1
|
Ratchford SM, Clifton HL, Gifford JR, LaSalle DT, Thurston TS, Bunsawat K, Alpenglow JK, Wright JB, Amann M, Ryan JJ, Wray DW. Impact of Acute Antioxidant and Tetrahydrobiopterin (BH 4) Administration on Locomotor Muscle Microvascular Function in Patients With Heart Failure. Circ Heart Fail 2025:e012446. [PMID: 40270242 DOI: 10.1161/circheartfailure.124.012446] [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: 09/16/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Peripheral microvascular dysfunction is a hallmark feature of both heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF) pathophysiology, due partly to impairments in nitric oxide signaling secondary to tetrahydrobiopterin (BH4) deficiency and oxidative stress. METHODS Using a randomized, double-blind, placebo-controlled crossover design, this study examined the impact of enteral BH4 (10 mg/kg), an antioxidant cocktail (AOx), and coadministration of these 2 agents (BH4+AOx) on microvascular function in patients with HFrEF (n=14, 64±10 years) and HFpEF (n=19, 74±9 years). Passive limb movement was utilized to assess locomotor muscle microvascular function, and biomarkers of inflammation and oxidative damage were measured. RESULTS Compared with placebo, the peak change in leg blood flow was not statistically different after AOx administration (HFrEF, P=0.60; HFpEF, P=0.61), but improved following BH4 (P=0.033) and BH4+AOx (P=0.019) in both HFrEF (placebo: 234±31; BH4: 357±45; BH4+AOx: 355±49 mL/min) and HFpEF (placebo: 269±33; BH4: 367±47; BH4+AOx: 394±65 mL/min). The total hyperemic response to passive limb movement (leg blood flow area under the curve) was not statistically different across treatments in patients with HFrEF (P=0.29), but increased following BH4 (P=0.016) and BH4+AOx (P=0.040) in the HFpEF group. CRP (C-reactive protein) was lower following BH4 (P=0.007) and BH4+AOx (P=0.007) in HFpEF (placebo: 4268±547; BH4: 2721±391; BH4+AOx: 2779±376 ng/mL), but was not statistically different in HFrEF (P=0.39). CONCLUSIONS Together, these results provide new evidence for the efficacy of acute BH4 administration to improve some aspects of locomotor muscle microvascular function in patients with HFrEF and HFpEF, with no apparent benefit of AOx administration, alone or in combination with BH4, in either group. These findings lend further conceptual support for the nitric oxide pathway as a modifiable target in the treatment of heart failure.
Collapse
Affiliation(s)
- Stephen M Ratchford
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
| | - Heather L Clifton
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
| | - Jayson R Gifford
- Department of Exercise Sciences, Brigham Young University, Salt Lake City, UT (J.R.G.)
| | - D Taylor LaSalle
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| | - Taylor S Thurston
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| | - Kanokwan Bunsawat
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
| | - Jeremy K Alpenglow
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| | - Josephine B Wright
- Division of Cardiovascular Medicine (J.B.W., J.J.R.), University of Utah, Salt Lake City
| | - Markus Amann
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Anesthesiology (M.A.) University of Utah, Salt Lake City
| | - John J Ryan
- Division of Cardiovascular Medicine (J.B.W., J.J.R.), University of Utah, Salt Lake City
| | - D Walter Wray
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| |
Collapse
|
2
|
