1
|
Pinaffi-Langley ACDC, Dajani RM, Prater MC, Nguyen HVM, Vrancken K, Hays FA, Hord NG. Dietary Nitrate from Plant Foods: A Conditionally Essential Nutrient for Cardiovascular Health. Adv Nutr 2024; 15:100158. [PMID: 38008359 PMCID: PMC10776916 DOI: 10.1016/j.advnut.2023.100158] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023] Open
Abstract
Under specific conditions, such as catabolic stress or systemic inflammation, endogenous nutrient production becomes insufficient and exogenous supplementation (for example, through dietary intake) is required. Herein, we propose consideration of a dietary nitrate from plant foods as a conditionally essential nutrient for cardiovascular health based on its role in nitric oxide homeostasis. Nitrate derived from plant foods may function as a conditionally essential nutrient, whereas nitrate obtained from other dietary sources, such as drinking water and cured/processed meats, warrants separate consideration because of the associated health risks. We have surveyed the literature and summarized epidemiological evidence regarding the effect of dietary nitrate on cardiovascular disease and risk factors. Meta-analyses and population-based observational studies have consistently demonstrated an inverse association of dietary nitrate with blood pressure and cardiovascular disease outcomes. Considering the available evidence, we suggest 2 different approaches to providing dietary guidance on nitrate from plant-based dietary sources as a nutrient: the Dietary Reference Intakes developed by the National Academies of Sciences, Engineering, and Medicine, and the dietary guidelines evaluated by the Academy of Nutrition and Dietetics. Ultimately, this proposal underscores the need for food-based dietary guidelines to capture the complex and context-dependent relationships between nutrients, particularly dietary nitrate, and health.
Collapse
Affiliation(s)
- Ana Clara da C Pinaffi-Langley
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rosa M Dajani
- Nutrition and Food Services, San Francisco Health, University of California, San Francisco, CA, United States
| | - M Catherine Prater
- Department of Foods and Nutrition, Dawson Hall, University of Georgia, Athens, GA, United States
| | - Hoang Van M Nguyen
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | | | - Franklin A Hays
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Norman G Hord
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Department of Nutritional Sciences, College of Education and Human Sciences, Oklahoma State University, Stillwater, OK, United States.
| |
Collapse
|
2
|
De Luca M, Crisci G, Armentaro G, Cicco S, Talerico G, Bobbio E, Lanzafame L, Green CG, McLellan AG, Debiec R, Caferra P, Scicali R, Cannatà A, Israr MZ, Heaney LM, Salzano A. Endothelial Dysfunction and Heart Failure with Preserved Ejection Fraction-An Updated Review of the Literature. Life (Basel) 2023; 14:30. [PMID: 38255646 PMCID: PMC10817572 DOI: 10.3390/life14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Heart failure (HF) is a clinical syndrome consisting of typical symptoms and signs due to structural and/or functional abnormalities of the heart, resulting in elevated intracardiac pressures and/or inadequate cardiac output. The vascular system plays a crucial role in the development and progression of HF regardless of ejection fraction, with endothelial dysfunction (ED) as one of the principal features of HF. The main ED manifestations (i.e., impaired endothelium-dependent vasodilation, increased oxidative stress, chronic inflammation, leukocyte adhesion, and endothelial cell senescence) affect the systemic and pulmonary haemodynamic and the renal and coronary circulation. The present review is aimed to discuss the contribution of ED to HF pathophysiology-in particular, HF with preserved ejection fraction-ED role in HF patients, and the possible effects of pharmacological and non-pharmacological approaches. For this purpose, relevant data from a literature search (PubMed, Scopus, EMBASE, and Medline) were reviewed. As a result, ED, assessed via venous occlusion plethysmography or flow-mediated dilation, was shown to be independently associated with poor outcomes in HF patients (e.g., mortality, cardiovascular events, and hospitalization due to worsening HF). In addition, SGLT2 inhibitors, endothelin antagonists, endothelial nitric oxide synthase cofactors, antioxidants, and exercise training were shown to positively modulate ED in HF. Despite the need for future research to better clarify the role of the vascular endothelium in HF, ED represents an interesting and promising potential therapeutic target.
Collapse
Affiliation(s)
- Mariarosaria De Luca
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), 80131 Naples, Italy
| | - Giulia Crisci
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), 80131 Naples, Italy
| | - Giuseppe Armentaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy
| | - Sebastiano Cicco
- Internal Medicine Unit “Guido Baccelli” and Arterial Hypertension Unit “Anna Maria Pirrelli”, Department of Precision and Regenerative Medicine and Jonic Area (DiMePReJ), University of Bari Aldo Moro, Azienda Ospedaliero-Universitaria Policlinico, 70124 Bari, Italy
| | | | - Emanuele Bobbio
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Kuggen, 417 56 Gothenburg, Sweden
| | - Lorena Lanzafame
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Christopher G. Green
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Abbie G. McLellan
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Radek Debiec
- Department of Cardiovascular Sciences, University of Leicester, Leicester (UK), IHR Leicester Biomedical Research Centre, Groby Road, Leicester LE3 9QP, UK
| | - Paolo Caferra
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Antonio Cannatà
- Department of Cardiology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College, London SE1 8WA, UK
| | - Muhammad Zubair Israr
- Department of Cardiovascular Sciences, University of Leicester, Leicester (UK), IHR Leicester Biomedical Research Centre, Groby Road, Leicester LE3 9QP, UK
| | - Liam M. Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Andrea Salzano
- Cardiac Unit, AORN A Cardarelli, 80131 Naples, Italy
- Cardiac Unit, University Hospital of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
3
|
Coggan AR, Park LK, Racette SB, Davila-Roman VG, Lenzen P, Vehe K, Dore PM, Schechtman KB, Peterson LR. The inorganic NItrate and eXercise performance in Heart Failure (iNIX-HF) phase II clinical trial: Rationale and study design. Contemp Clin Trials Commun 2023; 36:101208. [PMID: 37842318 PMCID: PMC10568282 DOI: 10.1016/j.conctc.2023.101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/16/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Background Heart failure (HF) is a debilitating and often fatal disease that affects millions of people worldwide. Diminished nitric oxide synthesis, signaling, and bioavailability are believed to contribute to poor skeletal muscle function and aerobic capacity. The aim of this clinical trial (iNIX-HF) is to determine the acute and longer-term effectiveness of inorganic nitrate supplementation on exercise performance in patients with HF with reduced ejection fraction (HFrEF). Methods This clinical trial is a double-blind, placebo-controlled, randomized, parallel-arm design study in which patients with HFrEF (n = 75) are randomized to receive 10 mmol potassium nitrate (KNO3) or a placebo capsule daily for 6 wk. Primary outcome measures are muscle power determined by isokinetic dynamometry and peak aerobic capacity (VO2peak) determined during an incremental treadmill exercise test. Endpoints include the acute effects of a single dose of KNO3 and longer-term effects of 6 wk of KNO3. The study is adequately powered to detect expected increases in these outcomes at P < 0.05 with 1-β>0.80. Discussion The iNIX-HF phase II clinical trial will evaluate the effectiveness of inorganic nitrate supplements as a new treatment to ameliorate poor exercise capacity in HFrEF. This study also will provide critical preliminary data for a future 'pivotal', phase III, multi-center trial of the effectiveness of nitrate supplements not only for improving exercise performance, but also for improving symptoms and decreasing other major cardiovascular endpoints. The potential public health impact of identifying a new, relatively inexpensive, safe, and effective treatment that improves overall exercise performance in patients with HFrEF is significant.
Collapse
Affiliation(s)
- Andrew R. Coggan
- Department of Kinesiology, School of Health & Human Sciences, And Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Lauren K. Park
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Susan B. Racette
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | | | - Pattie Lenzen
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Peter M. Dore
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kenneth B. Schechtman
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Linda R. Peterson
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
4
|
Engelen MPKJ, Kirschner SK, Coyle KS, Argyelan D, Neal G, Dasarathy S, Deutz NEP. Sex related differences in muscle health and metabolism in chronic obstructive pulmonary disease. Clin Nutr 2023; 42:1737-1746. [PMID: 37542951 DOI: 10.1016/j.clnu.2023.06.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/05/2023] [Accepted: 06/28/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND & AIMS Sex differences in muscle function and mass, dyspnea, and clinical outcomes have been observed in patients with Chronic Obstructive Pulmonary Disease (COPD) despite a similar level of airflow obstruction. Protein and amino acid metabolism is altered in COPD, however, it remains unclear whether a difference in metabolic signature exists between males and females with COPD that may explain the observed differences in muscle health and clinical outcomes. METHODS In 234 moderate to severe COPD patients (males/females: 113/121) and 182 healthy controls (males/females: 77/105), we assessed, besides presence of comorbidities and clinical features, muscle function by handgrip and leg dynamometry, and body composition by dual-energy x-ray absorptiometry. In the postabsorptive state, a mixture of 18 stable isotopes of amino acids was administered by pulse and arterialized blood was sampled for 2 h. Amino acid concentrations and enrichments were analyzed by LC-MS/MS to calculate whole body (net) protein breakdown (WBnetPB) and whole body production (WBP) rates (μmol/hour) of the amino acids playing a known role in muscle health. Statistics was done by ANCOVA to examine the effects of sex, COPD, and sex-by-COPD interaction with as covariates age and lean mass. Significance was set as p < 0.05. RESULTS Lung function was comparable between males and females with COPD. Being a female and presence of COPD were independently associated with lower appendicular lean mass, muscle strength, and WBnetPB (p < 0.05). Being a male was associated with higher visceral adipose tissue, C-reactive protein (CRP) (p < 0.05), and higher prevalence of heart failure and obstructive sleep apnea. Sex-by-COPD interactions were found indicating lower fat mass (p = 0.0005) and WBPs of phenylalanine (measure of whole body protein turnover) and essential amino acids (p < 0.05), particularly in COPD females. Higher visceral adipose tissue (p = 0.025), CRP (p < 0.0001), and WBP of tau-methylhistidine (p = 0.010) (reflecting enhanced myofibrillar protein breakdown) were observed in COPD males. CONCLUSIONS Presence of sex specific changes in protein and amino acid metabolism and cardiometabolic health in COPD need to be considered when designing treatment regimens to restore muscle health in males and females with COPD. CLINICAL TRIAL REGISTRY www. CLINICALTRIALS gov, NCT01787682, NCT01624792, NCT02157844, NCT02065141, NCT02770092, NCT02780219, NCT03327181, NCT03796455, NCT01173354, NCT01154400.
Collapse
Affiliation(s)
- Mariëlle P K J Engelen
- Center for Translational Research in Aging & Longevity, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA; Primary Care & Rural Medicine, Texas A&M University, College Station, TX, USA.
| | - Sarah K Kirschner
- Center for Translational Research in Aging & Longevity, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Kimberly S Coyle
- Center for Translational Research in Aging & Longevity, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - David Argyelan
- Center for Translational Research in Aging & Longevity, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Gabriel Neal
- Primary Care & Rural Medicine, Texas A&M University, College Station, TX, USA
| | - Srinivasan Dasarathy
- Department of Gastroenterology and Hepatology, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA; Department of Inflammation and Immunity, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA; Primary Care & Rural Medicine, Texas A&M University, College Station, TX, USA
| |
Collapse
|
5
|
Awada C, Boucherat O, Provencher S, Bonnet S, Potus F. The future of group 2 pulmonary hypertension: Exploring clinical trials and therapeutic targets. Vascul Pharmacol 2023; 151:107180. [PMID: 37178949 DOI: 10.1016/j.vph.2023.107180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Pulmonary hypertension due to left heart disease (PH-LHD) or group 2 PH is the most common and lethal form of PH, occurring secondary to left ventricular systolic or diastolic heart failure (HF), left-sided valvular diseases, and congenital abnormalities. It is subdivided into isolated postcapillary PH (IpcPH) and combined pre- and post-capillary PH (CpcPH), with the latter sharing many similarities with group 1 PH. CpcPH is associated with worse outcomes and increased morbidity and mortality when compared to IpcPH. Although IpcPH can be improved by treatment of the underlying LHD, CpcPH is an incurable disease for which no specific treatment exists, likely due to the lack of understanding of its underlying mechanisms. Furthermore, drugs approved for PAH are not recommended for group 2 PH, as they are either ineffective or even deleterious. With this major unmet medical need, a better understanding of mechanisms and the identification of effective treatment strategies for this deadly condition are urgently needed. This review presents relevant background of the molecular mechanisms underlying PH-LHD that could translate into innovative therapeutic targets and explores novel targets currently being evaluated in clinical trials.
