-Kyoto (WKY) rats of both sexes. Stress was induced by reducing living space to 70 cm(2)/100 g body mass of rat for 2 weeks, while the controls were kept at 200 cm(2)/100 g. Langendorff-perfused hearts, subjected to I/R, exhibited higher vulnerability to ventricular tachycardia in crowd-stressed SHR vs. the control rats, and this was more pronounced in the males. Myocardial infarction was not affected by crowding stress in any of the groups. Male and female SHR showed increased activation of cardiac Akt, whereas nitric oxide synthase activity (NOS) with pro-apoptotic signaling decreased in the males but was not altered in the females (vs. WKY rats). NOS was enhanced in the female SHR and WKY groups by comparison with the respective males. Stress only reduced NOS activity in the SHR groups, and without changes in apoptotic markers. In conclusion, we showed that stress in young SHR mainly affects the nonlethal markers for I/R, and has no impact on myocardial infarction and apoptosis, despite reduced NOS activity.
Collapse
Affiliation(s)
| | - Iveta Bernátová
- b Institute of Normal and Pathological Physiology, Slovak Academy of Sciences and Centre of Excellence for Examination of Regulatory Role of Nitric Oxide in Civilization Diseases, Bratislava, Slovak Republic
| | - Peter Balis
- b Institute of Normal and Pathological Physiology, Slovak Academy of Sciences and Centre of Excellence for Examination of Regulatory Role of Nitric Oxide in Civilization Diseases, Bratislava, Slovak Republic
| | - Angelika Puzserova
- b Institute of Normal and Pathological Physiology, Slovak Academy of Sciences and Centre of Excellence for Examination of Regulatory Role of Nitric Oxide in Civilization Diseases, Bratislava, Slovak Republic
| | - Monika Barteková
- a Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Ivan Gablovsky
- a Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Tana Ravingerová
- a Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| |
Collapse
|
9
|
Drosatos K, Lymperopoulos A, Kennel PJ, Pollak N, Schulze PC, Goldberg IJ. Pathophysiology of sepsis-related cardiac dysfunction: driven by inflammation, energy mismanagement, or both? Curr Heart Fail Rep 2015; 12:130-140. [PMID: 25475180 PMCID: PMC4474734 DOI: 10.1007/s11897-014-0247-z] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Sepsis is a systemic inflammatory response that follows bacterial infection. Cardiac dysfunction is an important consequence of sepsis that affects mortality and has been attributed to either elevated inflammation or suppression of both fatty acid and glucose oxidation and eventual ATP depletion. Moreover, cardiac adrenergic signaling is compromised in septic patients and this aggravates further heart function. While anti-inflammatory therapies are important for the treatment of the disease, administration of anti-inflammatory drugs did not improve survival in septic patients. This review article summarizes findings on inflammatory and other mechanisms that are triggered in sepsis and affect cardiac function and mortality. Particularly, it focuses on the effects of the disease in metabolic pathways, as well as in adrenergic signaling and the potential interplay of the latter with inflammation. It is suggested that therapeutic approaches should include combination of anti-inflammatory treatments, stimulation of energy production, and restoration of adrenergic signaling in the heart.
Collapse
Affiliation(s)
- Konstantinos Drosatos
- Metabolic Biology Laboratory, Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, MERB-951, Philadelphia, PA 19140, USA
| | - Anastasios Lymperopoulos
- Neurohormonal Control of the Circulation Lab, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, 3200 S. University Dr., Health Professions Division (Terry) Bldg/Room 1338, Fort Lauderdale, FL 33328, USA
| | - Peter Johannes Kennel
- Division of Cardiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Nina Pollak
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - P. Christian Schulze
- Division of Cardiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes & Metabolism, NYU-Langone School of Medicine, 522 First Avenue, New York, NY 10016, USA
| |
Collapse
|
10
|
Borlaug BA, Lewis GD, McNulty SE, Semigran MJ, LeWinter M, Chen H, Lin G, Deswal A, Margulies KB, Redfield MM. Effects of sildenafil on ventricular and vascular function in heart failure with preserved ejection fraction. Circ Heart Fail 2015; 8:533-41. [PMID: 25782985 DOI: 10.1161/circheartfailure.114.001915] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/12/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Early studies showed beneficial effects of phosphodiesterase 5 inhibitors on cardiovascular function in heart failure (HF) patients, but the RELAX trial observed no improvement in exercise capacity with sildenafil treatment in subjects with HF and preserved ejection fraction. METHODS AND RESULTS A subgroup of participants in the RELAX trial (n=48) underwent comprehensive noninvasive cardiovascular assessment before and after treatment with sildenafil or placebo in a prospective ancillary study. Left ventricular contractility was assessed by peak power index and stroke work index. Systemic arterial load was assessed by arterial elastance (Ea) and right ventricular afterload by pulmonary artery systolic pressure. Endothelial function was assessed by reactive hyperemia index after upper arm cuff occlusion. Compared with placebo (n=25), sildenafil (n=23) decreased Ea (-0.29±0.28 mm Hg/mL versus +0.02±0.29, P=0.008) and tended to improve reactive hyperemia index (+0.30±0.45 versus -0.17±0.30, P=0.054). In contrast, left ventricular contractility was reduced by 11% to 16% with sildenafil compared with placebo (ΔPWR/EDV -52±70 versus +0±40 mm Hg/s, P=0.006; ΔSW/EDV +0.3±5.8 versus -6.0±5.1 mm Hg, P=0.04). Sildenafil had no effect on pulmonary artery systolic pressure. CONCLUSIONS In subjects with HF and preserved ejection fraction, sildenafil displayed opposing effects on ventricular and vascular function. We speculate that beneficial effects of phosphodiesterase 5 inhibitors in the systemic vasculature and endothelium were insufficient to improve clinical status or that the deleterious effects on left ventricular function offset any salutary vascular effects, contributing to the absence of benefit observed with sildenafil in subjects with HF and preserved ejection fraction in the RELAX trial. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00094302.
Collapse
Affiliation(s)
- Barry A Borlaug
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.).
| | - Gregory D Lewis
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Steven E McNulty
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Marc J Semigran
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Martin LeWinter
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Horng Chen
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Grace Lin
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Anita Deswal
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Kenneth B Margulies
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| | - Margaret M Redfield
- From the Department of Medicine, Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., H.C., G.L., M.M.R.); Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.J.S.); Department of Medicine, Duke Clinical Research Institute, Durham, NC (S.E.M.); Department of Medicine, Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Department of Medicine, Michael E. DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX (A.D.); and Department of Medicine, University of Pennsylvania, Translational Research Center, Philadelphia (K.B.M.)
| |
Collapse
|
11
|
Haouzi P, Chenuel B, Sonobe T. High-dose hydroxocobalamin administered after H2S exposure counteracts sulfide-poisoning-induced cardiac depression in sheep. Clin Toxicol (Phila) 2015; 53:28-36. [PMID: 25546714 DOI: 10.3109/15563650.2014.990976] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CONTEXT Severe H2S poisoning leads to death by rapid respiratory and cardiac arrest, the latter can occur within seconds or minutes in severe forms of intoxication. OBJECTIVES To determine the time course and the nature of H2S-induced cardiac arrest and the effects of high-dose hydroxocobalamin administered after the end of sulfide exposure. MATERIALS AND METHODS NaHS was infused in 16 sedated mechanically ventilated sheep to reach concentrations of H2S in the blood, which was previously found to lead to cardiac arrest within minutes following the cessation of H2S exposure. High-dose hydroxocobalamin (5 g) or saline solution was administered intravenously, 1 min after the cessation of NaHS infusion. RESULTS All animals were still alive at the cessation of H2S exposure. Three animals (18%) presented a cardiac arrest within 90 s and were unable to receive any antidote or vehicle. In the animals that survived long enough to receive either hydroxocobalamin or saline, 71% (5/7) died in the control group by cardiac arrest within 10 min. In all instances, cardiac arrest was the result of a pulseless electrical activity (PEA). In the group that received the antidote, intravenous injection of 5 g of hydroxocobalamin provoked an abrupt increase in blood pressure and blood flow; PEA was prevented in all instances. However, we could not find any evidence for a recovery in oxidative metabolism in the group receiving hydroxocobalamin, as blood lactate remained elevated and even continued to rise after 1 h, despite restored hemodynamics. This, along with an unaltered recovery of H2S kinetics, suggests that hydroxocobalamin did not act through a mechanism of H2S trapping. CONCLUSION In this sheep model, there was a high risk for cardiac arrest, by PEA, persisting up to 10 min after H2S exposure. Very high dose of hydroxocobalamin (5 g), injected very early after the cessation of H2S exposure, improved cardiac contractility and prevented PEA.