Selvaraj S, Karaj A, Chirinos JA, Denney N, Grosso G, Fernando M, Chambers K, Demastus C, Reddy R, Langham M, Kumar D, Maynard H, Pourmussa B, Prenner SB, Cohen JB, Ischiropoulos H, Rickels MR, Poole DC, Church DD, Wolfe RR, Kelly DP, Putt M, Margulies KB, Zamani P. Crossover Trial of Exogenous Ketones on Cardiometabolic Endpoints in Heart Failure With Preserved Ejection Fraction. JACC. HEART FAILURE 2025:S2213-1779(25)00237-9. [PMID: 40243975 DOI: 10.1016/j.jchf.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The etiology of exercise intolerance in heart failure with preserved ejection fraction (HFpEF) is multifactorial. Several contributing pathways may be improved by ketone ester (KE). OBJECTIVES This study aims to determine whether KE improves exercise tolerance in HFpEF. METHODS KETO-HFpEF (Ketogenic Exogenous Therapies in HFpEF) is a randomized, crossover, placebo-controlled trial of acute KE dosing in 20 symptomatic HFpEF participants. Coprimary endpoints include peak oxygen consumption (VO2) during incremental cardiopulmonary exercise testing and time to exhaustion during an additional constant-intensity exercise (75% peak workload) bout. RESULTS The average age was 71 ± 8 years, 60% were women, and 65% were White. KE did not improve peak VO2 (KE: 10.4 ± 3.6 vs placebo: 10.5 ± 4.0 mL/kg/min; P = 0.75). At rest, heart rate, biventricular systolic function, and cardiac output (0.6 L/min [95% CI: 0.3-1.0 L/min]) were greater with KE vs placebo, whereas total peripheral resistance (-3.2 WU [95% CI: -5.2 to -1.2 WU]) and the arteriovenous oxygen content difference (-0.7 mL of O2/dL blood [95% CI: -1.2 to -0.2 mL]) were lower. These differences mostly disappeared during incremental exercise. KE did not improve exercise endurance during the constant-intensity protocol (9.7 ± 7.3 minutes vs 8.7 ± 4.4 minutes; P = 0.51). In 6 participants receiving 6,6-2H2-glucose infusions during constant-intensity exercise, plasma glucose appearance rate before and during exercise was lower with KE (-0.24 mg/kg/min; P < 0.001). During both exercise protocols, KE lowered: 1) respiratory exchange ratios, demonstrating decreased systemic carbohydrate use; 2) nonesterified fatty acids and glucose; and 3) estimated left ventricular filling pressures (E/e'). CONCLUSIONS Despite robust ketosis, shifting substrate use away from carbohydrates, and decreasing estimated left ventricular filling pressures, acute KE supplementation did not improve peak VO2 or constant-intensity exercise in HFpEF. (Ketogenic Exogenous Therapies in HFpEF [KETO-HFpEF]; NCT04633460).
Collapse
Affiliation(s)
- Senthil Selvaraj
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Durham, North Carolina, USA; Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Antoneta Karaj
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Julio A Chirinos
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicole Denney
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gabby Grosso
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Melissa Fernando
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kishon Chambers
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cassandra Demastus
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ravinder Reddy
- Department of Medicine and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Langham
- Department of Medicine and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dushyant Kumar
- Department of Medicine and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hannah Maynard
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bianca Pourmussa
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stuart B Prenner
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordana B Cohen
- Division of Nephrology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Michael R Rickels
- Division of Endocrinology, Diabetes & Metabolism, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David C Poole
- Departments of Kinesiology, Anatomy, and Physiology, Kansas State University, Manhattan, Kansas, USA
| | - David D Church
- Department of Geriatrics, Donald W. Reynolds Institute on Aging, Center for Translational Research in Aging and Longevity, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Robert R Wolfe
- Department of Geriatrics, Donald W. Reynolds Institute on Aging, Center for Translational Research in Aging and Longevity, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Daniel P Kelly