Collapse
Affiliation(s)
- Charifa Awada
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - François Potus
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada.
| |
Collapse
|
6
|
Packer M. The First Dedicated Comprehensive Heart Failure Program in the United States: The Division of Circulatory Physiology at Columbia Presbyterian (1992-2004). J Card Fail 2023; 29:1078-1090. [PMID: 37075940 DOI: 10.1016/j.cardfail.2023.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
The first dedicated multidisciplinary heart failure program in the United States was founded as the Division of Circulatory Physiology at the Columbia University College of Physicians & Surgeons in 1992. The Division was administratively and financially independent of the Division of Cardiology and grew to 24 faculty members at its peak. Its administrative innovations included (1) a comprehensive full-integrated service line, with 2 differentiated clinical teams, one devoted to drug therapy and the other to heart transplantation and ventricular assist devices; (2) a nurse specialist/physician assistant-led clinical service; and (3) a financial structure independent of (and not supported by) other cardiovascular medical or surgical services. The division had 3 overarching missions: (1) to promote a unique career development path for each faculty member to be linked to recognition in a specific area of heart failure expertise; (2) to change the trajectory and enhance the richness of intellectual discourse in the discipline of heart failure, so as to foster an understanding of fundamental mechanisms and to develop new therapeutics; and (3) to provide optimal medical care to patients and to promote the ability of other physicians to provide optimal care. The major research achievements of the division included (1) the development of beta-blockers for heart failure, from initial hemodynamic assessments to proof-of-concept studies to large-scale international trials; (2) the development and definitive assessment of flosequinan, amlodipine, and endothelin antagonists; (3) initial clinical trials and concerns with nesiritide; (4) large-scale trials evaluating dosing of angiotensin converting-enzyme inhibitors and the efficacy and safety of neprilysin inhibition; (5) identification of key mechanisms in heart failure, including neurohormonal activation, microcirculatory endothelial dysfunction, deficiencies in peripheral vasodilator pathways, noncardiac factors in driving dyspnea, and the first identification of subphenotypes of heart failure and a preserved ejection fraction; (6) the development of a volumetric approach to the assessment of myocardial shortening; (7) conceptualization and early studies of cardiac contractility modulation as a treatment for heart failure; (8) novel approaches to the identification of cardiac allograft rejection and new therapeutics to prevent allograft vasculopathy; and (9) demonstration of the effect of left ventricular assist devices to induce reverse remodeling, and the first randomized trial showing a survival benefit with ventricular assist devices. Above all, the division served as an exceptional incubator for a generation of leaders in the field of heart failure.
Collapse
Affiliation(s)
- Milton Packer
- From the Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas, and Imperial College, London, UK.
| |
Collapse
|
7
|
Fuertes-Kenneally L, Manresa-Rocamora A, Blasco-Peris C, Ribeiro F, Sempere-Ruiz N, Sarabia JM, Climent-Paya V. Effects and Optimal Dose of Exercise on Endothelial Function in Patients with Heart Failure: A Systematic Review and Meta-Analysis. SPORTS MEDICINE - OPEN 2023; 9:8. [PMID: 36739344 PMCID: PMC9899305 DOI: 10.1186/s40798-023-00553-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Exercise-based cardiac rehabilitation (CR) is considered an effective treatment for enhancing endothelial function in patients with heart failure (HF). However, recent studies have been published and the optimal "dose" of exercise required to increase the benefits of exercise-based CR programmes on endothelial function is still unknown. OBJECTIVES (a) To estimate the effect of exercise-based CR on endothelial function, assessed by flow-mediated dilation (FMD), in patients with HF; (b) to determine whether high-intensity interval training (HIIT) is better than moderate-intensity training (MIT) for improving FMD; and (c) to investigate the influence of exercise modality (i.e. resistance exercise vs. aerobic exercise and combined exercise vs. aerobic exercise) on the improvement of endothelial function. METHODS Electronic searches were carried out in PubMed, Embase, and Scopus up to February 2022. Random-effects models of between-group mean differences were estimated. Heterogeneity analyses were performed by means of the chi-square test and I2 index. Subgroup analyses and meta-regressions were used to test the influence of potential moderator variables on the effect of exercise. RESULTS We found a FMD increase of 3.09% (95% confidence interval [CI] = 2.01, 4.17) in favour of aerobic-based CR programmes compared with control groups in patients with HF and reduced ejection fraction (HFrEF). However, the results of included studies were inconsistent (p < .001; I2 = 95.2%). Higher FMD improvement was found in studies which were randomised, reported radial FMD, or performed higher number of training sessions a week. Moreover, HIIT enhanced FMD to a greater extent than MIT (2.35% [95% CI = 0.49, 4.22]) in patients with HFrEF. Insufficient data prevented pooled analyses for the effect of exercise in patients with HF and preserved ejection fraction and the influence of exercise modality on the improvement of endothelial function. CONCLUSION Aerobic-based CR is a non-pharmacological treatment for enhancing endothelial function in patients with HFrEF. However, higher training frequency and HIIT induce greater adaptation of endothelial function in these patients, which should betaken into consideration when designing exercise-based CR programmes. Trial registration The protocol was prospectively registered on the PROSPERO database (CRD42022304687).
Collapse
Affiliation(s)
- Laura Fuertes-Kenneally
- grid.513062.30000 0004 8516 8274Institute for Health and Biomedical Research of Alicante (ISABIAL), 03010 Alicante, Spain ,Cardiology Department, Alicante General University Hospital (HGUA), 03010 Alicante, Spain
| | - Agustín Manresa-Rocamora
- grid.513062.30000 0004 8516 8274Institute for Health and Biomedical Research of Alicante (ISABIAL), 03010 Alicante, Spain ,grid.26811.3c0000 0001 0586 4893Department of Sport Sciences, Sports Research Centre, Miguel Hernández University of Elche, 03202 Elche, Spain
| | - Carles Blasco-Peris
- grid.513062.30000 0004 8516 8274Institute for Health and Biomedical Research of Alicante (ISABIAL), 03010 Alicante, Spain ,grid.5338.d0000 0001 2173 938XDepartment of Physical Education and Sport, University of Valencia, 46010 Valencia, Spain
| | - Fernando Ribeiro
- grid.7311.40000000123236065Institute of Biomedicine‑iBiMED and School of Health Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Noemí Sempere-Ruiz
- grid.513062.30000 0004 8516 8274Institute for Health and Biomedical Research of Alicante (ISABIAL), 03010 Alicante, Spain ,grid.26811.3c0000 0001 0586 4893Department of Sport Sciences, Sports Research Centre, Miguel Hernández University of Elche, 03202 Elche, Spain
| | - José Manuel Sarabia
- grid.513062.30000 0004 8516 8274Institute for Health and Biomedical Research of Alicante (ISABIAL), 03010 Alicante, Spain ,grid.26811.3c0000 0001 0586 4893Department of Sport Sciences, Sports Research Centre, Miguel Hernández University of Elche, 03202 Elche, Spain
| | - Vicente Climent-Paya
- grid.513062.30000 0004 8516 8274Institute for Health and Biomedical Research of Alicante (ISABIAL), 03010 Alicante, Spain ,Cardiology Department, Alicante General University Hospital (HGUA), 03010 Alicante, Spain
| |
Collapse
|
8
|
Park LK, Coggan AR, Peterson LR. Skeletal Muscle Contractile Function in Heart Failure With Reduced Ejection Fraction-A Focus on Nitric Oxide. Front Physiol 2022; 13:872719. [PMID: 35721565 PMCID: PMC9198547 DOI: 10.3389/fphys.2022.872719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022] Open
Abstract
Despite advances over the past few decades, heart failure with reduced ejection fraction (HFrEF) remains not only a mortal but a disabling disease. Indeed, the New York Heart Association classification of HFrEF severity is based on how much exercise a patient can perform. Moreover, exercise capacity-both aerobic exercise performance and muscle power-are intimately linked with survival in patients with HFrEF. This review will highlight the pathologic changes in skeletal muscle in HFrEF that are related to impaired exercise performance. Next, it will discuss the key role that impaired nitric oxide (NO) bioavailability plays in HFrEF skeletal muscle pathology. Lastly, it will discuss intriguing new data suggesting that the inorganic nitrate 'enterosalivary pathway' may be leveraged to increase NO bioavailability via ingestion of inorganic nitrate. This ingestion of inorganic nitrate has several advantages over organic nitrate (e.g., nitroglycerin) and the endogenous nitric oxide synthase pathway. Moreover, inorganic nitrate has been shown to improve exercise performance: both muscle power and aerobic capacity, in some recent small but well-controlled, cross-over studies in patients with HFrEF. Given the critical importance of better exercise performance for the amelioration of disability as well as its links with improved outcomes in patients with HFrEF, further studies of inorganic nitrate as a potential novel treatment is critical.
Collapse
Affiliation(s)
- Lauren K. Park
- Department of Medicine, Cardiology Division, Washington University School of Medicine, Saint Louis, MO, United States
| | - Andrew R. Coggan
- Department of Kinesiology, Indiana University Purdue University, Indianapolis, IN, United States
| | - Linda R. Peterson
- Department of Medicine, Cardiology Division, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
9
|
Abstract
The development of pulmonary hypertension (PH) is common and has adverse prognostic implications in patients with heart failure due to left heart disease (LHD), and thus far, there are no known treatments specifically for PH-LHD, also known as group 2 PH. Diagnostic thresholds for PH-LHD, and clinical classification of PH-LHD phenotypes, continue to evolve and, therefore, present a challenge for basic and translational scientists actively investigating PH-LHD in the preclinical setting. Furthermore, the pathobiology of PH-LHD is not well understood, although pulmonary vascular remodeling is thought to result from (1) increased wall stress due to increased left atrial pressures; (2) hemodynamic congestion-induced decreased shear stress in the pulmonary vascular bed; (3) comorbidity-induced endothelial dysfunction with direct injury to the pulmonary microvasculature; and (4) superimposed pulmonary arterial hypertension risk factors. To ultimately be able to modify disease, either by prevention or treatment, a better understanding of the various drivers of PH-LHD, including endothelial dysfunction, abnormalities in vascular tone, platelet aggregation, inflammation, adipocytokines, and systemic complications (including splanchnic congestion and lymphatic dysfunction) must be further investigated. Here, we review the diagnostic criteria and various hemodynamic phenotypes of PH-LHD, the potential biological mechanisms underlying this disorder, and pressing questions yet to be answered about the pathobiology of PH-LHD.
Collapse
Affiliation(s)
- Jessica H Huston
- Division of Cardiology, Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA (J.H.H.)