Collapse
Affiliation(s)
- Philippe Haouzi
- Division of Pulmonary and Critical Medicine, Pennsylvania State University College of Medicine , Hershey, PA , USA
| | | | | |
Collapse
|
12
|
Andreadou I, Mikros E, Ioannidis K, Sigala F, Naka K, Kostidis S, Farmakis D, Tenta R, Kavantzas N, Bibli SI, Gikas E, Skaltsounis L, Kremastinos DT, Iliodromitis EK. Oleuropein prevents doxorubicin-induced cardiomyopathy interfering with signaling molecules and cardiomyocyte metabolism. J Mol Cell Cardiol 2014; 69:4-16. [DOI: 10.1016/j.yjmcc.2014.01.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/16/2014] [Accepted: 01/19/2014] [Indexed: 11/30/2022]
|
13
|
Greene SJ, Gheorghiade M, Borlaug BA, Pieske B, Vaduganathan M, Burnett JC, Roessig L, Stasch JP, Solomon SD, Paulus WJ, Butler J. The cGMP signaling pathway as a therapeutic target in heart failure with preserved ejection fraction. J Am Heart Assoc 2013; 2:e000536. [PMID: 24334823 PMCID: PMC3886746 DOI: 10.1161/jaha.113.000536] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Stephen J Greene
- Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Chen Y, Zhang P, Li J, Xu X, Bache RJ. Inducible nitric oxide synthase inhibits oxygen consumption in collateral-dependent myocardium. Am J Physiol Heart Circ Physiol 2013; 306:H356-62. [PMID: 24322607 DOI: 10.1152/ajpheart.00308.2013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Following coronary artery occlusion growth of collateral vessels can provide an effective blood supply to the dependent myocardium. The ischemia, which results in growth of collateral vessels, recruits an inflammatory response with expression of cytokines and growth factors, upregulation of endothelial nitric oxide (NO) synthase (eNOS) in vascular endothelial cells, and expression of inducible nitric oxide synthase (iNOS) in both vessels and cardiac myocytes. Because NO is a potent collateral vessel dilator, this study examined whether NO derived from iNOS or constitutive NOS regulates myocardial blood flow (MBF) in the collateral region. Nonselective NOS inhibition with N(G)-nitro-l-arginine (LNA) caused vasoconstriction with a significant decrease in MBF to the collateral region during exercise. In contrast, the highly selective iNOS inhibitor 1400W caused a 21 ± 5% increase of MBF in the collateral region. This increase in MBF following selective iNOS blockade was proportionate to an increase in myocardial O2 consumption (MVo2). The results suggest that NO produced by iNOS inhibits MVo2 in the collateralized region, so that the increase in MBF following iNOS blockade was the result of metabolic vasodilation secondary to an increase in MVo2. Thus the coordinated expression of iNOS to restrain MVo2 and eNOS to maintain collateral vasodilation act to optimize the O2 supply-demand relationship and protect the collateralized myocardium from ischemia.
Collapse
Affiliation(s)
- Yingjie Chen
- Departments of Medicine and Integrative Biology/Physiology, University of Minnesota Medical School, Minneapolis, Minnesota; and
| | | | | | | | | |
Collapse
|
15
|
Romero-Bermejo FJ, Ruiz-Bailen M, Gil-Cebrian J, Huertos-Ranchal MJ. Sepsis-induced cardiomyopathy. Curr Cardiol Rev 2013; 7:163-83. [PMID: 22758615 PMCID: PMC3263481 DOI: 10.2174/157340311798220494] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 02/24/2011] [Accepted: 02/24/2011] [Indexed: 01/20/2023] Open
Abstract
Myocardial dysfunction is one of the main predictors of poor outcome in septic patients, with mortality rates next to 70%. During the sepsis-induced myocardial dysfunction, both ventricles can dilate and diminish its ejection fraction, having less response to fluid resuscitation and catecholamines, but typically is assumed to be reversible within 7-10 days. In the last 30 years, It´s being subject of substantial research; however no explanation of its etiopathogenesis or effective treatment have been proved yet. The aim of this manuscript is to review on the most relevant aspects of the sepsis-induced myocardial dysfunction, discuss its clinical presentation, pathophysiology, etiopathogenesis, diagnostic tools and therapeutic strategies proposed in recent years.
Collapse
Affiliation(s)
- Francisco J Romero-Bermejo
- Intensive Care Unit, Critical Care and Emergency Department, Puerto Real University Hospital, Cadiz, Spain.
| | | | | | | |
Collapse
|
16
|
Damy T, Lesault PF, Guendouz S, Eddahibi S, Tu L, Marcos E, Guellich A, Dubois-Randé JL, Teiger E, Hittinger L, Adnot S. Pulmonary hemodynamic responses to inhaled NO in chronic heart failure depend on PDE5 G(-1142)T polymorphism. Pulm Circ 2012; 1:377-82. [PMID: 22140627 PMCID: PMC3224429 DOI: 10.4103/2045-8932.87303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To evaluate the vasoconstrictor component of PH in CHF by investigating the hemodynamic response to inhaled nitric oxide (iNO) and to determine whether this response was influenced by the phosphodiesterase 5 gene (PDE5) G(1142)T polymorphism. CHF patients underwent right heart catheterization at rest and after 20 ppm of iNO and plasma cGMP and PDE5 G(1142)T polymorphism determinations. Of the 72 included CHF patients (mean age, 53±1 years; mean left ventricular ejection fraction, 29±1%; and mean pulmonary artery pressure, 25.5±1.3 mmHg), 54% had ischemic heart disease. Proportions of patients with the TT, GT, and GG genotypes were 39%, 42% and 19% respectively. Baseline hemodynamic characteristics were not significantly different across PDE5 genotype groups, although pulmonary capillary wedge pressure (PCWP) tended to be lower in the TT group (P=0.09). Baseline plasma cGMP levels were significantly lower in the TT than in the GG and GT patients. With iNO, PVR diminished in TT (-33%) but not GG (-1.6%) or GT (0%) patients (P=0.002); and PCWP increased more in TT than in GT (P<0.05) or GG (P<0.003) patients. The PDE5 G(-1142) polymorphism is therefore a major contributor to the iNO-induced PVR decrease in CHF.
Collapse
Affiliation(s)
- Thibaud Damy
- Department of Cardiology, all at AP-HP, Groupe Hospitalier Albert Chenevier-Henri Mondor, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Brunetti-Pierri N, Lamance KM, Lewis RA, Craigen WJ. 30-year follow-up of a patient with classic citrullinemia. Mol Genet Metab 2012; 106:248-50. [PMID: 22494546 DOI: 10.1016/j.ymgme.2012.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 03/16/2012] [Accepted: 03/16/2012] [Indexed: 10/28/2022]
Abstract
Citrullinemia is a urea cycle defect requiring long-term care with nutritional and pharmacological management. Despite treatment, morbidity and mortality of this disease remain high, and long-term complications include mild to profound mental retardation, seizures, and growth deficiency. We report a 31-year old woman with classic, neonatal-onset citrullinemia who developed progressive hypertrophic cardiomyopathy and cataracts, neither of which has been recognized previously as a complication of the disease or a consequence of long-term drug treatment.
Collapse
Affiliation(s)
- Nicola Brunetti-Pierri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
18
|
Reassessment of a suggested pharmacological approach to heart failure: L-arginine is only a marginal NO donor in pigs. J Cardiovasc Pharmacol 2012; 60:262-8. [PMID: 22592773 DOI: 10.1097/fjc.0b013e31825de0bf] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES L-Arginine has been tested in various cardiovascular diseases, mainly to improve endothelial function through NO production. However, as the results have been partly unpredictable, we assessed the hemodynamic, energetic and metabolic effects of L-arginine to clarify any potential benefits in postischemic left ventricular (LV) dysfunction. METHODS LV dysfunction was induced by repetitive brief coronary occlusions in 12 anesthetized, open chest pigs. L-Arginine was subsequently infused (bolus 400 mg·kg and continuously for 1 hour, 250 mg·kg·h). Hemodynamic parameters, metabolites of L-arginine and myocardial energetics were assessed sequentially. RESULTS L-Arginine infusions caused a substantial rise in plasma L-arginine (3474 ± 358 μmole·L) accompanied by a 2-fold increase in plasma L-citrulline. No significant alterations in vascular resistance or LV contractility were observed from L-arginine. Mean arterial pressure dropped from 78 ± 11 to 72 ± 10 mm Hg (P = 0.019) and 70 ± 8 mm Hg (P = 0.003) after bolus and infusions, respectively. Myocardial oxygen consumption was unaltered, and myocardial creatine content was not increased after 90 minutes of L-arginine infusion. CONCLUSION L-Arginine infusion did not influence the energetic cost of myocardial contractility, and only minor hemodynamic changes were observed despite a demonstrable turnover of L-arginine. These findings question the use of L-arginine to promote therapeutic NO formation in the acute setting.