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mary Putt
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kenneth B Margulies
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Payman Zamani
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
3
|
Gonzaga LA, Porto AA, Takahashi C, Gomes RL, Vanderlei LCM, Valenti VE. Acute effects of beetroot extract and resveratrol ingestion on cardiovascular and cardiac autonomic modulation recovery after moderate-intensity aerobic exercise in individuals with coronary artery disease: a triple-blinded, randomized, placebo-controlled trial. Eur J Nutr 2025; 64:67. [PMID: 39853480 DOI: 10.1007/s00394-025-03582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 01/01/2025] [Indexed: 01/26/2025]
Abstract
PURPOSE This study aimed to evaluate the acute effects of beetroot extract and resveratrol supplementation (isolated and combined) on cardiac autonomic modulation and cardiovascular parameters recovery after exercise in individuals with coronary artery disease (CAD). METHODS 14 males with CAD were submitted to 4 protocols consisting of 30 min (min) of rest, 30 min of aerobic exercise on a treadmill (60% of the heart rate reserve HRR), followed by 30 min of recovery. Before each protocol, the subjects consumed 500 mg of starch (placebo protocol), 500 mg of beetroot (beetroot protocol), or 500 mg of resveratrol (resveratrol protocol), or 500 mg of beetroot and 500 mg of resveratrol (combined protocol). Heart rate variability (HRV) indices and cardiorespiratory parameters were determined at different times during the protocols. RESULTS Regarding HR, significantly higher values about rest in the placebo protocol at all recovery moments (1st to 30th min) were observed. Significant differences were observed in the other protocols (beetroot, resveratrol and combined) from the first to the 20th min recovery. For SBP, significantly higher values concerning rest were observed at the first minute of recovery for all protocols. No differences were found for the HRV index between time and protocols. CONCLUSIONS The single supplementation of beetroot and resveratrol (isolated and combined) did not alter HRV and cardiovascular parameter responses between protocols. The consumption of beetroot extract and resveratrol enhanced vagal modulation and heart rate recovery compared to rest.
Collapse
Affiliation(s)
- Luana Almeida Gonzaga
- Postgraduate Program in Movement Sciences, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Andrey Alves Porto
- Postgraduate Program in Movement Sciences, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil.
| | - Carolina Takahashi
- Postgraduate Program in Movement Sciences, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Rayana Loch Gomes
- Faculty of Health Sciences, Federal University of Grande Dourados (UFGD), Dourados, Mato Grosso do Sul, Brazil
| | - Luiz Carlos Marques Vanderlei
- Postgraduate Program in Movement Sciences, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Vitor Engrácia Valenti
- Postgraduate Program in Movement Sciences, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
| |
Collapse
|
4
|
Billingsley HE, Carbone S, Driggin E, Kitzman DW, Hummel SL. Dietary Interventions in Heart Failure With Preserved Ejection Fraction: A Scoping Review. JACC. ADVANCES 2025; 4:101465. [PMID: 39801812 PMCID: PMC11719370 DOI: 10.1016/j.jacadv.2024.101465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025]
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) are burdened by multiple diet-sensitive comorbidities, including obesity and malnutrition. Despite this, a low percentage of patients with HFpEF have been enrolled in dietary intervention trials in heart failure and few dietary interventions have been conducted in HFpEF exclusively. This scoping review will examine available evidence regarding dietary interventions in patients with HFpEF, highlight existing gaps in knowledge, and discuss emerging dietary therapies in this population.