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.)
| |
Collapse
|
10
|
Manresa-Rocamora A, Ribeiro F, Casanova-Lizón A, Flatt AA, Sarabia JM, Moya-Ramón M. Cardiac Rehabilitation Improves Endothelial Function in Coronary Artery Disease Patients. Int J Sports Med 2022; 43:905-920. [PMID: 35468652 DOI: 10.1055/a-1717-1798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Exercise-based cardiac rehabilitation may be an effective non-pharmacological intervention for improving endothelial function in coronary artery disease patients. Therefore, this systematic review with meta-analysis aimed to (a) estimate the training-induced effect on endothelial and vascular smooth muscle function, assessed by flow-mediated dilation and nitroglycerin-mediated dilation, respectively, in coronary artery disease patients; and to (b) study the influence of potential trial-level variables (i. e. study and intervention characteristics) on the training-induced effect on endothelial and vascular smooth muscle function. Electronic searches were performed in Pubmed, Scopus, and Embase up to February 2021. Random-effects models of standardised mean change were estimated. Heterogeneity analyses were performed by using the Chi 2 test and I 2 index. Our results showed that exercise-based cardiac rehabilitation significantly enhanced flow-mediated dilation (1.04 [95% confidence interval=0.76 to 1.31]) but did not significantly change nitroglycerin-mediated dilation (0.05 [95% confidence interval=-0.03 to 0.13]). Heterogeneity testing reached statistical significance (p<.001) with high inconsistency for flow-mediated dilation (I 2 =92%). Nevertheless, none of the analysed variables influenced the training-induced effect on flow-mediated dilation. Exercise-based cardiac rehabilitation seems to be an effective therapeutic strategy for improving endothelial-dependent dilation in coronary artery disease patients, which may aid in the prevention of cardiovascular events.
Collapse
Affiliation(s)
- Agustín Manresa-Rocamora
- Sports Research Centre, Department of Sport Sciences, Miguel Hernández University of Elche, 03202 Elche, Spain.,Institute for Health and Biomedical Research (ISABIAL Foundation), Miguel Hernandez University, 03550 Alicante, Spain
| | - Fernando Ribeiro
- School of Health Sciences and Institute of Biomedicine- iBiMED, Universidade de Aveiro, Aveiro, Portugal
| | - Antonio Casanova-Lizón
- Sports Research Centre, Department of Sport Sciences, Miguel Hernández University of Elche, 03202 Elche, Spain
| | - Andrew A Flatt
- Health Sciences and Kinesiology, Georgia Southern University - Armstrong Campus, Savannah, United States
| | - José Manuel Sarabia
- Sports Research Centre, Department of Sport Sciences, Miguel Hernández University of Elche, 03202 Elche, Spain.,Institute for Health and Biomedical Research (ISABIAL Foundation), Miguel Hernandez University, 03550 Alicante, Spain
| | - Manuel Moya-Ramón
- Sports Research Centre, Department of Sport Sciences, Miguel Hernández University of Elche, 03202 Elche, Spain.,Institute for Health and Biomedical Research (ISABIAL Foundation), Miguel Hernandez University, 03550 Alicante, Spain
| |
Collapse
|
11
|
Porro B, Eligini S, Conte E, Cosentino N, Capra N, Cavalca V, Banfi C. An Optimized MRM-Based Workflow of the l-Arginine/Nitric Oxide Pathway Metabolites Revealed Disease- and Sex-Related Differences in the Cardiovascular Field. Int J Mol Sci 2022; 23:ijms23031136. [PMID: 35163055 PMCID: PMC8835333 DOI: 10.3390/ijms23031136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/29/2022] Open
Abstract
Clinical data indicate that low circulating l-homoarginine (HArg) concentrations are associated with cardiovascular (CV) disease, CV mortality, and all-cause mortality. A high number of LC-based analytical methods for the quantification of HArg, in combination with the l-arginine (Arg)-related pathway metabolites, have been reported. However, these methods usually consider a limited panel of analytes. Thus, in order to achieve a comprehensive picture of the Arg metabolism, we described an improved targeted metabolomic approach based on a multiple reaction monitoring (MRM) mass spectrometry method for the simultaneous quantification of the Arg/nitric oxide (NO) pathway metabolites. This methodology was then employed to quantify the plasma concentrations of these analytes in a cohort of individuals with different grades/types of coronary artery disease (CAD) in order to increase knowledge about the role of HArg and its associated metabolites in the CV field. Our results showed that the MRM method here implemented is suitable for the simultaneous assessment of a wide panel of amino acids involved in the Arg/NO metabolic pathway in plasma samples from patients with CV disease. Further, our findings highlighted an impairment of the Arg/NO metabolic pathway, and suggest a sex-dependent regulation of this metabolic route.
Collapse
|
12
|
Soman D, Hodovan J, Macon CJ, Davidson BP, Belcik JT, Mudd JO, Park BS, Lindner JR. Contrast Ultrasound Assessment of Skeletal Muscle Recruitable Perfusion after Permanent Left Ventricular Assist Device Implantation: Implications for Functional Recovery. J Am Soc Echocardiogr 2021; 35:495-502. [PMID: 34973393 PMCID: PMC9081119 DOI: 10.1016/j.echo.2021.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND In heart failure with reduced ejection fraction (HFrEF), abnormal regulation of skeletal muscle perfusion contributes to reduced exercise tolerance. The aim of this study was to test the hypothesis that improvement in functional status after permanent left ventricular assist device (LVAD) implantation in patients with HFrEF is related to improvement in muscle perfusion during work, which was measured using contrast-enhanced ultrasound (CEUS). METHODS CEUS perfusion imaging of calf muscle at rest and during low-intensity plantar flexion exercise (20 W, 0.2 Hz) was performed in patients with HFrEF (n = 22) at baseline and 3 months after placement of permanent LVADs. Parametric analysis of CEUS data was used to quantify muscle microvascular blood flow (MBF), blood volume index, and red blood cell flux rate. For subjects alive at 3 months, comparisons were made between those with New York Heart Association functional class I or II (n = 13) versus III or IV (n = 7) status after LVAD. Subjects were followed for a median of 5.7 years for mortality. RESULTS Echocardiographic data before and after LVAD placement and LVAD parameters were similar in subjects classified with New York Heart Association functional class I-II versus functional class III-IV after LVAD. Skeletal muscle MBF at rest and during exercise before LVAD implantation was also similar between groups. After LVAD placement, resting MBF remained similar between groups, but during exercise those with New York Heart Association functional class I or II had greater exercise MBF (111 ± 60 vs 52 ± 38 intensity units/sec, P = .03), MBF reserve (median, 4.45 [3.95 to 6.80] vs 2.22 [0.98 to 3.80]; P = .02), and percentage change in exercise MBF (median, 73% [-28% to 83%] vs -45% [-80% to 26%]; P = .03). During exercise, increases in MBF were attributable to faster microvascular flux rate, with little change in blood volume index, indicating impaired exercise-mediated microvascular recruitment. The only clinical or echocardiographic feature that correlated with post-LVAD exercise MBF was a history of diabetes mellitus. There was a trend toward better survival in patients who demonstrated improvement in muscle exercise MBF after LVAD placement (P = .05). CONCLUSIONS CEUS perfusion imaging can quantify peripheral vascular responses to advanced therapies for HFrEF. After LVAD implantation, improvement in functional class is seen in patients with improvements in skeletal muscle exercise perfusion and flux rate, implicating a change in vasoactive substances that control resistance arteriolar tone.
Collapse
Affiliation(s)
- Divya Soman
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - James Hodovan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Conrad J Macon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Brian P Davidson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - J Todd Belcik
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - James O Mudd
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Byung S Park
- School of Public Health, Oregon Health & Science University, Portland, Oregon
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
13
|
Gee LC, Massimo G, Lau C, Primus C, Fernandes D, Chen J, Rathod KS, Hamers AJP, Filomena F, Nuredini G, Ibrahim AS, Khambata RS, Gupta AK, Moon JC, Kapil V, Ahluwalia A. Inorganic nitrate attenuates cardiac dysfunction: role for xanthine oxidoreductase and nitric oxide. Br J Pharmacol 2021; 179:4757-4777. [PMID: 34309015 DOI: 10.1111/bph.15636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/28/2022] Open
Abstract
Nitric oxide (NO) is a vasodilator and independent modulator of cardiac remodelling. Commonly, in cardiac disease (e.g. heart failure) endothelial dysfunction (synonymous with NO-deficiency) has been implicated in increased blood pressure (BP), cardiac hypertrophy and fibrosis. Currently no effective therapies replacing NO have succeeded in the clinic. Inorganic nitrate (NO3 - ), through chemical reduction to nitrite and then NO, exerts potent BP-lowering but whether it might be useful in treating undesirable cardiac remodelling is unknown. In a nested age- and sex-matched case-control study of hypertensive patients +/- left ventricular hypertrophy (NCT03088514) we show that lower plasma nitrite concentration and vascular dysfunction accompany cardiac hypertrophy and fibrosis in patients. In mouse models of cardiac remodelling, we also show that restoration of circulating nitrite levels using dietary nitrate improves endothelial dysfunction through targeting of xanthine oxidoreductase (XOR)-driven H2 O2 and superoxide, and reduces cardiac fibrosis through NO-mediated block of SMAD-phosphorylation leading to improvements in cardiac structure and function. We show that via these mechanisms dietary nitrate offers easily translatable therapeutic options for treatment of cardiac dysfunction.
Collapse
Affiliation(s)
- Lorna C Gee
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Clement Lau
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Christopher Primus
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Daniel Fernandes
- Departamento de Farmacologia, Federal University of Santa Catarina, Florianópolis, Santa Catarina,, Brazil
| | - Jianmin Chen
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jozua Pedro Hamers
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Federica Filomena
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Gani Nuredini
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Abdiwahab Shidane Ibrahim
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Ajay K Gupta
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - James C Moon
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Vikas Kapil
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
14
|
Nishat S, Gumina RJ. The chicken, the egg, and the elephant: eNOS and NRG1 in fibrosis. Am J Physiol Heart Circ Physiol 2021; 321:H292-H293. [PMID: 34170198 DOI: 10.1152/ajpheart.00308.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shamama Nishat
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Department of Cellular Biology and Physiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Richard J Gumina
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Department of Cellular Biology and Physiology, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
15
|
Miller RJH, Howlett JG, Fine NM. A Novel Approach to Medical Management of Heart Failure With Reduced Ejection Fraction. Can J Cardiol 2021; 37:632-643. [PMID: 33453357 DOI: 10.1016/j.cjca.2020.12.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 01/05/2023] Open
Abstract
The advent of newly available medical therapies for heart failure with reduced ejection fraction (HFrEF) has resulted in many potential therapeutic combinations, increasing treatment complexity. Publication of expert consensus guidelines and initiatives aimed to improve implementation of treatment has emphasized sequential stepwise initiation and titration of medical therapy, which is labour intensive. Data taken from heart failure registries show suboptimal use of medications, prolonged titration times, and consequently little change in dose intensity, all of which indicate therapeutic inertia. Recently published evidence indicates that 4 medication classes-renin-angiotensin-neprilysin inhibitors, β-blockers, mineralocorticoid antagonists, and sodium-glucose cotransporter inhibitors-which we refer to as Foundational Therapy, confer rapid and robust reduction in both morbidity and mortality in most patients with HFrEF and that they work in additive fashion. Additional morbidity and mortality may be observed following addition of several personalized therapies in specific subgroups of patients. In this review, we discuss mechanisms of action of these therapies and propose a framework for their implementation, based on several principles. These include the critical importance of rapid initiation of all 4 Foundational Therapies followed by their titration to target doses, emphasis on multiple simultaneous drug changes with each patient encounter, attention to patient-specific factors in choice of medication class, leveraging inpatient care, use of the entire health care team, and alternative (ie, virtual visits) modes of care. We have incorporated these principles into a Cluster Scheme designed to facilitate timely and optimal medical treatment for patients with HFrEF.