Collapse
|
19
|
Martinelli NC, Santos KG, Biolo A, La Porta VL, Cohen CR, Silvello D, Andrades ME, Clausell N, Rohde LE. Polymorphisms of endothelial nitric oxide synthase gene in systolic heart failure: An haplotype analysis. Nitric Oxide 2012; 26:141-7. [PMID: 22290017 DOI: 10.1016/j.niox.2012.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 12/14/2011] [Accepted: 01/06/2012] [Indexed: 10/14/2022]
|
20
|
Bishu K, Hamdani N, Mohammed SF, Kruger M, Ohtani T, Ogut O, Brozovich FV, Burnett JC, Linke WA, Redfield MM. Sildenafil and B-type natriuretic peptide acutely phosphorylate titin and improve diastolic distensibility in vivo. Circulation 2011; 124:2882-91. [PMID: 22144574 DOI: 10.1161/circulationaha.111.048520] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In vitro studies suggest that phosphorylation of titin reduces myocyte/myofiber stiffness. Titin can be phosphorylated by cGMP-activated protein kinase. Intracellular cGMP production is stimulated by B-type natriuretic peptide (BNP) and degraded by phosphodiesterases, including phosphodiesterase-5A. We hypothesized that a phosphodiesterase-5A inhibitor (sildenafil) alone or in combination with BNP would increase left ventricular diastolic distensibility by phosphorylating titin. METHODS AND RESULTS Eight elderly dogs with experimental hypertension and 4 young normal dogs underwent measurement of the end-diastolic pressure-volume relationship during caval occlusion at baseline, after sildenafil, and BNP infusion. To assess diastolic distensibility independently of load/extrinsic forces, the end-diastolic volume at a common end-diastolic pressure on the sequential end-diastolic pressure-volume relationships was measured (left ventricular capacitance). In a separate group of dogs (n=7 old hypertensive and 7 young normal), serial full-thickness left ventricular biopsies were harvested from the beating heart during identical infusions to measure myofilament protein phosphorylation. Plasma cGMP increased with sildenafil and further with BNP (7.31±2.37 to 26.9±10.3 to 70.3±8.1 pmol/mL; P<0.001). Left ventricular diastolic capacitance increased with sildenafil and further with BNP (51.4±16.9 to 53.7±16.8 to 60.0±19.4 mL; P<0.001). Changes were similar in old hypertensive and young normal dogs. There were no effects on phosphorylation of troponin I, troponin T, phospholamban, or myosin light chain-1 or -2. Titin phosphorylation increased with sildenafil and BNP, whereas titin-based cardiomyocyte stiffness decreased. CONCLUSION Short-term cGMP-enhancing treatment with sildenafil and BNP improves left ventricular diastolic distensibility in vivo, in part by phosphorylating titin.
Collapse
Affiliation(s)
- Kalkidan Bishu
- Mayo Clinic Cardiovascular Division, 200 First St SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Effects of phosphodiesterase type 5 inhibitors on endothelial function and cardiovascular autonomic nerve function in men. JOURNAL OF MENS HEALTH 2011. [DOI: 10.1016/j.jomh.2011.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
23
|
Schulman IH, Hare JM. Regulation of cardiovascular cellular processes by S-nitrosylation. Biochim Biophys Acta Gen Subj 2011; 1820:752-62. [PMID: 21536106 DOI: 10.1016/j.bbagen.2011.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 04/07/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Nitric oxide (NO), a highly versatile signaling molecule, exerts a broad range of regulatory influences in the cardiovascular system that extends from vasodilation to myocardial contractility, angiogenesis, inflammation, and energy metabolism. Considerable attention has been paid to deciphering the mechanisms for such diversity in signaling. S-nitrosylation of cysteine thiols is a major signaling pathway through which NO exerts its actions. An emerging concept of NO pathophysiology is that the interplay between NO and reactive oxygen species (ROS), the nitroso/redox balance, is an important regulator of cardiovascular homeostasis. SCOPE OF REVIEW ROS react with NO, limit its bioavailability, and compete with NO for binding to the same thiol in effector molecules. The interplay between NO and ROS appears to be tightly regulated and spatially confined based on the co-localization of specific NO synthase (NOS) isoforms and oxidative enzymes in unique subcellular compartments. NOS isoforms are also in close contact with denitrosylases, leading to crucial regulation of S-nitrosylation. MAJOR CONCLUSIONS Nitroso/redox balance is an emerging regulatory pathway for multiple cells and tissues, including the cardiovascular system. Studies using relevant knockout models, isoform specific NOS inhibitors, and both in vitro and in vivo methods have provided novel insights into NO- and ROS-based signaling interactions responsible for numerous cardiovascular disorders. GENERAL SIGNIFICANCE An integrated view of the role of nitroso/redox balance in cardiovascular pathophysiology has significant therapeutic implications. This is highlighted by human studies where pharmacologic manipulation of oxidative and nitrosative pathways exerted salutary effects in patients with advanced heart failure. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation.
Collapse
Affiliation(s)
- Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | | |
Collapse
|
24
|
The Renin-Angiotensin System in the Development of Salt-Sensitive Hypertension in Animal Models and Humans. Pharmaceuticals (Basel) 2010; 3:940-960. [PMID: 27713283 PMCID: PMC4034015 DOI: 10.3390/ph3040940] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 02/25/2010] [Accepted: 03/08/2010] [Indexed: 02/07/2023] Open
Abstract
Hypertension is still one of the major causes of death from cardiovascular failure. Increased salt intake may aggravate the rise in blood pressure and the development of consequential damage of the heart, the vessels and other organs. The general necessity of restricted salt intake regardless of blood pressure or salt sensitivity has been a matter of debate over the past decades. This review summarizes the main pathogenic mechanisms of hypertension and salt sensitivity in rat models, particularly in the spontaneously hypertensive rat (SHR), and in patients with essential hypertension (EH). Although SHRs are commonly considered to be salt-resistant, there is much evidence that salt loading may deteriorate blood pressure and cardiovascular function even in these animals. Similarly, EH is not a homogenous disorder - some patients, but not all, exhibit pronounced salt sensitivity. The renin-angiotensin system (RAS) plays a key role in the regulation of blood pressure and salt and fluid homeostasis and thus is one of the main targets of antihypertensive therapy. This review focuses on the contribution of the RAS to the pathogenesis of salt-sensitive hypertension in SHRs and patients with EH.
Collapse
|
25
|
Mattar EH, Haffor ASA. Effect of dobutamine and hyperoxia on free radicals production in relation to the ultrastructural alterations in the endothelial of myocardial capillary in rats, Rattus norvigicus. Ultrastruct Pathol 2010; 33:209-15. [PMID: 19895293 DOI: 10.3109/01913120903275626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hyperoxia has been widely used as model for oxidative stress. Free radicals (FR), suproxide anion (*O(-)(2)) and nitric oxide anion (*NO(-)), are highly toxic and have detrimental effects on nitroso-redox balance in the myocyte. Myocardium is rich with beta-adrenergic receptors and endothelial is the site of NO production. The authors hypothesized that graded doses of dobutamine result in hyperkinetic state, which shifts the nitroso-redox balance toward the buildup of reactive species in dose-dependent excess. The purpose of the present study was to investigate free radicals production and coronary endothelial cell pathological changes following increasing length of breathing oxygen (100% O(2)) and progressive doses of dobutamine. Thirty-five adult male rats, matched with age and body weight, were randomly assigned to 7 groups. The first group served as control (C) and the 2nd, 3rd, and 4th groups were exposed to hyperoxia (100% O(2) breathing) for 24,48, and 72 h, whereas the 5th, 6th, and 7th groups were injected dobutamine 10,20, and 30 microg kg(-1), respectively. Following the treatment condition for each group, animals were sacrificed and heart tissues were divided randomly into two parts. The first part was processed for the ultrastructure, using transmission electron microscope (TEM), and the second was homogenized for FR determination. TEM examination showed that O2 breathing for 24 h resulted in hypertrophy and proliferation of endothelial cells lining the coronary capillary, which was lodged by lymphoid cells. Distended and irregular contour of endothelium, enlarged nucleus, protrusion membrane, as well as pinocytotic vesicles were also observed. Free radicals (FR) production at all levels of hyperoxia exposures and dobutamine injections were significantly (p < .05) higher than control group. In addition, dobutamine induced higher relative FR production, as compared with hyperoxia, implying more severe myocyte injury. Based on the results of the present study, it can be concluded that O2 breathing for 24 h or higher resulted in variety of pathological changes of the endothelium of coronary capillary that were induced by the buildup of oxidants by-products. Because dobutamine caused relative higher in FR production levels, as compared with hyperoxia levels, throughout this implied it aggravated the myocyte capillaries' endothelium more heavily, which could have resulted in more intense ultrastructural deteriorations.