Collapse
Affiliation(s)
- Hayley E. Billingsley
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Salvatore Carbone
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
- Virginia Commonwealth University Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Elissa Driggin
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| | - Dalane W. Kitzman
- Sections on Cardiovascular Medicine and Geriatrics and Gerontology, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Scott L. Hummel
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cardiology, Veteran’s Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Zamani P, Shah SJ, Cohen JB, Zhao M, Yang W, Afable JL, Caturla M, Maynard H, Pourmussa B, Demastus C, Mohanty I, Miyake MM, Adusumalli S, Margulies KB, Prenner SB, Poole DC, Wilson N, Reddy R, Townsend RR, Ischiropoulos H, Cappola TP, Chirinos JA. Potassium Nitrate in Heart Failure With Preserved Ejection Fraction: A Randomized Clinical Trial. JAMA Cardiol 2024:2827840. [PMID: 39693096 DOI: 10.1001/jamacardio.2024.4417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Importance Nitric oxide deficiency may contribute to exercise intolerance in patients with heart failure with preserved ejection fraction (HFpEF). Prior pilot studies have shown improvements in exercise tolerance with single-dose and short-term inorganic nitrate administration. Objective To assess the impact of chronic inorganic nitrate administration on exercise tolerance in a larger trial of participants with HFpEF. Design, Setting, and Participants This multicenter randomized double-blinded crossover trial was conducted at the University of Pennsylvania, the Philadelphia Veterans Affairs Medical Center, and Northwestern University between October 2016 and July 2022. Participants included patients with symptomatic (New York Heart Association class II/III) HFpEF who had objective signs of elevated left ventricular filling pressures. Image quantification, physiological data modeling and biochemical measurements, unblinding, and statistical analyses were completed in 2024. Intervention Potassium nitrate (KNO3) (6 mmol 3 times daily) vs equimolar doses of potassium chloride (KCl) for 6 weeks, each with a 1-week washout in between. MAIN OUTCOMES AND MEASURES The coprimary end points included peak oxygen uptake and total work performed during a maximal effort incremental cardiopulmonary exercise test. Secondary end points included the exercise systemic vasodilatory reserve (ie, reduction in systemic vascular resistance with exercise) and quality of life assessed using the Kansas City Cardiomyopathy Questionnaire. Results Eighty-four participants were enrolled. Median age was 68 years and 58 participants were women (69.0%). Most participants had NYHA class II disease (69%) with a mean 6-minute walk distance of 335.5 (SD, 97.3) m. Seventy-seven participants received the KNO3 intervention and 74 received the KCl intervention. KNO3 increased trough levels of serum nitric oxide metabolites after 6 weeks (KNO3, 418.4 [SD, 26.9] uM vs KCl, 40.1 [SD, 28.3] uM; P < .001). KNO3 did not improve peak oxygen uptake (KNO3, 10.23 [SD, 0.43] mL/min/kg vs KCl, 10.17 [SD, 0.43] mL/min/kg; P = .73) or total work performed (KNO3, 25.9 [SD, 3.65] kilojoules vs KCl, 23.63 [SD, 3.63] kilojoules; P = .29). KNO3 nitrate did not improve the vasodilatory reserve or quality of life, though it was well-tolerated. Conclusions and Relevance In this study, potassium nitrate did not improve aerobic capacity, total work, or quality of life in participants with HFpEF. Trial Registration ClinicalTrials.gov Identifier: NCT02840799.
Collapse
Affiliation(s)
- Payman Zamani
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Sanjiv J Shah
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jordana B Cohen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Manyun Zhao
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Wei Yang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jessica L Afable
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Maria Caturla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Hannah Maynard
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Bianca Pourmussa
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | | - Ipsita Mohanty
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Michelle Menon Miyake
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | | | | | - Stuart B Prenner
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - David C Poole