Collapse
Affiliation(s)
- Robert J H Miller
- Division of Cardiology, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan G Howlett
- Division of Cardiology, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Nowell M Fine
- Division of Cardiology, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
16
|
Lago-Rodríguez Á, Domínguez R, Ramos-Álvarez JJ, Tobal FM, Jodra P, Tan R, Bailey SJ. The Effect of Dietary Nitrate Supplementation on Isokinetic Torque in Adults: A Systematic Review and Meta-Analysis. Nutrients 2020; 12:E3022. [PMID: 33023118 PMCID: PMC7601047 DOI: 10.3390/nu12103022] [Citation(s) in RCA: 2] [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] [Received: 08/26/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Dietary nitrate (NO3-) supplementation, which can enhance performance in exercise settings involving repeated high-intensity efforts, has been linked to improved skeletal muscle contractile function. Although muscular strength is an important component of explosive movements and sport-specific skills, few studies have quantified indices of muscular strength following NO3- supplementation, particularly isokinetic assessments at different angular velocities. We performed a systematic review and meta-analysis to determine whether dietary NO3- supplementation improves peak torque, as assessed by the gold standard method of isokinetic dynamometry, and if this effect was linked to the angular velocity imposed during the assessment. Dialnet, Directory of Open Access Journals, MEDLINE, PubMed, SciELO, Scopus, and SPORTDiscus were searched for articles using the following search strategy: (nitrate OR beet*) AND (supplement* OR nutr* OR diet*) AND (isokinetic OR strength OR "resistance exercise" OR "resistance training" OR "muscular power"). The meta-analysis of data from 5 studies with 60 participants revealed an overall effect size of -0.01 for the effect of nitrate supplementation on isokinetic peak torque, whereas trivial effect sizes ranging from -0.11 to 0.16 were observed for independent velocity-specific (90°/s, 180°/s, 270°/s, and 360°/s) isokinetic peak torque. Four of the five studies indicated that dietary NO3- supplementation is not likely to influence voluntary knee extensor isokinetic torque across a variety of angular velocities. These results suggest that NO3- supplementation does not influence isokinetic peak torque, but further work is required to elucidate the potential of NO3- supplementation to influence other indices of muscular strength, given the dearth of experimental evidence on this topic.
Collapse
Affiliation(s)
| | - Raúl Domínguez
- Studies Research Group in Neuromuscular Responses (GEPREN), University of Lavras, 37200-000 Lavras, Brazil
| | - Juan José Ramos-Álvarez
- Faculty of Medicine, School of Medicine of Physical Education and Sport, Complutense University, 28040 Madrid, Spain; (J.J.R.-Á.); (F.M.T.)
| | - Francisco Miguel Tobal
- Faculty of Medicine, School of Medicine of Physical Education and Sport, Complutense University, 28040 Madrid, Spain; (J.J.R.-Á.); (F.M.T.)
| | - Pablo Jodra
- Faculty of Education Sciences, University of Alcalá, 19001 Guadalajara, Spain;
| | - Rachel Tan
- Faculty of Sports Medicine, Natural Sciences Division, Pepperdine University, Malibu, CA 90263, USA;
| | - Stephen J. Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| |
Collapse
|
17
|
Bosso G, Valvano A, Apuzzi V, Mercurio V, Di Simone V, Cittadini A, Napoli R, Oliviero U. Peripheral Vascular Function in Dilated Cardiomyopathy of Different Etiology. Angiology 2020; 71:726-733. [DOI: 10.1177/0003319720932803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular function in dilated cardiomyopathy of different etiology has been poorly investigated. Moreover, reference values of flow-mediated dilation (FMD) in chronic heart failure (CHF) need to be updated according to the new standardized protocols. We characterized the vascular impairment in different stages of post-ischemic dilated cardiomyopathy (PI-DC) or idiopathic dilated cardiomyopathy (I-DC). Eighty consecutive outpatients with CHF in different New York Heart Association (NYHA) classes (45 PI-DC, 35 I-DC) and 50 control subjects underwent FMD and brachial distensibility coefficient measurement. Patients with CHF showed a marked impairment in FMD compared with controls that worsened from classes NYHA I-II to III-IV, independently of etiology ( P < .05). New York Heart Association I-II PI-DC patients showed a worse FMD compared with NYHA I-II I-DC patients ( P < .05). Brachial distensibility coefficient values were significantly lower in patients with CHF compared with controls ( P < .001) without differences between PI-DC and I-DC. In conclusion, advanced CHF is characterized by vascular impairment that is independent of etiology. In the early stages of CHF, endothelial dysfunction is more severe in patients with PI-DC compared with I-DC probably due to the high cardiovascular risk profile. In I-DC, vascular function impairment is independent of cardiovascular risk factors and could participate in the pathogenesis of I-DC.
Collapse
Affiliation(s)
- Giorgio Bosso
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Antonio Valvano
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Valentina Apuzzi
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Valeria Di Simone
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Raffaele Napoli
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Ugo Oliviero
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| |
Collapse
|
18
|
Taher R, Sara JD, Toya T, Borlaug BA, Lerman LO, Lerman A. Peripheral endothelial dysfunction is a novel risk factor for systolic dysfunction and heart failure progression. IJC HEART & VASCULATURE 2020; 30:100584. [PMID: 32743042 PMCID: PMC7385446 DOI: 10.1016/j.ijcha.2020.100584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 07/06/2020] [Indexed: 11/20/2022]
Abstract
Peripheral endothelial dysfunction (PED) predicts adverse outcomes in patients with stage B HF. PED was independently associated with stage B HF, even after adjusting for co-variables. PED was strongly associated with progression from stage B to overt stage C HF. Detecting PED may stratify the risk of HF progression.
Background ACC/AHA guidelines recognize the progressive nature of heart failure (HF). Patients with risk factors (Stage A) are at risk for developing asymptomatic cardiac dysfunction (Stage B), which may then lead to symptomatic HF (Stage C). As such, therapies targeting abnormalities in stages A and B may protect against development of symptomatic HF. peripheral endothelial dysfunction (PED) is an independent predictor of adverse outcomes in patients with stage C HF. The aim of the current study was to evaluate whether PED might be associated with Stage B HF, where therapeutic interventions to prevent progression might be more efficacious. Methods We performed a retrospective cross-sectional analysis of patients who were referred for routine cardiovascular evaluation that included an assessment of peripheral endothelial function with reactive hyperemia-peripheral arterial tonometry. Individuals in this study underwent routine clinically indicated echocardiography within 2 months of testing for PED. Patients with clinical HF were excluded. Results The study included 355 patients (mean age 51.5 ± 14.6 years, 231 (65.1%) female). There was a significant association between PED and Stage B HF (Odds Ratio (OR) 6.38; P < 0.0001) that persisted after stratifying by sex. In multivariate analyses PED was significantly associated with Stage B HF (OR 5.33; P = 0.0038), and was also associated with progression to overt stage C HF (OR 4.63; P = 0.033). Conclusion Peripheral endothelial dysfunction is risk factor for Stage B HF, even in low risk individuals. Further study is warranted to better understand the mechanistic basis of PED in HF to reduce the risk of symptomatic progression.
Collapse
Affiliation(s)
- Riad Taher
- Internal Medicine Department and Department of Endocrinology, Rambam HealthCare Campus, Haifa, Israel
| | - Jaskanwal D. Sara
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Takumi Toya
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Division of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Barry A. Borlaug
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Corresponding author at: Department of Cardiovascular Diseases, Mayo College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
19
|
Sara JD, Toya T, Taher R, Lerman A, Gersh B, Anavekar NS. Asymptomatic Left Ventricle Systolic Dysfunction. Eur Cardiol 2020; 15:e13. [PMID: 32373186 PMCID: PMC7199190 DOI: 10.15420/ecr.2019.14] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/06/2020] [Indexed: 12/22/2022] Open
Abstract
Heart failure is a common debilitating illness, associated with significant morbidity and mortality, rehospitalisation and societal costs. Current guidelines and position statements emphasise the management of patients with overt symptomatic disease, but the increasing prevalence of congestive heart failure underscores the need to identify and manage patients with early left ventricular dysfunction prior to symptom onset. Asymptomatic left ventricular systolic dysfunction (ALVSD), classified as stage B heart failure, is defined as depressed left ventricular systolic function in the absence of clinical heart failure. Early initiation of therapies in patients with presumed ALVSD has been shown to lead to better outcomes. In this article, the authors clarify issues surrounding the definition and natural history of ALVSD, outline clinical tools that may be of value in identifying patients with ALVSD and highlight potential opportunities for future investigations to better address aspects of our understanding of this complex syndrome.
Collapse
Affiliation(s)
- Jaskanwal D Sara
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science Rochester, MN, US
| | - Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science Rochester, MN, US.,Division of Cardiology, National Defense Medical College Tokorozawa, Japan
| | - Riad Taher
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science Rochester, MN, US
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science Rochester, MN, US
| | - Bernard Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science Rochester, MN, US
| | - Nandan S Anavekar
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science Rochester, MN, US
| |
Collapse
|
20
|
Williams JK, Smallwood MJ, Benjamin N, D'Souza RJ, Shore AC, Winyard PG, Gilchrist M. Renal nitrate clearance in chronic kidney disease. Nitric Oxide 2020; 97:16-19. [PMID: 32007629 DOI: 10.1016/j.niox.2020.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nitric oxide (NO) is rapidly oxidised in humans to nitrite and nitrate, with nitrate being present in much greater abundance. These oxidation products can be recycled back into nitric oxide via a complex entero-salivary pathway, thus preserving NO activity. Approximately 65% of circulating nitrate is excreted in the urine in 48 h, with the excretory pathway of the remainder unknown. The effect of declining renal function on nitrate clearance is unknown METHODS: Forty five subjects, 21 M, 24F, median age 69 (range 27-75 years) with renal function assessed by CKD-EPI eGFR between 9 and 89 ml/min/1.73 m2 completed the study. Following a 24 h low nitrate diet a microplate spectrophotometric method was employed to measure plasma nitrate concentration and 24 h urinary nitrate excretion were measured to determine renal nitrate clearance. RESULTS There was a strong positive correlation between urinary nitrate clearance and eGFR, (Spearman R = 0.7665, p < 0.0001) with a moderate negative correlation between plasma nitrate concentration and CKD-EPI eGFR, (Spearman's R = -0.37, p = 0.012). There was a trend between fractional excretion of nitrate and CKD-EPI eGFR (ml/min/1.73 m2) Spearman's R 0.27, p = 0.07 though this did not reach statistical significance. Plasma nitrate concentration and serum creatinine concentration were positively correlated, Spearman's R = 0.39, p = 0.008. CONCLUSIONS We have observed a strong positive association between renal nitrate clearance and renal function such that plasma nitrate rises as renal function falls. Fractional excretion of nitrate appears to decline as renal function falls. As such, urinary nitrate excretion is unlikely to be a reliable marker of endogenous NO synthesis in settings where renal function is altered.