Collapse
Affiliation(s)
- Essam H Mattar
- Department of Radiological Sciences, King Saud University, Kharj, Kingdom of Saudi Arabia
| | | |
Collapse
|
26
|
Abstract
β-blockers are among the most widely used drugs in the prevention and treatment of cardiovascular disease, although they are associated with increased peripheral resistance. Third-generation β-blockers avoid this adverse effect by inducing vasodilation through different mechanisms. In particular, nebivolol, a highly selective blocker of β1-adrenergic receptors, is the only β-blocker known to induce vascular production of nitric oxide, the main endothelial vasodilator. The specific mechanism of nebivolol is particularly relevant in hypertension, where nitric oxide dysfunction occurs. Indeed, nebivolol is able to reverse endothelial dysfunction. Nebivolol induces nitric oxide production via activation of β3-adrenergic receptors, which can explain the good metabolic profile observed after treatment with this drug. Moreover, nebivolol can also stimulate the β3-adrenergic receptor-mediated production of nitric oxide in the heart, and this stimulation can result in a greater protection against heart failure. In conclusion, nebivolol has a unique profile among antihypertensive drugs, adding to a very high selectivity against β1 adrenergic receptors, and an agonist action on β3 receptors and nitric oxide (NO), which has led to clinically significant improvements in hypertensive patients.
Collapse
Affiliation(s)
- Angelo Maffei
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Giuseppe Lembo
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS), Italy,
| |
Collapse
|
27
|
Brooks WW, Conrad CH, Robinson KG, Colucci WS, Bing OHL. L-arginine fails to prevent ventricular remodeling and heart failure in the spontaneously hypertensive rat. Am J Hypertens 2009; 22:228-34. [PMID: 19057517 DOI: 10.1038/ajh.2008.334] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The effects of long-term oral administration of L-arginine, a substrate for nitric oxide (NO) production, on left ventricular (LV) remodeling, myocardial function and the prevention of heart failure (HF) was compared to the angiotensin-converting enzyme (ACE) inhibitor captopril in a rat model of hypertensive HF (aged spontaneously hypertensive rat (SHR)). METHODS SHRs and age-matched normotensive Wistar-Kyoto (WKY) rats were assigned to either no treatment, treatment with L-arginine (7.5 g/l in drinking water) or captopril (1 g/l in drinking water) beginning at 14 months of age, a time when SHRs exhibit stable compensated hypertrophy with no hemodynamic impairment; animals were studied at 23 months of age or at the time of HF. RESULTS In untreated SHR, relative to WKY, there was significant LV hypertrophy, myocardial fibrosis, and isolated LV muscle performance and response to isoproterenol (ISO) were depressed; and, 7 of 10 SHRs developed HF. Captopril administration to six SHRs attenuated hypertrophy and prevented impaired inotropic responsiveness to ISO, contractile dysfunction, fibrosis, increased passive stiffness, and HF. In contrast, L-arginine administration to SHR increased LV hypertrophy and myocardial fibrosis while cardiac performance was depressed; and 7 of 9 SHRs developed HF. In WKY, L-arginine treatment but not captopril resulted in increased LV weight and the contractile response to ISO was blunted. Neither L-arginine nor captopril treatment of WKY changed fibrosis and HF did not occur. CONCLUSION These data demonstrate that in contrast to captopril, long-term treatment with L-arginine exacerbates age-related cardiac hypertrophy, fibrosis, and did not prevent contractile dysfunction or the development of HF in aging SHR.
Collapse
|
28
|
Hu YF, Chen YC, Cheng CC, Higa S, Chen YJ, Chen SA. Fluvastatin reduces pulmonary vein spontaneous activity through nitric oxide pathway. J Cardiovasc Electrophysiol 2008; 20:200-6. [PMID: 18775044 DOI: 10.1111/j.1540-8167.2008.01281.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Pulmonary veins (PVs) are the most important focus for the generation of atrial fibrillation. The HMG-CoA reductase inhibitors (statins) can reduce the occurrence of atrial fibrillation. The purposes of this study were to evaluate whether statins may inhibit the PV arrhythmogenic activity to prevent atrial arrhythmias from PVs and to investigate the link between fluvastatin, nitric oxide synthase (NOS) activity, mechanical activity, and electrical activity. METHODS Conventional microelectrodes and Western blot were used to record the electrical activity, diastolic tension, contractility and expression of Akt, endothelial nitric oxide synthase (eNOS), neuronal nitric oxide synthase (nNOS), and phosphorylated Akt and eNOS before and after the administration of fluvastatin in rabbit PVs or atria. RESULTS Fluvastatin decreased the PV spontaneous activity, diastolic tension, and contractility, but did not change the action potential duration or resting membrane potential. The effects of fluvastatin on the PV firing rate and diastolic tension were attenuated in the presence of L-NAME (100 microM), wortmannin (100 nM), and ODQ (3 microM). Fluvastatin (1 muM) increased the phosphorylated Akt and eNOS, but did not change the total Akt or eNOS in the PVs and atria. In contrast, fluvastatin (1 microM) decreased the total nNOS in the PVs and atria. CONCLUSIONS AND IMPLICATIONS Fluvastatin produced nitric oxide through the PI3kinase/Akt pathway, thus reducing the PV vascular diastolic tension and PV spontaneous activity. These results may contribute to the beneficial effects of statins.
Collapse
Affiliation(s)
- Yu-Feng Hu
- Division of Cardiology and Cardiovascular Research Center, Veterans General Hospital-Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
29
|
Spániková A, Simoncíková P, Ravingerová T, Pechánová O, Barancík M. The effect of chronic nitric oxide synthases inhibition on regulatory proteins in rat hearts. Mol Cell Biochem 2008; 312:113-20. [PMID: 18327702 DOI: 10.1007/s11010-008-9726-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Accepted: 02/25/2008] [Indexed: 11/27/2022]
Abstract
The aim of the present study was to characterize the effects of chronic nitric oxide synthase (NOS) inhibition on the alterations of regulatory myocardial proteins of intracellular signaling pathways (mitogen-activated protein kinase (MAPK) and Akt kinase cascades) and matrix metalloproteinases (MMP). Chronic NO deficiency (NOD) was induced by NG-nitro-L-arginine methyl ester (L-NAME, 40 mg/kg/day, 4 weeks). Protein levels and activation of protein kinases were determined using specific antibodies, activities of MMP were analyzed by zymography in gels containing gelatin as a substrate. The development of NOD was associated with decreased activation of endothelial NOS (eNOS) and down-regulation of protein level of inducible NOS (iNOS). Investigation of kinase pathways revealed that the activation of extracellular signal-regulated kinases (ERK) and the levels of upstream activators of ERK (aFGF, H-Ras) were decreased after L-NAME treatment. Western blot analysis revealed that chronic application of L-NAME also decreased the activation of Akt kinase as compared with control hearts. Study of MMPs showed that in L-NAME-treated rat hearts activities of tissue MMP-2 were decreased. It is concluded that development of NOD resulted in inhibition of ERK and Akt kinase pathways and these changes suggest the involvement of these cascades in responses of myocardium to NOD. The results also point to the possible relationship between ERK and Akt kinase pathways and activation of eNOS and/or MMP-2.