- Departments of Kinesiology, Anatomy, and Physiology, Kansas State University, Manhattan
| | - Neil Wilson
- Center for Advanced Metabolic Imaging in Precision Medicine, Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Thomas P Cappola
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Julio A Chirinos
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
6
|
Derella CC, Anderson KC, Woessner MN, Paterson C, Allen JD. Ergogenic Effect of Nitrate Supplementation in Clinical Populations: A Systematic Review and Meta-Analysis. Nutrients 2024; 16:3832. [PMID: 39599618 PMCID: PMC11597481 DOI: 10.3390/nu16223832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Inorganic nitrate (NO3-) supplementation, via its conversion to nitric oxide (NO), has been purported to be ergogenic in healthy individuals. Many disease states are characterized by reduced NO bioavailability and are expected to derive a benefit from NO3-. This systematic review and meta-analysis evaluate the current literature on the ergogenic effect of NO3- supplementation in individuals with cardiopulmonary and metabolic diseases (CPMD). Methods: Relevant databases were searched up to December 2023 for randomized, placebo-controlled crossover trials for aerobic exercise outcome variables with CPMD. Results: Twenty-two studies were included, and 46% reported ergogenic benefits of inorganic nitrate supplementation. NO3- supplementation had no effect on aerobic performance with respect to maximal (SMD = 0.11, 95% CI: -0.12 to 0.34, p = 0.34) and submaximal (SMD = 0.16, 95% CI: -0.13 to 0.46, p = 0.27) TTE, VO2peak (SMD = 0.002, 95% CI: -0.37 to 0.38, p = 0.99), or 6MW (SMD = 0.01, 95% CI: -0.29 to 0.28, p = 0.96). When the studies were limited to only cardiovascular disease conditions, NO3- supplementation had trivial effects on aerobic performance with respect to Timed Trials (SMD = 0.14, 95% CI: -0.04 to 0.33, p = 0.13), VO2 (SMD = -0.02, 95% CI: -0.32 to 0.27, p = 0.87), and small effects on Distance Trials (SMD = 0.25, 95% CI: -0.18 to 0.69, p = 0.25). Sunset funnel plots revealed low statistical power in all trials. Conclusions: The results of this systematic review revealed that 46% of the individual studies showed a positive benefit from inorganic nitrate supplementation. However, the meta-analysis revealed a trivial effect on physical function in CPMD populations. This is likely due to the large heterogeneity and small sample sizes in the current literature.
Collapse
Affiliation(s)
- Cassandra C. Derella
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Kara C. Anderson
- Division of Endocrinology and Metabolism, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Mary N. Woessner
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia
| | - Craig Paterson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Jason D. Allen
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA
- Division of Cardiovascular Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
7
|
Tian S, Ding T, Li H. Oral microbiome in human health and diseases. MLIFE 2024; 3:367-383. [PMID: 39359681 PMCID: PMC11442140 DOI: 10.1002/mlf2.12136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/13/2024] [Accepted: 05/05/2024] [Indexed: 10/04/2024]
Abstract
The oral cavity contains the second-largest microbiota in the human body. The cavity's anatomically and physiologically diverse niches facilitate a wide range of symbiotic bacteria living at distinct oral sites. Consequently, the oral microbiota exhibits site specificity, with diverse species, compositions, and structures influenced by specific aspects of their placement. Variations in oral microbiota structure caused by changes in these influencing factors can impact overall health and lead to the development of diseases-not only in the oral cavity but also in organs distal to the mouth-such as cancer, cardiovascular disease, and respiratory disease. Conversely, diseases can exacerbate the imbalance of the oral microbiota, creating a vicious cycle. Understanding the heterogeneity of both the oral microbiome and individual humans is important for investigating the causal links between the oral microbiome and diseases. Additionally, understanding the intricacies of the oral microbiome's composition and regulatory factors will help identify the potential causes of related diseases and develop interventions to prevent and treat illnesses in this domain. Therefore, turning to the extant research in this field, we systematically review the relationship between oral microbiome dynamics and human diseases.