Collapse
Affiliation(s)
- J K Williams
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK
| | - M J Smallwood
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK
| | - N Benjamin
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK
| | - R J D'Souza
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK
| | - A C Shore
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK
| | - P G Winyard
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK
| | - M Gilchrist
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Road, EX2 5AX, UK.
| |
Collapse
|
21
|
Engelen MPKJ, Jonker R, Thaden JJ, Ten Have GAM, Jeon MS, Dasarathy S, Deutz NEP. Comprehensive metabolic flux analysis to explain skeletal muscle weakness in COPD. Clin Nutr 2020; 39:3056-3065. [PMID: 32035752 DOI: 10.1016/j.clnu.2020.01.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 01/10/2020] [Accepted: 01/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Metabolic characterization of a well-defined group of patients could be a powerful tool in revealing metabolic signatures to explain limb muscle weakness in chronic diseases. Studies are currently limited in Chronic Obstructive Pulmonary Disease (COPD) to the identification of differential amino acid concentrations but lack comprehensive analysis of the flux through relevant muscle function related metabolic pathways. METHODS In 23 stable patients with moderate to very severe COPD and 19 healthy controls, a comprehensive metabolic flux analysis was conducted by administering an intravenous pulse and primed constant infusion of multiple stable tracers of amino acids known to play a role in muscle health. Blood samples were obtained to calculate production (WBP) and interconversion rates, and plasma concentrations of these amino acids. Lower and upper limb muscle strength, muscle mass, lung function, physical activity level, and disease history and characteristics were assessed. RESULTS The COPD group was characterized by lower and upper limb muscle weakness (P < 0.01) despite preserved muscle mass. Higher values were found in COPD for plasma glutamine, WBP of leucine (P < 0.001), 3-methylhistidine (P < 0.01) (marker of enhanced myofibrillar protein breakdown), citrulline (P < 0.05), and arginine to citrulline conversion (P < 0.05) (reflecting enhanced nitric oxide synthesis). Plasma concentration of β-hydroxy β-methylbutyrate (HMB with anticatabolic, anabolic and contractile properties), WBP of glycine (precursor of creatine and glutathione), and transcutaneous O2 saturation explained up to 79% and 65% of the variation in strength of the lower and upper limb muscles, respectively, in COPD. CONCLUSIONS Comprehensive metabolic flux analysis revealed a homogenous metabolic signature in stable patients with COPD and a specific metabolic profile in those with skeletal muscle weakness. CLINICAL TRIAL REGISTRY ClinicalTrials.gov; No. NCT01787682; URL: www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Mariëlle P K J Engelen
- Center for Translational Research in Aging & Longevity, Dept of Health and Kinesiology, Texas A&M University, College Station, TX, USA.
| | - Renate Jonker
- Center for Translational Research in Aging & Longevity, Dept of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - John J Thaden
- Center for Translational Research in Aging & Longevity, Dept of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - Gabriella A M Ten Have
- Center for Translational Research in Aging & Longevity, Dept of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - Moon Sun Jeon
- Center for Translational Research in Aging & Longevity, Dept of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - Srinivasan Dasarathy
- Departments of Gastroenterology, Hepatology and Pathobiology, Cleveland Clinic, Cleveland, OH, USA
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Dept of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
22
|
Chioncel O, Mebazaa A. Microcirculatory Dysfunction in Acute Heart Failure. Microcirculation 2020. [DOI: 10.1007/978-3-030-28199-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
23
|
Abstract
Advanced heart failure (HF) is a progressive disease characterized by recurrent hospitalizations and high risk of mortality. Indeed, outcomes in late stages of HF approximate those seen in patients with various aggressive malignancies. Clinical trials assessing beneficial outcomes of new treatments in patients with cancer have used innovative approaches to measure impact on total disease burden or surrogates to assess treatment efficacy. Although most cardiovascular outcomes trials continue to use time-to-first event analyses to assess the primary efficacy end point, such analyses do not adequately reflect the impact of new treatments on the totality of the chronic disease burden. Consequently, patient enrichment and other strategies for ongoing clinical trial design, as well as new statistical methodologies, are important considerations, particularly when studying a population with advanced chronic HF. The DREAM-HF trial (Double-Blind Randomized Assessment of Clinical Events With Allogeneic Mesenchymal Precursor Cells in Advanced Heart Failure) is an ongoing, randomized, sham-controlled phase 3 study of the efficacy and safety of mesenchymal precursor cells as immunotherapy in patients with advanced chronic HF with reduced ejection fraction. Mesenchymal precursor cells have a unique multimodal mechanism of action that is believed to result in polarization of proinflammatory type 1 macrophages in the heart to an anti-inflammatory type 2 macrophage state, inhibition of maladaptive adverse left ventricular remodeling, reversal of cardiac and peripheral endothelial dysfunction, and recovery of deranged vasculature. The objective of DREAM-HF is to confirm earlier phase 2 results and evaluate whether mesenchymal precursor cells will reduce the rate of nonfatal recurrent HF-related major adverse cardiac events while delaying or preventing progression of HF to terminal cardiac events. DREAM-HF is an example of an ongoing contemporary events-driven cardiovascular cell-based immunotherapy study that has utilized the concepts of baseline disease enrichment, prognostic enrichment, and predictive enrichment to improve its efficiency by using accumulating data from within as well as external to the trial. Adaptive enrichment designs and strategies are important components of a rational approach to achieve clinical research objectives in shorter clinical trial timelines and with increased cost-effectiveness without compromising ethical standards or the overall statistical integrity of the study. The DREAM-HF trial also presents an alternative approach to traditional composite time-to-first event primary efficacy end points. Statistical methodologies such as the joint frailty model provide opportunities to expand the scope of events-driven HF with reduced ejection fraction clinical trials to utilize time to recurrent nonfatal HF-related major adverse cardiac events as the primary efficacy end point without compromising the integrity of the statistical analyses for terminal cardiac events. In advanced chronic HF with reduced ejection fraction studies, the joint frailty model is utilized to reflect characteristics of the high-risk patient population with important unmet therapeutic needs. In some cases, use of the joint frailty model may substantially reduce sample size requirements. In addition, using an end point that is acceptable to the Food and Drug Administration and the European Medicines Agency, such as recurrent nonfatal HF-related major adverse cardiac events, enables generation of clinically relevant pharmacoeconomic data while providing comprehensive views of the patient's overall cardiovascular disease burden. The major goal of this review is to provide lessons learned from the ongoing DREAM-HF trial that relate to biologic plausibility and flexible clinical trial design and are potentially applicable to other development programs of innovative therapies for patients with advanced cardiovascular disease. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02032004.
Collapse
Affiliation(s)
| | | | - Barry Greenberg
- University of California, San Diego School of Medicine, La Jolla (B.G.)
- Advanced Heart Failure Treatment Program, Sulpizio Cardiovascular Center, University of California, San Diego Healthcare System, La Jolla (B.G.)
| | - Emerson C. Perin
- Stem Cell Center and Adult Cardiology, Texas Heart Institute, Houston (E.C.P.)
| |
Collapse
|
24
|
Alem MM. Endothelial Dysfunction in Chronic Heart Failure: Assessment, Findings, Significance, and Potential Therapeutic Targets. Int J Mol Sci 2019; 20:E3198. [PMID: 31261886 PMCID: PMC6651535 DOI: 10.3390/ijms20133198] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/13/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure (CHF) is a complex syndrome that results from structural and functional disturbances that affect the ability of the heart to supply oxygen to tissues. It largely affects and reduces the patient's quality of life, socio-economic status, and imposes great costs on health care systems worldwide. Endothelial dysfunction (ED) is a newly discovered phenomenon that contributes greatly to the pathophysiology of numerous cardiovascular conditions and commonly co-exists with chronic heart failure. However, the literature lacks clarity as to which heart failure patients might be affected, its significance in CHF patients, and its reversibility with pharmacological and non-pharmacological means. This review will emphasize all these points and summarize them for future researchers interested in vascular pathophysiology in this particular patient population. It will help to direct future studies for better characterization of these two phenomena for the potential discovery of therapeutic targets that might reduce future morbidity and mortality in this "at risk" population.
Collapse
Affiliation(s)
- Manal M Alem
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| |
Collapse
|
25
|
Coggan AR, Broadstreet SR, Mikhalkova D, Bole I, Leibowitz JL, Kadkhodayan A, Park S, Thomas DP, Thies D, Peterson LR. Dietary nitrate-induced increases in human muscle power: high versus low responders. Physiol Rep 2019; 6. [PMID: 29368802 PMCID: PMC5789728 DOI: 10.14814/phy2.13575] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 01/26/2023] Open
Abstract
Maximal neuromuscular power is an important determinant of athletic performance and also quality of life, independence, and perhaps even mortality in patient populations. We have shown that dietary nitrate (NO3−), a source of nitric oxide (NO), improves muscle power in some, but not all, subjects. The present investigation was designed to identify factors contributing to this interindividual variability. Healthy men (n = 13) and women (n = 7) 22–79 year of age and weighing 52.1–114.9 kg were studied using a randomized, double‐blind, placebo‐controlled, crossover design. Subjects were tested 2 h after ingesting beetroot juice (BRJ) either containing or devoid of 12.3 ± 0.8 mmol of NO3−. Plasma NO3− and nitrite (NO2−) were measured as indicators of NO bioavailability and maximal knee extensor speed (Vmax), power (Pmax), and fatigability were determined via isokinetic dynamometry. On average, dietary NO3− increased (P < 0.05) Pmax by 4.4 ± 8.1%. Individual changes, however, ranged from −9.6 to +26.8%. This interindividual variability was not significantly correlated with age, body mass (inverse of NO3− dose per kg), body mass index (surrogate for body composition) or placebo trial Vmax or fatigue index (in vivo indicators of muscle fiber type distribution). In contrast, the relative increase in Pmax was significantly correlated (r = 0.60; P < 0.01) with the relative increase in plasma NO2− concentration. In multivariable analysis female sex also tended (P = 0.08) to be associated with a greater increase in Pmax. We conclude that the magnitude of the dietary NO3−‐induced increase in muscle power is dependent upon the magnitude of the resulting increase in plasma NO2− and possibly female sex.
Collapse
Affiliation(s)
- Andrew R Coggan
- Departments of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana.,Cellular and Integrative Physiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana.,Departments of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Seth R Broadstreet
- Departments of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Deana Mikhalkova
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Indra Bole
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Joshua L Leibowitz
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ana Kadkhodayan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Soo Park
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Deepak P Thomas
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Dakkota Thies
- Departments of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Linda R Peterson
- Departments of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
26
|
Johnson DM, Antoons G. Arrhythmogenic Mechanisms in Heart Failure: Linking β-Adrenergic Stimulation, Stretch, and Calcium. Front Physiol 2018; 9:1453. [PMID: 30374311 PMCID: PMC6196916 DOI: 10.3389/fphys.2018.01453] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Heart failure (HF) is associated with elevated sympathetic tone and mechanical load. Both systems activate signaling transduction pathways that increase cardiac output, but eventually become part of the disease process itself leading to further worsening of cardiac function. These alterations can adversely contribute to electrical instability, at least in part due to the modulation of Ca2+ handling at the level of the single cardiac myocyte. The major aim of this review is to provide a definitive overview of the links and cross talk between β-adrenergic stimulation, mechanical load, and arrhythmogenesis in the setting of HF. We will initially review the role of Ca2+ in the induction of both early and delayed afterdepolarizations, the role that β-adrenergic stimulation plays in the initiation of these and how the propensity for these may be altered in HF. We will then go onto reviewing the current data with regards to the link between mechanical load and afterdepolarizations, the associated mechano-sensitivity of the ryanodine receptor and other stretch activated channels that may be associated with HF-associated arrhythmias. Furthermore, we will discuss how alterations in local Ca2+ microdomains during the remodeling process associated the HF may contribute to the increased disposition for β-adrenergic or stretch induced arrhythmogenic triggers. Finally, the potential mechanisms linking β-adrenergic stimulation and mechanical stretch will be clarified, with the aim of finding common modalities of arrhythmogenesis that could be targeted by novel therapeutic agents in the setting of HF.
Collapse
Affiliation(s)
- Daniel M Johnson
- Department of Cardiothoracic Surgery, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Gudrun Antoons
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
27
|
Shannon OM, Stephan BCM, Minihane AM, Mathers JC, Siervo M. Nitric Oxide Boosting Effects of the Mediterranean Diet: A Potential Mechanism of Action. J Gerontol A Biol Sci Med Sci 2018; 73:902-904. [DOI: 10.1093/gerona/gly087] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 04/16/2018] [Indexed: 02/05/2023] Open
Affiliation(s)
- Oliver M Shannon
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, UK
| | - Blossom C M Stephan
- Institute of Health and Society, Newcastle University, UK
- Institute of Ageing, Newcastle University, UK
| | - Anne-Marie Minihane
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia (UEA), UK
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, UK
| | - Mario Siervo
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, UK
| |
Collapse
|
28
|
Wray DW, Amann M, Richardson RS. Peripheral vascular function, oxygen delivery and utilization: the impact of oxidative stress in aging and heart failure with reduced ejection fraction. Heart Fail Rev 2018; 22:149-166. [PMID: 27392715 DOI: 10.1007/s10741-016-9573-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [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
The aging process appears to be a precursor to many age-related diseases, perhaps the most impactful of which is cardiovascular disease (CVD). Heart disease, a manifestation of CVD, is the leading cause of death in the USA, and heart failure (HF), a syndrome that develops as a consequence of heart disease, now affects almost six million American. Importantly, as this is an age-related disease, this number is likely to grow along with the ever-increasing elderly population. Hallmarks of the aging process and HF patients with a reduced ejection fraction (HFrEF) include exercise intolerance, premature fatigue, and limited oxygen delivery and utilization, perhaps as a consequence of diminished peripheral vascular function. Free radicals and oxidative stress have been implicated in this peripheral vascular dysfunction, as a redox imbalance may directly impact the function of the vascular endothelium. This review aims to bring together studies that have examined the impact of oxidative stress on peripheral vascular function and oxygen delivery and utilization with both healthy aging and HFrEF.