Collapse
Affiliation(s)
- Anna Spániková
- Institute for Heart Research, Centre of Excellence for Cardiovascular Research, Slovak Academy of Sciences, Dúbravská cesta 9, P.O. Box 104, 840 05 Bratislava, Slovak Republic
| | | | | | | | | |
Collapse
|
30
|
Cariou A, Pinsky MR, Monchi M, Laurent I, Vinsonneau C, Chiche JD, Charpentier J, Dhainaut JF. Is myocardial adrenergic responsiveness depressed in human septic shock? Intensive Care Med 2008; 34:917-22. [PMID: 18259725 DOI: 10.1007/s00134-008-1022-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 01/16/2008] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess left ventricular (LV) contractile function and adrenergic responsiveness in septic patients. METHODS We used echocardiographically defined fractional area of contraction (FAC), and LV area to end-systolic arterial pressure estimates of end-systolic elastance (E'es) and its change in response to dobutamine (5 microg/kg/min) in 10 subjects in septic shock admitted to an intensive care unit of an academic medical center. Subjects were studied on admission and again at both 5 days and 8-10 days after admission. RESULTS Three of the 10 subjects died as a result of their acute process, while the others were discharged from hospital. Nine out of 10 subjects required intravenous vasopressor therapy on day 1, while only 1 of 9 subjects required vasopressor support at day 5. LV end-diastolic area (EDA) increased from day 1 to day 5 and days 8-10 (p<0.05), but neither FAC nor E'es was altered by time (EDA 15.7+/-5.8, 21.4+/-5.1, and 19.4+/-5.6 cm2; FAC 0.46+/-0.19, 0.50+/-0.20, and 0.48+/-0.15%; E'es 21.6+/-12.6, 23.2+/-8.5, and 19.2+/-6.3 mmHg/cm2, mean+/-SD, for days 1, 5 and 8-10 respectively). Although dobutamine did not alter E'es on day 1 or day 5, E'es increased in all of the 5 subjects studied on days 8-10 (p<0.05). CONCLUSIONS Adrenergic hyporesponsiveness is present in septic shock and persists for at least 5 days into recovery, resolving by days 8-10 in survivors.
Collapse
Affiliation(s)
- Alain Cariou
- Cochin-Saint Vincent de Paul Hospital and Paris Descartes University, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Inducible nitric oxide synthase depresses cardiac contractile function in Zucker diabetic fatty rats. Eur J Pharmacol 2008; 579:253-9. [DOI: 10.1016/j.ejphar.2007.09.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2007] [Revised: 09/21/2007] [Accepted: 09/27/2007] [Indexed: 10/22/2022]
|
32
|
Ohta S, Shinke T, Hata K, Takaoka H, Shite J, Kijima Y, Murata T, Yoshikawa R, Masai H, Hirata KI, Yokoyama M. Inhibition of endogenous nitric oxide synthase augments contractile response to adenylyl cyclase stimulation without altering mechanical efficiency in patients with idiopathic dilated cardiomyopathy. Circ J 2007; 71:1268-73. [PMID: 17652893 DOI: 10.1253/circj.71.1268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Increased nitric oxide (NO) in the failing heart attenuates the myocardial contractile response to beta-adrenergic receptor stimulation. However, the physiological effects of NO on the beta-adrenergic post-receptor signaling system are unknown. The objective of the present study was to examine the effects of cardiac NO synthase (NOS) inhibition on left ventricular (LV) hemodynamics and mechanoenergetics in response to adenylyl cyclase stimulation in human heart failure. METHODS AND RESULTS The study group comprised 13 patients with heart failure because of idiopathic cardiomyopathy (IDC). Emax was examined as an index of LV contractility, LV external work (EW), pressure-volume area (PVA), myocardial oxygen consumption (MVO2), and mechanical efficiency (EW/MVO2) with the use of conductance and coronary sinus thermodilution catheters before and during colforsin daropate infusion, and during concurrent infusion of colforsin daropate with the NOS inhibitor, NG-monomethyl-L-arginine (L-NMMA; 200 micromol). Colforsin daropate increased Emax by 53% and EW by 18%, and reduced PVA by 14%, without altering MVO2 or mechanical efficiency. The combination of colforsin daropate with L-NMMA further increased Emax by 26% and reduced PVA by 9%, without altering MVO2 or mechanical efficiency. CONCLUSIONS These findings suggest endogenous NO may modulate beta-adrenergic post-receptor pathways and preserve myocardial efficiency in patients with IDC.
Collapse
Affiliation(s)
- Soichiro Ohta
- Division of Cardiovascular and Respiratory Medicine, Department of Internal Medicine, Kobe University, Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
OBJECTIVES To review mechanisms underlying sepsis-induced cardiac dysfunction in general and intrinsic myocardial depression in particular. DATA SOURCE MEDLINE database. DATA SYNTHESIS Myocardial depression is a well-recognized manifestation of organ dysfunction in sepsis. Due to the lack of a generally accepted definition and the absence of large epidemiologic studies, its frequency is uncertain. Echocardiographic studies suggest that 40% to 50% of patients with prolonged septic shock develop myocardial depression, as defined by a reduced ejection fraction. Sepsis-related changes in circulating volume and vessel tone inevitably affect cardiac performance. Although the coronary circulation during sepsis is maintained or even increased, alterations in the microcirculation are likely. Mitochondrial dysfunction, another feature of sepsis-induced organ dysfunction, will also place the cardiomyocytes at risk of adenosine triphosphate depletion. However, clinical studies have demonstrated that myocardial cell death is rare and that cardiac function is fully reversible in survivors. Hence, functional rather than structural changes seem to be responsible for intrinsic myocardial depression during sepsis. The underlying mechanisms include down-regulation of beta-adrenergic receptors, depressed postreceptor signaling pathways, impaired calcium liberation from the sarcoplasmic reticulum, and impaired electromechanical coupling at the myofibrillar level. Most, if not all, of these changes are regulated by cytokines and nitric oxide. CONCLUSIONS Integrative studies are needed to distinguish the hierarchy of the various mechanisms underlying septic cardiac dysfunction. As many of these changes are related to severe inflammation and not to infection per se, a better understanding of septic myocardial dysfunction may be usefully extended to other systemic inflammatory conditions encountered in the critically ill. Myocardial depression may be arguably viewed as an adaptive event by reducing energy expenditure in a situation when energy generation is limited, thereby preventing activation of cell death pathways and allowing the potential for full functional recovery.
Collapse
Affiliation(s)
- Alain Rudiger
- Bloomsbury Institute of Intensive Care Medicine, Wolfson Institute for Biomedical Research and Department of Medicine, University College London, UK
| | | |
Collapse
|
34
|
Kittleson MM, Lowenstein CJ, Hare JM. Novel pathogenetic mechanisms in myocarditis: nitric oxide signaling. Heart Fail Clin 2007; 1:345-61. [PMID: 17386859 DOI: 10.1016/j.hfc.2005.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
35
|
Kumar A, Paladugu B, Mensing J, Kumar A, Parrillo JE. Nitric oxide-dependent and -independent mechanisms are involved in TNF-alpha -induced depression of cardiac myocyte contractility. Am J Physiol Regul Integr Comp Physiol 2007; 292:R1900-6. [PMID: 17234961 DOI: 10.1152/ajpregu.00146.2006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Previous studies have demonstrated the presence of myocardial depression in clinical and experimental septic shock. This response is mediated, in part, through circulating TNF-alpha-induced, nitric oxide-dependent, depression of basal myocyte contractility. Other mechanisms of early myocardial dysfunction involving decreased response to adrenergic stimulation may exist. This study evaluated the presence and nitric oxide dependence of impaired adrenergic response to TNF-alpha in in vitro cardiac myocytes. The contraction of electrically paced neonatal rat cardiac myocytes in tissue culture was quantified using a closed-loop video tracking system. TNF-alpha induced depression of baseline contractility over the first 20 min of cardiac myocyte exposure. This effect was blocked by N-methyl-arginine (NMA), a nitric oxide synthase inhibitor, in all studies. Contractile and cAMP response to increasing concentrations of isoproterenol was deficient in cardiac myocytes exposed to TNF-alpha regardless of the presence of NMA. In contrast, increasing concentrations of forskolin (a direct stimulant of adenylate cyclase) and dibutyryl cAMP (a metabolically active membrane-soluble analog of cAMP) completely reversed TNF-alpha-mediated depression, though only in the presence of NMA. Forskolin-stimulated cAMP generation remained intact regardless of NMA. Increasing concentrations of exogenous calcium chloride, unlike other inotropic agents, corrected TNF-alpha-mediated defects of contractility independent of the presence of NMA. These data suggest that TNF-alpha exposure is associated with a second nitric oxide-independent but calcium-dependent early depressant mechanism that is manifested by reduced contractile and cAMP response to beta-adrenergic stimulation.
Collapse
Affiliation(s)
- Anand Kumar
- Section of Critical Care Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada.
| | | | | | | | | |
Collapse
|
36
|
Abstract
Patients with heart failure (HF) due to left ventricular (LV) systolic dysfunction have abnormal endothelium-dependent, nitric oxide-cyclic guanosine monophosphate-mediated vasodilation in the pulmonary and skeletal muscle vasculature. Therefore, inhibition of type 5 phosphodiesterase (PDE5), the principle enzyme responsible for cyclic guanosine monophosphate catabolism in the lungs and skeletal muscle, has been targeted in an effort to counteract vasoconstriction that contributes to increased right and LV afterload in HF. The efficacy of PDE5 inhibition in the treatment of pulmonary arterial hypertension has led to the investigation of its potential utility in the treatment of HF patients with secondary pulmonary hypertension. Moreover, recent preclinical studies suggest direct myocardial effects of PDE5 inhibition that may counteract beta-adrenergic, hypertrophic, and pro-apoptotic signaling, three critical pathways in the development of LV dysfunction. This review outlines both the underlying rationale and the results of initial studies of the therapeutic effects of PDE5 inhibition in HF.
Collapse
Affiliation(s)
- Gregory D Lewis
- Cardiology Division, Bigelow 800, MassachusettsGeneral Hospital, 55 Fruit St., Boston, MA 02114, USA
| | | |
Collapse
|
37
|
Donzelli S, Espey MG, Thomas DD, Mancardi D, Tocchetti CG, Ridnour LA, Paolocci N, King SB, Miranda KM, Lazzarino G, Fukuto JM, Wink DA. Discriminating formation of HNO from other reactive nitrogen oxide species. Free Radic Biol Med 2006; 40:1056-66. [PMID: 16540401 DOI: 10.1016/j.freeradbiomed.2005.10.058] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Revised: 10/20/2005] [Accepted: 10/25/2005] [Indexed: 02/07/2023]
Abstract
Nitroxyl (HNO) exhibits unique pharmacological properties that often oppose those of nitric oxide (NO), in part due to differences in reactivity toward thiols. Prior investigations suggested that the end products arising from the association of HNO with thiols were condition-dependent, but were inconclusive as to product identity. We therefore used HPLC techniques to examine the chemistry of HNO with glutathione (GSH) in detail. Under biological conditions, exposure to HNO donors converted GSH to both the sulfinamide [GSONH2] and the oxidized thiol (GSSG). Higher thiol concentrations generally favored a higher GSSG ratio, suggesting that the products resulted from competitive consumption of a single intermediate (GSNHOH). Formation of GSONH2 was not observed with other nitrogen oxides (NO, N2O3, NO2, or ONOO(-)),indicating that it is a unique product of the reaction of HNO with thiols. The HPLC assay was able to detect submicromolar concentrations of GSONH2. Detection of GSONH2 was then used as a marker for HNO production from several proposed biological pathways, including thiol-mediated decomposition of S-nitrosothiols and peroxidase-driven oxidation of hydroxylamine (an end product of the reaction between GSH and HNO) and NG-hydroxy-l-arginine (an NO synthase intermediate). These data indicate that free HNO can be biosynthesized and thus may function as an endogenous signaling agent that is regulated by GSH content.
Collapse
Affiliation(s)
- Sonia Donzelli
- Tumor Biology Section, Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The availability of selective inhibitors of the cyclic guanosine monophosphate (cGMP)-specific type 5 phosphodiesterase (PDE5) has created increasing interest in unlocking the therapeutic potential of PDE5 inhibition in cardiovascular diseases that are marked by dysfunction of nitric oxide (NO)-cGMP signaling. Pulmonary arterial hypertension (PAH) and heart failure (HF) are characterized by pulmonary arterial vasoconstriction that is thought to be caused by relative deficiencies of vasodilators such as NO and exaggerated production of vasoconstrictors such as endothelin. PDE5 is abundant in the pulmonary vasculature where it catabolizes cGMP, the second messenger of NO. Inhibition of PDE5 has been shown to lower pulmonary vascular resistance in PAH and HF by augmenting local cGMP. This review outlines the therapeutic potential of PDE5 inhibition for the treatment of PAH and HF.
Collapse
Affiliation(s)
- Gregory D Lewis
- Cardiology Division, Bigelow 800, Massachusetts General Hospital, Fruit Street, Boston, MA 02114, USA
| | | |
Collapse
|
39
|
Mak S, Overgaard CB, Newton GE. Effect of vitamin C and L-NMMA on the inotropic response to dobutamine in patients with heart failure. Am J Physiol Heart Circ Physiol 2005; 289:H2424-8. [PMID: 16040714 DOI: 10.1152/ajpheart.00453.2005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The positive effect of vitamin C on left ventricular (LV) inotropic responses to dobutamine, observed in patients with preserved LV function, is lost in heart failure (HF). We tested the hypothesis that in HF, endogenous nitric oxide (NO) opposes the positive effect of vitamin C on adrenergically stimulated contractility by examining the effects of vitamin C on dobutamine responses during NO synthase inhibition. In 11 HF patients, a micromanometer-tipped catheter was inserted into the LV and an infusion catheter was positioned in the left main coronary artery. The peak positive rate of change of LV pressure (LV +dP/dt) was measured in response to intravenous dobutamine (Dob-1). After recontrol, intracoronary N(G)-monomethyl-L-arginine (l-NMMA) was infused before reinfusion of dobutamine (L-NMMA + Dob-2). Finally, intracoronary vitamin C was infused in addition to intracoronary L-NMMA and dobutamine (L-NMMA + Dob-2 + vitamin C). Intracoronary L-NMMA alone had no effect on LV +dP/dt. After a stable inotropic response to intracoronary L-NMMA and dobutamine was established, the addition of intracoronary vitamin C resulted in a modest but significant increase in LV +dP/dt. The change in LV +dP/dt in response to dobutamine alone was 25 +/- 5%, with intracoronary L-NMMA, 27 +/- 6%, and with intracoronary L-NMMA plus vitamin C, 37 +/- 5% (P < 0.05 vs. Dob-1 and L-NMMA + Dob-2). These findings demonstrate that an interaction between endogenous NO and redox environment exists and exerts some influence on stimulated contractility in HF.
Collapse
Affiliation(s)
- Susanna Mak
- Cardiovascular Clinical Research Laboratory, Div. of Cardiology, Mount Sinai Hospital, 600 University Ave., Rm. 1543, Toronto, Ontario, Canada M5G 1X5
| | | | | |
Collapse
|
40
|
Athavale K, Claure N, D'Ugard C, Everett R, Swaminathan S, Bancalari E. Acute effects of inhaled nitric oxide on pulmonary and cardiac function in preterm infants with evolving bronchopulmonary dysplasia. J Perinatol 2004; 24:769-74. [PMID: 15496967 DOI: 10.1038/sj.jp.7211216] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Inhaled nitric oxide (iNO) reduces pulmonary vascular resistance by preferential vasodilation in ventilated lung units. In experimental animals, iNO also reduces airway resistance by smooth muscle relaxation. Hence, there may be a therapeutic role for iNO in evolving bronchopulmonary dysplasia (BPD). OBJECTIVE To evaluate the acute effects of low-dose iNO on lung mechanics, ventilation distribution, oxygenation, and cardiac function in preterm infants with evolving BPD. METHODS Measurements of lung compliance (C(L)), airway resistance (R(L)), ventilation-distribution (N(2) clearance in multiple-breath washout), oxygenation (SpO(2)), left ventricular ejection fraction (LVEF) and right ventricular shortening fraction were obtained before and during 2 hours of iNO (10 ppm) in a group of ventilated preterm infants with evolving BPD. RESULTS A total of 13 preterm infants with (mean+/-SD) BW: 663.8+/-116 g, GA: 24.9+/-1.2 weeks, age: 32+/-14 days, mean airway pressure: 6.7+/-0.9 cmH(2)O and fraction of inspired oxygen: 0.35+/-0.06 were studied. iNO did not affect C(L), R(L) or N(2) clearance. There was a small increase in LVEF. Mean SpO(2) remained unchanged, but the duration of spontaneous hypoxemic episodes increased during iNO. CONCLUSION Low-dose iNO had no acute effects on lung function, cardiac function and oxygenation in evolving BPD.