Collapse
Affiliation(s)
- D Walter Wray
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Bldg 2, Rm 1D25, 500 Foothill Drive, Salt Lake City, UT, 84148, USA
- Department of Exercise and Sport Science, University of Utah, Salt Lake City, UT, USA
| | - Markus Amann
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Bldg 2, Rm 1D25, 500 Foothill Drive, Salt Lake City, UT, 84148, USA
- Department of Exercise and Sport Science, University of Utah, Salt Lake City, UT, USA
| | - Russell S Richardson
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Bldg 2, Rm 1D25, 500 Foothill Drive, Salt Lake City, UT, 84148, USA.
- Department of Exercise and Sport Science, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
29
|
Trivedi RK, Polhemus DJ, Li Z, Yoo D, Koiwaya H, Scarborough A, Goodchild TT, Lefer DJ. Combined Angiotensin Receptor-Neprilysin Inhibitors Improve Cardiac and Vascular Function Via Increased NO Bioavailability in Heart Failure. J Am Heart Assoc 2018; 7:JAHA.117.008268. [PMID: 29502102 PMCID: PMC5866338 DOI: 10.1161/jaha.117.008268] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background There is a paucity of data about the mechanisms by which sacubitril/valsartan (also known as LCZ696) improves outcomes in patients with heart failure. Specifically, the effects of sacubitril/valsartan on vascular function and NO bioavailability have not been investigated. We hypothesized that sacubitril/valsartan therapy increases circulating NO levels and improves vascular function in the setting of heart failure. Methods and Results Male spontaneously hypertensive rats underwent myocardial ischemia/reperfusion surgery to induce heart failure and were followed for up to 12 weeks with serial echocardiography. Rats received sacubitril/valsartan (68 mg/kg), valsartan (31 mg/kg), or vehicle starting at 4 weeks after reperfusion. At 8 or 12 weeks of reperfusion, animals were euthanized and tissues were collected for ex vivo analyses of NO bioavailability, aortic vascular reactivity, myocardial and vascular histology, and cardiac molecular assays. Left ventricular structure and function were improved by both valsartan and sacubitril/valsartan compared with vehicle. Sacubitril/valsartan resulted in superior cardiovascular benefits, as evidenced by sustained improvements in left ventricular ejection fraction and end‐diastolic pressure. Ex vivo vascular function, as measured by aortic vasorelaxation responses to acetylcholine and sodium nitroprusside, was significantly improved by valsartan and sacubitril/valsartan, with more sustained improvements afforded by sacubitril/valsartan. Furthermore, myocardial NO bioavailability was significantly enhanced in animals receiving sacubitril/valsartan therapy. Conclusions Sacubitril/valsartan offers superior cardiovascular protection in heart failure and improves vascular function to a greater extent than valsartan alone. Sacubitril/valsartan‐mediated improvements in cardiac and vascular function are likely related to increases in NO bioavailability and explain, in part, the benefits beyond angiotensin receptor blockade.
Collapse
Affiliation(s)
- Rishi K Trivedi
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - David J Polhemus
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Zhen Li
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Daniel Yoo
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Hiroshi Koiwaya
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Amy Scarborough
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Traci T Goodchild
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA .,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
30
|
Abstract
Sepsis-associated organ dysfunction involves multiple responses to inflammation, including endothelial and microvascular dysfunction, immune and autonomic dysregulation, and cellular metabolic reprogramming. The effect of targeting these mechanistic pathways on short- and long-term outcomes depends highly on the timing of therapeutic intervention. Furthermore, there is a need to understand the adaptive or maladaptive character of these mechanisms, to discover phase-specific biomarkers to guide therapy, and to conceptualize these mechanisms in terms of resistance and tolerance.
Collapse
Affiliation(s)
- Rachel Pool
- Department of Anesthesiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Hernando Gomez
- Center for Critical Care Nephrology, The CRISMA (Clinical Research, Investigation, and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh, 3347 Forbes Avenue, Suite 220, Pittsburgh, PA 15213, USA.
| | - John A Kellum
- Center for Critical Care Nephrology, The CRISMA (Clinical Research, Investigation, and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh, 3347 Forbes Avenue, Suite 220, Pittsburgh, PA 15213, USA
| |
Collapse
|
31
|
Coggan AR, Broadstreet SR, Mahmood K, Mikhalkova D, Madigan M, Bole I, Park S, Leibowitz JL, Kadkhodayan A, Thomas DP, Thies D, Peterson LR. Dietary Nitrate Increases VO 2peak and Performance but Does Not Alter Ventilation or Efficiency in Patients With Heart Failure With Reduced Ejection Fraction. J Card Fail 2017; 24:65-73. [PMID: 28916479 DOI: 10.1016/j.cardfail.2017.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Patients with heart failure with reduced ejection fraction (HFrEF) exhibit lower efficiency, dyspnea, and diminished peak oxygen uptake (VO2peak) during exercise. Dietary nitrate (NO3-), a source of nitric oxide (NO), has improved these measures in some studies of other populations. We determined the effects of acute NO3- ingestion on exercise responses in 8 patients with HFrEF using a randomized, double-blind, placebo-controlled, crossover design. METHODS AND RESULTS Plasma NO3-, nitrite (NO2-), and breath NO were measured at multiple time points and respiratory gas exchange was determined during exercise after ingestion of beetroot juice containing or devoid of 11.2 mmol of NO3-. NO3- intake increased (P < .05-0.001) plasma NO3- and NO2- and breath NO by 1469 ± 245%, 105 ± 34%, and 60 ± 18%, respectively. Efficiency and ventilation during exercise were unchanged. However, NO3- ingestion increased (P < .05) VO2peak by 8 ± 2% (ie, from 21.4 ± 2.1 to 23.0 ± 2.3 mL.min-1.kg-1). Time to fatigue improved (P < .05) by 7 ± 3 % (ie, from 582 ± 84 to 612 ± 81 seconds). CONCLUSIONS Acute dietary NO3- intake increases VO2peak and performance in patients with HFrEF. These data, in conjunction with our recent data demonstrating that dietary NO3- also improves muscle contractile function, suggest that dietary NO3- supplementation may be a valuable means of enhancing exercise capacity in this population.
Collapse
Affiliation(s)
- Andrew R Coggan
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana; Department of Cellular and Integrative Physiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana; Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| | - Seth R Broadstreet
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Kiran Mahmood
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Deana Mikhalkova
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Madigan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Indra Bole
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Soo Park
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Joshua L Leibowitz
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ana Kadkhodayan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Deepak P Thomas
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Dakkota Thies
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Linda R Peterson
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri; Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
32
|
Ichige MHA, Pereira MG, Brum PC, Michelini LC. Experimental Evidences Supporting the Benefits of Exercise Training in Heart Failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 999:181-206. [PMID: 29022264 DOI: 10.1007/978-981-10-4307-9_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Heart Failure (HF), a common end point for many cardiovascular diseases, is a syndrome with a very poor prognosis. Although clinical trials in HF have achieved important outcomes in reducing mortality, little is known about functional mechanisms conditioning health improvement in HF patients. In parallel with clinical studies, basic science has been providing important discoveries to understand the mechanisms underlying the pathophysiology of HF, as well as to identify potential targets for the treatment of this syndrome. In spite of being the end-point of cardiovascular derangements caused by different etiologies, autonomic dysfunction, sympathetic hyperactivity, oxidative stress, inflammation and hormonal activation are common factors involved in the progression of this syndrome. Together these causal factors create a closed link between three important organs: brain, heart and the skeletal muscle. In the past few years, we and other groups have studied the beneficial effects of aerobic exercise training as a safe therapy to avoid the progression of HF. As summarized in this chapter, exercise training, a non-pharmacological tool without side effects, corrects most of the HF-induced neurohormonal and local dysfunctions within the brain, heart and skeletal muscles. These adaptive responses reverse oxidative stress, reduce inflammation, ameliorate neurohormonal control and improve both cardiovascular and skeletal muscle function, thus increasing the quality of life and reducing patients' morbimortality.
Collapse
Affiliation(s)
- Marcelo H A Ichige
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Marcelo G Pereira
- Department of Biodynamics of Human Body Movement, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Patrícia C Brum
- Department of Biodynamics of Human Body Movement, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil. .,National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq - Niterói (RJ), Rio de Janeiro, Brazil.
| | - Lisete C Michelini
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.,National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq - Niterói (RJ), Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is a rare progressive disease of the pulmonary vasculature that is characterized by endothelial dysfunction, inflammation, and right ventricular dysfunction. OBJECTIVES The main objective was to determine whether endothelial, inflammatory, and cardiac biomarkers would be associated with the World Health Organization functional assessment and survival in patients with PAH. METHODS We performed a retrospective cohort study of patients with PAH enrolled in the Randomized Clinical Trial of Aspirin and Simvastatin for Pulmonary Arterial Hypertension (ASA-STAT). Biomarkers (N-terminal fragment of pro-BNP [NT-pro-BNP], von Willebrand factor [vWF], soluble P selectin, C-reactive protein, total and high-density lipoprotein cholesterol, triglycerides, tumor necrosis factor, IL-6, β-thromboglobulin, and thromboxane B2) were measured at baseline. Patients from the study were followed until lung transplantation, death, or August 1, 2013. Ordinal logistic regression and Cox regression analyses were performed. MEASUREMENTS AND MAIN RESULTS Sixty-five patients with PAH were enrolled. The mean age was 51 years, and 86% were women. Higher vWF activity, lower high-density lipoprotein cholesterol, and higher thromboxane B2 levels were associated with worse World Health Organization functional class after adjustment for age, sex, and etiology of PAH. Higher NT-pro-BNP levels, lower vWF activity, and lower total cholesterol were associated with an increased risk of death or lung transplant after adjustment for age, sex, etiology of PAH, and 6-minute-walk distance. CONCLUSIONS In patients with PAH, lower vWF activity and cholesterol levels and higher NT-pro-BNP levels at baseline were associated with an increased risk of death or transplantation. Clinical trial registered with www.clinicaltrials.gov (NCT00384865).
Collapse
|
34
|
Abstract
Heart failure (HF) patients suffer from exercise intolerance that diminishes their ability to perform normal activities of daily living and hence compromises their quality of life. This is due largely to detrimental changes in skeletal muscle mass, structure, metabolism, and function. This includes an impairment of muscle contractile performance, i.e., a decline in the maximal force, speed, and power of muscle shortening. Although numerous mechanisms underlie this reduction in contractility, one contributing factor may be a decrease in nitric oxide (NO) bioavailability. Consistent with this, recent data demonstrate that acute ingestion of NO3 (-)-rich beetroot juice, a source of NO via the NO synthase-independent enterosalivary pathway, markedly increases maximal muscle speed and power in HF patients. This review discusses the role of muscle contractile dysfunction in the exercise intolerance characteristic of HF, and the evidence that dietary NO3 (-) supplementation may represent a novel and simple therapy for this currently underappreciated problem.
Collapse
Affiliation(s)
- Andrew R Coggan
- Cardiovascular Imaging Laboratory, Mallinckrodt Institute of Radiology, Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd. - Campus Box 8225, St. Louis, MO, 63110, USA.
| | - Linda R Peterson
- Cardiovascular Imaging Laboratory, Mallinckrodt Institute of Radiology, Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd. - Campus Box 8225, St. Louis, MO, 63110, USA
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, 660 S. Euclid Ave. - Campus Box 8086, St. Louis, MO, 63110, USA
| |
Collapse
|
35
|
Linz W, Itter G, Dobrucki LW, Malinski T, Wiemer G. Ramipril improves nitric oxide availability in hypertensive rats with failing hearts after myocardial infarction. J Renin Angiotensin Aldosterone Syst 2016; 4:180-5. [PMID: 14608524 DOI: 10.3317/jraas.2003.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
A markedly decreased aortic nitric oxide (NO) availability, probably due to impaired endothelial nitric oxide synthase activity with enhanced O2- and peroxynitrite production, seems to be attributable to endothelial dysfunction in spontaneously hypertensive rats (SHR) with severe congestive heart failure (CHF). In this study, we investigated the chronic effect of the angiotensin-converting enzyme inhibitor, ramipril (RA) and the loop diuretic, frusemide (FU), as well as the combination of both on endothelial NO, O2- and peroxynitrite production in aortae from SHR with failing hearts after myocardial infarction (MI). Heart failure was induced by permanent occlusion of the left coronary artery. SHR were randomised to receive either placebo, RA, (1 mg/kg/day), FU (4 mg/kg/day) or RA+FU (1 and 4 mg/kg/day, respectively). Treatments were started two weeks following MI and continued for six weeks. Reduced aortic and coronary flow indices in the working heart, which can be considered as markers for endothelial function, were significantly normalised and improved, respectively, by RA, FU or RA+FU-treatment. Similarly, all three treatment regimens significantly enhanced the reduced calcium ionophore (CaI)-induced NO-release (assessed by a NO-sensitive microsensor) from aortic endothelial cells of placebo- treated animals with CHF. Concomitantly, the increased CaI-stimulated O 2production (assessed by an electrochemical sensor) in aortic endothelial cells of placebo-treated animals with CHF was significantly reduced by RA and RA+FUtreatment. Treatment with RA and RA+FU also attenuated the dramatic increase in endothelial peroxynitrite concentration (chemiluminescence method), which was observed in placebo-treated rats with CHF. FU did not counteract improved haemo- and cardiodynamic parameters by RA. Thus, RA and FU act synergistically to enhance bioavailability of endothelium-derived NO, and this may contribute to the clinical usefulness of the combination of these drugs in treatment of heart failure.
Collapse
Affiliation(s)
- Wolfgang Linz
- Aventis Pharma Deutschlands GmbH, DG Cardiovascular Diseases, Frankfurt, 65926 Germany.
| | | | | | | | | |
Collapse
|
36
|
Mónica FZ, Bian K, Murad F. The Endothelium-Dependent Nitric Oxide-cGMP Pathway. ADVANCES IN PHARMACOLOGY 2016; 77:1-27. [PMID: 27451093 DOI: 10.1016/bs.apha.2016.05.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nitric oxide (NO)-cyclic 3'-5' guanosine monophosphate (cGMP) signaling plays a critical role on smooth muscle tone, platelet activity, cardiac contractility, renal function and fluid balance, and cell growth. Studies of the 1990s established endothelium dysfunction as one of the major causes of cardiovascular diseases. Therapeutic strategies that benefit NO bioavailability have been applied in clinical medicine extensively. Basic and clinical studies of cGMP regulation through activation of soluble guanylyl cyclase (sGC) or inhibition of cyclic nucleotide phosphodiesterase type 5 (PDE5) have resulted in effective therapies for pulmonary hypertension, erectile dysfunction, and more recently benign prostatic hyperplasia. This section reviews (1) how endothelial dysfunction and NO deficiency lead to cardiovascular diseases, (2) how soluble cGMP regulation leads to beneficial effects on disorders of the circulation system, and (3) the epigenetic regulation of NO-sGC pathway components in the cardiovascular system. In conclusion, the discovery of the NO-cGMP pathway revolutionized the comprehension of pathophysiological mechanisms involved in cardiovascular and other diseases. However, considering the expression "from bench to bedside" the therapeutic alternatives targeting NO-cGMP did not immediately follow the marked biochemical and pathophysiological revolution. Some therapeutic options have been effective and released on the market for pulmonary hypertension and erectile dysfunction such as inhaled NO, PDE5 inhibitors, and recently sGC stimulators. The therapeutic armamentarium for many other disorders is expected in the near future. There are currently numerous active basic and clinical research programs in universities and industries attempting to develop novel therapies for many diseases and medical applications.
Collapse
Affiliation(s)
- F Z Mónica
- School of Medicine, George Washington University, Washington, DC, United States; State University of Campinas (UNICAMP), Campinas, Brazil
| | - K Bian
- School of Medicine, George Washington University, Washington, DC, United States.
| | - F Murad
- School of Medicine, George Washington University, Washington, DC, United States.
| |
Collapse
|
37
|
Rammos C, Zeus T, Balzer J, Kubatz L, Hendgen-Cotta UB, Veulemans V, Hellhammer K, Totzeck M, Luedike P, Kelm M, Rassaf T. Percutaneous Mitral Valve Repair in Mitral Regurgitation Reduces Cell-Free Hemoglobin and Improves Endothelial Function. PLoS One 2016; 11:e0151203. [PMID: 26986059 PMCID: PMC4795750 DOI: 10.1371/journal.pone.0151203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 02/23/2016] [Indexed: 11/18/2022] Open
Abstract
Background and Objective Endothelial dysfunction is predictive for cardiovascular events and may be caused by decreased bioavailability of nitric oxide (NO). NO is scavenged by cell-free hemoglobin with reduction of bioavailable NO up to 70% subsequently deteriorating vascular function. While patients with mitral regurgitation (MR) suffer from an impaired prognosis, mechanisms relating to coexistent vascular dysfunctions have not been described yet. Therapy of MR using a percutaneous mitral valve repair (PMVR) approach has been shown to lead to significant clinical benefits. We here sought to investigate the role of endothelial function in MR and the potential impact of PMVR. Methods and Results Twenty-seven patients with moderate-to-severe MR treated with the MitraClip® device were enrolled in an open-label single-center observational study. Patients underwent clinical assessment, conventional echocardiography, and determination of endothelial function by measuring flow-mediated dilation (FMD) of the brachial artery using high-resolution ultrasound at baseline and at 3-month follow-up. Patients with MR demonstrated decompartmentalized hemoglobin and reduced endothelial function (cell-free plasma hemoglobin in heme 28.9±3.8 μM, FMD 3.9±0.9%). Three months post-procedure, PMVR improved ejection fraction (from 41±3% to 46±3%, p = 0.03) and NYHA functional class (from 3.0±0.1 to 1.9±1.7, p<0.001). PMVR was associated with a decrease in cell free plasma hemoglobin (22.3±2.4 μM, p = 0.02) and improved endothelial functions (FMD 4.8±1.0%, p<0.0001). Conclusion We demonstrate here that plasma from patients with MR contains significant amounts of cell-free hemoglobin, which is accompanied by endothelial dysfunction. PMVR therapy is associated with an improved hemoglobin decompartmentalization and vascular function.
Collapse
Affiliation(s)
- Christos Rammos
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Tobias Zeus
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Jan Balzer
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Laura Kubatz
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Ulrike B. Hendgen-Cotta
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Verena Veulemans
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Katharina Hellhammer
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Matthias Totzeck
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Peter Luedike
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Malte Kelm
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Tienush Rassaf
- University Hospital Düsseldorf, Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, 40225, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
38
|
Kim CH, Jae SY, Johnson BD. Pulmonary Hypertension and Cardiopulmonary Exercise in Heart Failure. ACTA ACUST UNITED AC 2015; 1:143-51. [PMID: 26389080 PMCID: PMC4315347 DOI: 10.1159/000360964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In heart failure (HF), pulmonary hypertension (PH) is initially associated with a rise in the left ventricular filling pressure. PH is defined by pulmonary hemodynamic measurements including pulmonary capillary wedge pressure, mean pulmonary arterial pressure and pulmonary vascular resistance. Eventually, PH in HF may become more of a reactive process. Although the mechanism of the reactive PH development is not clearly understood, vascular dysfunction induced by remodeling, vasoactive substances and genetic variation appear to contribute significantly to this form of PH. Noninvasive cardiopulmonary exercise testing has been extensively utilized to assess disease severity in HF patients. It provides integrated information that is dependent on cardiopulmonary hemodynamics, lung mechanics, breathing pattern and strategy. In this review, we will discuss the mechanisms of PH development in HF and how noninvasive gas exchange measures obtained with submaximal exercise are influenced by PH in this population.
Collapse
Affiliation(s)
- Chul-Ho Kim
- Division of Cardiovascular Disease, Mayo Clinic, Rochester, Minn., USA
| | - Sae Young Jae
- The Health and Integrative Physiology Laboratory, University of Seoul, Seoul, Korea
| | - Bruce D Johnson
- Division of Cardiovascular Disease, Mayo Clinic, Rochester, Minn., USA
| |
Collapse
|
39
|
Moore J, Dyson A, Singer M, Fraser J. Microcirculatory dysfunction and resuscitation: why, when, and how. Br J Anaesth 2015; 115:366-75. [DOI: 10.1093/bja/aev163] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
40
|
Abstract
Despite existing therapies, patients with heart failure have a very poor quality of life and a high 1-year mortality rate. Given the impact of this syndrome on health outcomes, research is being directed toward identifying novel strategies to treat heart failure symptoms as well as to prolong survival. One molecule that has been tested in animal models for this purpose is resveratrol. Resveratrol is a naturally occurring polyphenol found in several plants, and administration of resveratrol has been shown to prevent and/or slow the progression of heart failure in animal models of heart failure induced by myocardial infarction, pressure overload, myocarditis, and chemotherapy-induced cardiotoxicity. In addition, some animal studies have shown that resveratrol improves cardiac function and survival when administered as a treatment for established heart failure. Furthermore, as heart failure induces alterations in skeletal muscle and vasculature that also contribute to certain heart failure symptoms, such as fatigue and exercise intolerance, it has also been shown that resveratrol acts on these peripheral tissues to improve skeletal muscle and endothelial/vascular function. Therefore, if these animal studies translate to humans, resveratrol may prove to be a novel therapy for the treatment of heart failure.
Collapse
Affiliation(s)
- Miranda M Sung
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
41
|
Coggan AR, Leibowitz JL, Spearie CA, Kadkhodayan A, Thomas DP, Ramamurthy S, Mahmood K, Park S, Waller S, Farmer M, Peterson LR. Acute Dietary Nitrate Intake Improves Muscle Contractile Function in Patients With Heart Failure: A Double-Blind, Placebo-Controlled, Randomized Trial. Circ Heart Fail 2015; 8:914-20. [PMID: 26179185 DOI: 10.1161/circheartfailure.115.002141] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/02/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Skeletal muscle strength, velocity, and power are markedly reduced in patients with heart failure, which contributes to their impaired exercise capacity and lower quality of life. This muscle dysfunction may be partially because of decreased nitric oxide (NO) bioavailability. We therefore sought to determine whether ingestion of inorganic nitrate (NO3 (-)) would increase NO production and improve muscle function in patients with heart failure because of systolic dysfunction. METHODS AND RESULTS Using a double-blind, placebo-controlled, randomized crossover design, we determined the effects of dietary NO3 (-) in 9 patients with heart failure. After fasting overnight, subjects drank beetroot juice containing or devoid of 11.2 mmol of NO3 (-). Two hours later, muscle function was assessed using isokinetic dynamometry. Dietary NO3 (-) increased (P<0.05-0.001) breath NO by 35% to 50%. This was accompanied by 9% (P=0.07) and 11% (P<0.05) increases in peak knee extensor power at the 2 highest movement velocities tested (ie, 4.71 and 6.28 rad/s). Maximal power (calculated by fitting peak power data with a parabola) was therefore greater (ie, 4.74±0.41 versus 4.20±0.33 W/kg; P<0.05) after dietary NO3 (-) intake. Calculated maximal velocity of knee extension was also higher after NO3 (-) ingestion (ie, 12.48±0.95 versus 11.11±0.53 rad/s; P<0.05). Blood pressure was unchanged, and no adverse clinical events occurred. CONCLUSIONS In this pilot study, acute dietary NO3 (-) intake was well tolerated and enhanced NO bioavailability and muscle power in patients with systolic heart failure. Larger-scale studies should be conducted to determine whether the latter translates into an improved quality of life in this population. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01682356.