Collapse
Affiliation(s)
- Kamlesh Athavale
- Department of Pediatrics, University of Miami School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | |
Collapse
|
41
|
Ohta T, Hasebe N, Tsuji S, Izawa K, Jin YT, Kido S, Natori S, Sato M, Kikuchi K. Unequal effects of renin-angiotensin system inhibitors in acute cardiac dysfunction induced by isoproterenol. Am J Physiol Heart Circ Physiol 2004; 287:H2914-21. [PMID: 15297251 DOI: 10.1152/ajpheart.00221.2004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several clinical trials have demonstrated that angiotensin-converting enzyme inhibitor (ACEI) and angiotensin II type 1 receptor blocker (ARB) are equally effective in the treatment of chronic heart failure. However, this has not been confirmed for acute cardiac dysfunction. We examined whether ACEI or ARB prevents isoproterenol-induced acute left ventricular (LV) dysfunction in dogs. LV dysfunction induced by a large dose of isoproterenol (1 microg.kg(-1).min(-1), 3-h infusion) was compared in dogs treated with ACEI (temocaprilat) or ARB (olmesartan). Atrial pacing induced a constant heart rate and use of adjustable aortic banding provided a nearly constant afterload. LV systolic function (LV dP/dt, fractional shortening, and ejection fraction) and diastolic function (tau and LV end-diastolic pressure) were significantly deteriorated after isoproterenol infusion. The LV dysfunction was almost totally prevented by ARB but was only partially prevented by ACEI. The partial effect of ACEI was complemented by cotreatment with HOE-140, a bradykinin B2 receptor antagonist. At baseline, the response to low doses of isoproterenol was significantly attenuated by ACEI but not by ARB, and the ACEI-induced attenuation was totally abolished by cotreatment with HOE-140. The response to isoproterenol was significantly attenuated after 3 h of excess isoproterenol loading, and it was almost completely preserved by ARB but not by ACEI. In conclusion, acute LV dysfunction and beta-adrenergic desensitization induced by excess isoproterenol administration were almost totally prevented by ARB but only partially prevented by ACEI. These differences were attributable at least in part to bradykinin pathways activated by ACEI administration in acute LV dysfunction.
Collapse
Affiliation(s)
- Takafumi Ohta
- First Dept. of Internal Medicine, Asahikawa Medical College, 2-1-1 Midorigaoka higashi, Asahikawa, Hokkaido 078-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ostrom RS, Bundey RA, Insel PA. Nitric oxide inhibition of adenylyl cyclase type 6 activity is dependent upon lipid rafts and caveolin signaling complexes. J Biol Chem 2004; 279:19846-53. [PMID: 15007069 DOI: 10.1074/jbc.m313440200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Several cell types, including cardiac myocytes and vascular endothelial cells, produce nitric oxide (NO) via both constitutive and inducible isoforms of NO synthase. NO attenuates cardiac contractility and contributes to contractile dysfunction in heart failure, although the precise molecular mechanisms for these effects are poorly defined. Adenylyl cyclase (AC) isoforms type 5 and 6, which are preferentially expressed in cardiac myocytes, may be inhibited via a direct nitrosylation by NO. Because endothelial NO synthase (eNOS and NOS3), beta-adrenergic (betaAR) receptors, and AC6 all can localize in lipid raft/caveolin-rich microdomains, we sought to understand the role of lipid rafts in organizing components of betaAR-G(s)-AC signal transduction together with eNOS. Using neonatal rat cardiac myocytes, we found that disruption of lipid rafts with beta-cyclodextrin inhibited forskolin-stimulated AC activity and cAMP production, eliminated caveolin-3-eNOS interaction, and increased NO production. betaAR- and G(s)-mediated activation of AC activity were inhibited by beta-cyclodextrin treatment, but prostanoid receptor-stimulated AC activity, which appears to occur outside caveolin-rich microdomains, was unaffected unless eNOS was overexpressed and lipid rafts were disrupted. An NO donor, SNAP, inhibited basal and forskolin-stimulated cAMP production in both native cardiac myocytes and cardiac myocytes and pulmonary artery endothelial cells engineered to overexpress AC6. These effects of SNAP were independent of guanylyl cyclase activity and were mimicked by overexpression of eNOS. The juxtaposition of eNOS with betaAR and AC types 5 and 6 results in selective regulation of betaAR by eNOS activity in lipid raft domains over other G(s)-coupled receptors localized in nonraft domains. Thus co-localization of multiple signaling components in lipid rafts provides key spatial regulation of AC activity.
Collapse
Affiliation(s)
- Rennolds S Ostrom
- Department of Pharmacology, and Vascular Biology Center of Excellence, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | |
Collapse
|
43
|
Mak S, Newton GE. Redox modulation of the inotropic response to dobutamine is impaired in patients with heart failure. Am J Physiol Heart Circ Physiol 2003; 286:H789-95. [PMID: 14551049 DOI: 10.1152/ajpheart.00633.2003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been suggested that oxidative stress contributes to impaired left ventricular (LV) contractility in the setting of heart failure (HF). To test this hypothesis, we studied the effect of an antioxidant on contractility at rest and in response to dobutamine in 10 HF patients. We hypothesized that vitamin C would augment contractility in HF and that this effect would be of a greater magnitude in HF patients compared with patients with normal LV (NLV) function. Data from 10 patients with NLV function who participated in this study are included in this report and have been published elsewhere. A micromanometer-tipped catheter was introduced into the LV. In the experimental protocol, an infusion catheter was positioned in the left main coronary artery. The peak positive rate of change of LV pressure (LV +dP/dt) was measured in response to the intravenous infusion of dobutamine before and during the intracoronary infusion of vitamin C (96 mg/min). Vitamin C had no effect on basal LV +dP/dt in either HF or NLV groups. The infusion of vitamin C augmented the LV +dP/dt response to dobutamine by 22 +/- 4% in the NLV function group. In contrast, vitamin C had no effect on the inotropic response to dobutamine in the HF group. In the control protocol, without vitamin C, no differences were observed between responses to two sequential dobutamine infusions in either group (HF, n = 11; NLV, n = 9). Therefore, a positive effect of vitamin C on contractility was limited to patients with NLV function. The absence of this effect in HF patients may suggest that normal redox responsiveness is lost in this disease state.
Collapse
Affiliation(s)
- Susanna Mak
- Bayer Cardiovascular Clinical Research Laboratory, Division of Cardiology, Mount Sinai Hospital, University of Toronto, Ontario, Canada
| | | |
Collapse
|
44
|
Kumar K, Nguyen K, Waxman S, Nearing BD, Wellenius GA, Zhao SX, Verrier RL. Potent antifibrillatory effects of intrapericardial nitroglycerin in the ischemic porcine heart. J Am Coll Cardiol 2003; 41:1831-7. [PMID: 12767672 DOI: 10.1016/s0735-1097(03)00340-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVES We investigated the antiarrhythmic effects of intrapericardial nitroglycerin (NTG) during acute myocardial ischemia in the porcine heart. BACKGROUND Nitroglycerin is a nitric oxide donor that exerts potent effects on the cardiovascular system. Intrapericardial administration allows investigation of pharmacologic actions on cardiac tissue in an in vivo system while minimizing the confounding influences of systemic effects. METHODS In 29 closed-chest pigs, myocardial ischemia was induced by intraluminal balloon occlusion of the left anterior descending coronary artery. Arrhythmia incidence was monitored during 5-min balloon inflations performed without drug and at 15, 45, 75, and 105 min after NTG (4,000 microg bolus) administered by percutaneous transatrial access into the pericardial space. Electrocardiograms were monitored for ischemia-induced T-wave alternans (TWA), a marker of electrical instability. The antiadrenergic potential of NTG was investigated by examining the drug's suppression of dobutamine-induced increase in myocardial contractility. RESULTS Control coronary artery occlusion provoked ventricular fibrillation (VF) in all animals. Intrapericardial NTG suppressed VF at 45 min in all six pigs (p < 0.05) and reduced TWA across a parallel time course (from 459.1 +/- 144.4 microV before drug to 42.22 +/- 13.96 microV at 45 min, p = 0.047). The antifibrillatory effect occurred as early as 15 min and persisted for up to 75 min. Augmentation of maximum of the first time derivative of left ventricular pressure by dobutamine was blunted by intrapericardial NTG (from 3,999 +/- 196 mm Hg/s before NTG to 3,543 +/- 220 mm Hg/s at 15 min, p = 0.012). CONCLUSIONS Intrapericardial NTG exerts a robust antifibrillatory action. Potential mechanisms include reduction in electrical instability and blunting of adrenergic effects.