Collapse
Affiliation(s)
- Andrew R Coggan
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO.
| | - Joshua L Leibowitz
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Catherine Anderson Spearie
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Ana Kadkhodayan
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Deepak P Thomas
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Sujata Ramamurthy
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Kiran Mahmood
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Soo Park
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Suzanne Waller
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Marsha Farmer
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| | - Linda R Peterson
- From the Cardiovascular Imaging Laboratory, Division of Radiological Sciences, Department of Radiology (A.R.C., J.L.L., L.R.P.), Cardiovascular Division, Department of Medicine (J.L.L., A.K., D.P.T., S.R., K.M., S.P., M.F., L.R.P.), and Center for Applied Research Sciences (C.A.S.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
42
|
|
43
|
den Uil CA, Brugts JJ. Impact of Intravenous Nitroglycerin in the Management of Acute Decompensated Heart Failure. Curr Heart Fail Rep 2014; 12:87-93. [DOI: 10.1007/s11897-014-0230-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
44
|
Couto GK, Davel AP, Brum PC, Rossoni LV. Double disruption of α2A- and α2C-adrenoceptors induces endothelial dysfunction in mouse small arteries: role of nitric oxide synthase uncoupling. Exp Physiol 2014; 99:1427-38. [DOI: 10.1113/expphysiol.2014.079236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Gisele K. Couto
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo SP Brazil
| | - Ana P. Davel
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas; Campinas SP Brazil
| | - Patrícia C. Brum
- School of Physical Education and Sport; University of São Paulo; São Paulo SP Brazil
| | - Luciana V. Rossoni
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo SP Brazil
| |
Collapse
|
45
|
|
46
|
Bhushan S, Kondo K, Polhemus DJ, Otsuka H, Nicholson CK, Tao YX, Huang H, Georgiopoulou VV, Murohara T, Calvert JW, Butler J, Lefer DJ. Nitrite therapy improves left ventricular function during heart failure via restoration of nitric oxide-mediated cytoprotective signaling. Circ Res 2014; 114:1281-91. [PMID: 24599803 DOI: 10.1161/circresaha.114.301475] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
RATIONALE Nitric oxide (NO) bioavailability is reduced in the setting of heart failure. Nitrite (NO2) is a critically important NO intermediate that is metabolized to NO during pathological states. We have previously demonstrated that sodium nitrite ameliorates acute myocardial ischemia/reperfusion injury. OBJECTIVE No evidence exists as to whether increasing NO bioavailability via nitrite therapy attenuates heart failure severity after pressure-overload-induced hypertrophy. METHODS AND RESULTS Serum from patients with heart failure exhibited significantly decreased nitrosothiol and cGMP levels. Transverse aortic constriction was performed in mice at 10 to 12 weeks. Sodium nitrite (50 mg/L) or saline vehicle was administered daily in the drinking water postoperative from day 1 for 9 weeks. Echocardiography was performed at baseline and at 1, 3, 6, and 9 weeks after transverse aortic constriction to assess left ventricular dimensions and ejection fraction. We observed increased cardiac nitrite, nitrosothiol, and cGMP levels in mice treated with nitrite. Sodium nitrite preserved left ventricular ejection fraction and improved left ventricular dimensions at 9 weeks (P<0.001 versus vehicle). In addition, circulating and cardiac brain natriuretic peptide levels were attenuated in mice receiving nitrite (P<0.05 versus vehicle). Western blot analyses revealed upregulation of Akt-endothelial nitric oxide-nitric oxide-cGMP-GS3Kβ signaling early in the progression of hypertrophy and heart failure. CONCLUSIONS These results support the emerging concept that nitrite therapy may be a viable clinical option for increasing NO levels and may have a practical clinical use in the treatment of heart failure.
Collapse
Affiliation(s)
- Shashi Bhushan
- From the LSU Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA (S.B., D.J.P., H.O., D.J.L.); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K., T.M.); Division of Cardiothoracic Surgery, Department of Surgery, Carlyle Fraser Heart Center (C.K.N., J.W.C.) and Division of Cardiology, Department of Medicine (V.V.G., J.B.), Emory University School of Medicine, Atlanta, GA; and Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, AL (Y.-X.T., H.H.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The NO/ONOO-cycle as the central cause of heart failure. Int J Mol Sci 2013; 14:22274-330. [PMID: 24232452 PMCID: PMC3856065 DOI: 10.3390/ijms141122274] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 01/08/2023] Open
Abstract
The NO/ONOO-cycle is a primarily local, biochemical vicious cycle mechanism, centered on elevated peroxynitrite and oxidative stress, but also involving 10 additional elements: NF-κB, inflammatory cytokines, iNOS, nitric oxide (NO), superoxide, mitochondrial dysfunction (lowered energy charge, ATP), NMDA activity, intracellular Ca(2+), TRP receptors and tetrahydrobiopterin depletion. All 12 of these elements have causal roles in heart failure (HF) and each is linked through a total of 87 studies to specific correlates of HF. Two apparent causal factors of HF, RhoA and endothelin-1, each act as tissue-limited cycle elements. Nineteen stressors that initiate cases of HF, each act to raise multiple cycle elements, potentially initiating the cycle in this way. Different types of HF, left vs. right ventricular HF, with or without arrhythmia, etc., may differ from one another in the regions of the myocardium most impacted by the cycle. None of the elements of the cycle or the mechanisms linking them are original, but they collectively produce the robust nature of the NO/ONOO-cycle which creates a major challenge for treatment of HF or other proposed NO/ONOO-cycle diseases. Elevated peroxynitrite/NO ratio and consequent oxidative stress are essential to both HF and the NO/ONOO-cycle.
Collapse
|
48
|
Bech JN, Starklint J, Bentzen H, Nyvad O, Pedersen EB. Renal and hormonal effects of systemic nitric oxide inhibition in patients with congestive heart failure and in healthy control subjects. J Card Fail 2013; 19:776-785. [PMID: 24263123 DOI: 10.1016/j.cardfail.2013.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/10/2013] [Accepted: 10/02/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND The significance of basal renal nitric oxide (NO) availability in the regulation of renal perfusion and sodium excretion in human congestive heart failure (CHF) has not been described previously. METHODS AND RESULTS We studied the effects of acute systemic NO synthesis inhibition with N(G)-monomethyl-L-arginine (L-NMMA) in 12 patients with CHF and 10 healthy control subjects (CON) in a randomized placebo-controlled study. Effect parameters were renal plasma flow (RPF), renal vascular resistance (RVR), glomerular filtration rate (GFR), urine sodium excretion and plasma levels of vasoactive hormones. L-NMMA was associated with a significant decrease in RPF (CON-LNMMA: -13 ± 3% [P = .014]; CHF-LNMMA: -17 ± 7% [P = .017]) and a profound increase in RVR in both CHF and CON (CON-LNMMA: +26 ± 6% [P = .009]; CHF-LNMMA: +37 ± 70% [P = .005]). Significant decreases in sodium excretion were found in both CHF-LNMMA and CON-LNMMA. Relative changes from baseline were not statistically different between CHF-LNMMA and CON-LNMMA. After L-NMMA, RPF values correlated inversely with plasma aldosterone in CHF-LNMMA (P = .01). L-NMMA induced an increase in A-type natriuretic peptide (ANP) only in CHF-LNMMA (+18 ± 8%; P = .035), which correlated significantly with basal ANP levels (P = .034). CONCLUSIONS There was no difference in the renal response to L-NMMA in CHF vs CON, suggesting that the impact of NO on renal perfusion and sodium excretion is maintained in stable CHF. We suggest that NO influences the release of ANP during high levels of atrial stretch in CHF.
Collapse
Affiliation(s)
- J N Bech
- Department of Medical Research, Holstebro Hospital, Holstebro, Denmark and Aarhus University, Aarhus, Denmark.
| | | | | | | | | |
Collapse
|
49
|
Tardin OMA, Pereira SB, Velloso MWM, Balieiro HM, Costa B, Alves TOE, Giro C, Pessoa LP, Ribeiro GS, Mesquita ET. Genetic polymorphism G894T and the prognosis of heart failure outpatients. Arq Bras Cardiol 2013; 101:352-8. [PMID: 23949326 PMCID: PMC4062372 DOI: 10.5935/abc.20130167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/10/2013] [Accepted: 04/15/2013] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Previous studies have analyzed the role of the genetic polymorphism of endothelial nitric oxide synthase on heart failure prognosis. However, there are no studies relating the G894T and heart failure in Brazil. OBJECTIVE To evaluate the association between G894T GP and the prognosis of a sample of Brazilian outpatients with heart failure. METHODS Cohort study included 145 patients with systolic heart failure, followed for up to 40 months (mean = 22), at two university hospitals, in the State of Rio de Janeiro. We evaluated the relationship between G894T and the following outcomes: reverse remodeling, improvement in functional class (NYHA), and mortality and hospitalization rates. The diameters of the left atrium and ventricle, as well as the ejection fraction of the left ventricle, were evaluated at baseline and at 6 months to assess reverse remodeling. The improvement in functional class was evaluated after 6 months, and mortality rate and hospitalization were evaluated during follow-up. Race was self-declared. G894T polymorphism was analyzed by polymerase chain reaction and restriction fragment length polymorphism. RESULTS The genotypic frequencies were GG (40%), GT (48.3%) and TT (11.7%). The allele frequency was guanine (64.1%) and thiamine (35.8%). There were no differences between the genotype or allelic frequencies according to self-declared race, either as baseline characteristics. There was no relationship between genotype or allele frequency and the outcome measures. CONCLUSION No association was observed between the G894T polymorphism (Glu298Asp) and prognosis in this sample of Brazilian outpatients with systolic heart failure.
Collapse
Affiliation(s)
| | | | | | | | - Bruno Costa
- Universidade Federal Fluminense, Niterói, RJ - Brazil
| | | | - Camila Giro
- Universidade Federal Fluminense, Niterói, RJ - Brazil
| | | | | | | |
Collapse
|
50
|
Abstract
Statins lower serum cholesterol and are employed for primary and secondary prevention of cardiovascular events. Clinical evidence from observational studies, retrospective data, and post hoc analyses of data from large statin trials in various cardiovascular conditions, as well as small scale randomized trials, suggest survival and other outcome benefits for heart failure. Two recent large randomized controlled trials, however, appear to suggest statins do not have beneficial effects in heart failure. In addition to lowering cholesterol, statins are believed to have many pleotropic effects which could possibly influence the pathophysiology of heart failure. Following the two large trials, evidence from recent studies appears to support the use of statins in heart failure. This review discusses the role of statins in the pathophysiology of heart failure, current evidence for statin use in heart failure, and suggests directions for future research.
Collapse
Affiliation(s)
- Kwadwo Osei Bonsu
- School of Medicine and Health Sciences, Monash University Sunway Campus, Bandar Sunway, Malaysia
| | | | | |
Collapse
|