Collapse
Affiliation(s)
- Kapil Kumar
- Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Natori S, Hasebe N, Jin YT, Matsusaka T, Ido A, Matsuhashi H, Ihara T, Kikuchi K. Inhaled nitric oxide modifies left ventricular diastolic stress in the presence of vasoactive agents in heart failure. Am J Respir Crit Care Med 2003; 167:895-901. [PMID: 12519739 DOI: 10.1164/rccm.200201-057oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nitric oxide (NO) inhalation therapy has been widely used in several diseases with pulmonary hypertension. However, application of NO inhalation therapy remains controversial in heart failure. Cardiovascular effects of inhaled NO (iNO) were evaluated in dogs before and after induction of heart failure with and without infusion of vasoactive agents. iNO did not affect the baseline left ventricular (LV) function or the response to isoproterenol in control conditions or heart failure induced by procainamide. Pulmonary vascular resistance was significantly decreased by iNO in heart failure with infusion of vasoactive agents. Unexpectedly, LV end-diastolic pressure was significantly elevated by iNO in heart failure in the presence of infusion of vasoactive agents independent of their types; either the vasodilating agents of acetylcholine and nitroglycerin or the vasoconstricting agents of norepinephrine and angiotensin-II. The end-diastolic LV dimension and wall stress were also significantly increased by iNO, however, those at end systole were not affected. These results suggested that NO inhalation therapy reduced pulmonary vascular resistance, whereas in the presence of additional stress of vasoactive agents, it increased LV preload and end-diastolic wall stress in heart failure.
Collapse
Affiliation(s)
- Shunsuke Natori
- First Department of Internal Medicine, Asahikawa Medical College, 2-1-1-1 Midorigaoka higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Nitric oxide (NO) plays critical roles in the regulation of integrated cardiac and vascular function and homeostasis. An understanding of the physiologic role and relative contribution of the three NO synthase isoforms (neuronal--NOS1, inducible--NOS2, and endothelial--NOS3) is imperative to comprehend derangements of the NO signaling pathway in the failing cardiovascular system. Several theories of NO and its regulation have developed as explanations for the divergent observations from studies in health and disease states. Here we review the physiologic and pathophysiologic influence of NO on cardiac function, in a framework that considers several theories of altered NO signaling in heart failure. We discuss the notion of spatial compartmentalization of NO signaling within the myocyte in an effort to reconcile many controversies about derangements in the influences of NO in the heart and vasculature.
Collapse
Affiliation(s)
- Hunter C Champion
- Division of Cardiology, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
47
|
Brutsaert DL. Cardiac endothelial-myocardial signaling: its role in cardiac growth, contractile performance, and rhythmicity. Physiol Rev 2003; 83:59-115. [PMID: 12506127 DOI: 10.1152/physrev.00017.2002] [Citation(s) in RCA: 498] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Experimental work during the past 15 years has demonstrated that endothelial cells in the heart play an obligatory role in regulating and maintaining cardiac function, in particular, at the endocardium and in the myocardial capillaries where endothelial cells directly interact with adjacent cardiomyocytes. The emerging field of targeted gene manipulation has led to the contention that cardiac endothelial-cardiomyocytal interaction is a prerequisite for normal cardiac development and growth. Some of the molecular mechanisms and cellular signals governing this interaction, such as neuregulin, vascular endothelial growth factor, and angiopoietin, continue to maintain phenotype and survival of cardiomyocytes in the adult heart. Cardiac endothelial cells, like vascular endothelial cells, also express and release a variety of auto- and paracrine agents, such as nitric oxide, endothelin, prostaglandin I(2), and angiotensin II, which directly influence cardiac metabolism, growth, contractile performance, and rhythmicity of the adult heart. The synthesis, secretion, and, most importantly, the activities of these endothelium-derived substances in the heart are closely linked, interrelated, and interactive. It may therefore be simplistic to try and define their properties independently from one another. Moreover, in relation specifically to the endocardial endothelium, an active transendothelial physicochemical gradient for various ions, or blood-heart barrier, has been demonstrated. Linkage of this blood-heart barrier to the various other endothelium-mediated signaling pathways or to the putative vascular endothelium-derived hyperpolarizing factors remains to be determined. At the early stages of cardiac failure, all major cardiovascular risk factors may cause cardiac endothelial activation as an adaptive response often followed by cardiac endothelial dysfunction. Because of the interdependency of all endothelial signaling pathways, activation or disturbance of any will necessarily affect the others leading to a disturbance of their normal balance, leading to further progression of cardiac failure.
Collapse
|
48
|
Petrashevskaya NN, Koch SE, Bodi I, Schwartz A. Calcium cycling, historic overview and perspectives. Role for autonomic nervous system regulation. J Mol Cell Cardiol 2002; 34:885-96. [PMID: 12234761 DOI: 10.1006/jmcc.2002.2033] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The human heart proceeds from a relaxed state (diastole) to a fully contracted state (systole) and recovery in 600ms. During this period, Ca(2+) inside the myocardial cell rises from about 10nM to about 100nM and returns to the former. The contractile-relaxation cycle is tightly coupled to the Ca(2+)transient. In the normal physiological state, the autonomic nervous system (ANS) plays a major role in the regulation of cardiac function and important changes occur in diseases of the heart. Sympathetic overdrive is a major determinant of the critical transition from initial compensatory hypertrophy to decompensated failure. Cardiac myocytes from failing hearts are characterized by a number of abnormalities in excitation-contraction coupling, that are a direct consequence of beta-adrenergic signaling defects. Although desensitized in cardiac hypertrophy and failure, the beta-adrenergic signaling pathway retains receptor capacity, a characteristic that is used in therapeutic approaches. There are several putative Ca(2+)-dependent pathways that exert counterbalancing negative regulation over cAMP-dependent positive inotropic effect and may represent potential targets for contractile stimulation. This review is focused on the interactions between sympathetic drive and aspects of calcium signaling in the heart.
Collapse
Affiliation(s)
- Natalia N Petrashevskaya
- Institute of Molecular Pharmacology and Biophysics, Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0828, USA
| | | | | | | |
Collapse
|
49
|
Abstract
OBJECTIVE To review reversible myocardial dysfunction affecting critically ill patients without cardiac pathology. DATA SOURCES The bibliography for the study was compiled through a search of different databases for the period 1966-2001. References cited in the selected articles also were reviewed. STUDY SELECTION The selection criteria included all articles published on reversible myocardial dysfunction in critically ill patients. CONCLUSIONS Reversible myocardial dysfunction may develop in a situation of critical pathology, but the etiology of reversible myocardial dysfunction is not fully understood. This dysfunction may be accompanied by increases in enzyme concentrations and electrocardiographic changes. Reversible myocardial dysfunction probably is underdiagnosed, although its presence is associated with a worsening of the prognosis and with more specific therapeutic options. Further studies are necessary to define its true incidence and clinical implications.
Collapse
Affiliation(s)
- Manuel Ruiz Bailén
- Intensive Care Unit, Critical Care and Emergencies Department, Hospital de Poniente, El Ejido, Almería, Spain
| |
Collapse
|
50
|
Abstract
Nitric oxide (NO) participates in the regulation of the daily activities of cells as well as in cytotoxic events. Elucidating the mechanism(s) by which NO carries out its diverse functions has been the goal of numerous laboratories. In the cardiovascular system, evidence indicates that NO mediates its effects via an activation of soluble guanylyl cyclase (sGC). In other tissues, it is not clear if sGC is an exclusive target for NO or what the functions of cGMP might be. It is also unlikely that the diversity of NO actions is explained solely by changes in cGMP. This review focuses on the evidence that NO modulates cAMP signalling, with specific attention to the effects of NO on adenylyl cyclase (AC) as the target of NO regulation.
Collapse
Affiliation(s)
- Claudette Klein
- E.A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University Medical School, 1402 South Grand Boulevard, St. Louis, MO 63104, USA.
| |
Collapse
|