Collapse
Affiliation(s)
- Siqi Tian
- Department of Immunology and Microbiology, Zhongshan School of Medicine Sun Yat-Sen University Guangzhou China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University) Ministry of Education Guangzhou China
| | - Tao Ding
- Department of Immunology and Microbiology, Zhongshan School of Medicine Sun Yat-Sen University Guangzhou China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University) Ministry of Education Guangzhou China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University) Ministry of Education, China Guangzhou China
| | - Hui Li
- Department of Immunology and Microbiology, Zhongshan School of Medicine Sun Yat-Sen University Guangzhou China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University) Ministry of Education Guangzhou China
| |
Collapse
|
8
|
Yuschen X, Choi JH, Seo J, Sun Y, Lee E, Kim SW, Park HY. Effects of Acute Beetroot Juice Supplementation and Exercise on Cardiovascular Function in Healthy Men in Preliminary Study: A Randomized, Double-Blinded, Placebo-Controlled, and Crossover Trial. Healthcare (Basel) 2024; 12:1240. [PMID: 38998775 PMCID: PMC11241253 DOI: 10.3390/healthcare12131240] [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: 05/23/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Nitrate-rich beetroot juice (NRBRJ) can potentially enhance exercise performance and improve cardiovascular function, leading to an increased use of NRBRJ over the years. However, the combined effects of NRBRJ supplementation and exercise on cardiovascular function remain unclear. Therefore, this study compared cardiovascular function responses to submaximal exercise with either placebo (PLA) or NRBRJ supplementation in healthy men. Twelve healthy men (aged 25.2 ± 2.3 years) completed the 30-min submaximal cycle ergometer exercise trials corresponding to 70% maximal heart rate (HRmax) with either PLA or NRBRJ supplementation in a random order. The mean exercise load, heart rate (HR), stroke volume (SV), cardiac output (CO), systolic blood pressure (SBP), diastolic blood pressure (DBP), mean arterial pressure (MAP), and total peripheral resistance (TPR) were measured during exercise. The brachial-ankle pulse wave velocity (baPWV) and flow-mediated dilation (FMD) were measured before and after exercise. NRBRJ supplementation was more effective than PLA in increasing the mean exercise load and decreasing DBP and MAP during submaximal exercise. Furthermore, baPWV decreased in the NRBRJ trial and was considerably lower after exercise in the NRBRJ-supplemented group than in the PLA-supplemented group. FMD significantly increased in the PLA and NRBRJ trials; however, NRBRJ supplementation demonstrated a significantly higher FMD before and after exercise than PLA supplementation. In conclusion, acute NRBRJ supplementation and exercise were more effective than PLA supplementation and exercise in improving aerobic exercise capacity and cardiovascular function in healthy men.
Collapse
Affiliation(s)
- Xie Yuschen
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jae-Ho Choi
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jisoo Seo
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yerin Sun
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Eunjoo Lee
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sung-Woo Kim
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- Physical Activity and Performance Institute, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hun-Young Park
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- Physical Activity and Performance Institute, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
9
|
Silva-Cunha M, Lacchini R, Tanus-Santos JE. Facilitating Nitrite-Derived S-Nitrosothiol Formation in the Upper Gastrointestinal Tract in the Therapy of Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:691. [PMID: 38929130 PMCID: PMC11200996 DOI: 10.3390/antiox13060691] [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: 04/23/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are often associated with impaired nitric oxide (NO) bioavailability, a critical pathophysiological alteration in CVDs and an important target for therapeutic interventions. Recent studies have revealed the potential of inorganic nitrite and nitrate as sources of NO, offering promising alternatives for managing various cardiovascular conditions. It is now becoming clear that taking advantage of enzymatic pathways involved in nitrite reduction to NO is very relevant in new therapeutics. However, recent studies have shown that nitrite may be bioactivated in the acidic gastric environment, where nitrite generates NO and a variety of S-nitrosating compounds that result in increased circulating S-nitrosothiol concentrations and S-nitrosation of tissue pharmacological targets. Moreover, transnitrosation reactions may further nitrosate other targets, resulting in improved cardiovascular function in patients with CVDs. In this review, we comprehensively address the mechanisms and relevant effects of nitrate and nitrite-stimulated gastric S-nitrosothiol formation that may promote S-nitrosation of pharmacological targets in various CVDs. Recently identified interfering factors that may inhibit these mechanisms and prevent the beneficial responses to nitrate and nitrite therapy were also taken into consideration.
Collapse
Affiliation(s)
- Mila Silva-Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil;
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto 14040-902, Brazil;
| | - Jose E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil;
| |
Collapse
|
10
|
Alyahya AI, Charman SJ, Okwose NC, Fuller AS, Eggett C, Luke P, Bailey K, MacGowan GA, Jakovljevic DG. Lifestyle Intervention Improves Parasympathetic Activity in Hypertrophic Cardiomyopathy. Am J Lifestyle Med 2024:15598276241253187. [PMID: 39554911 PMCID: PMC11562454 DOI: 10.1177/15598276241253187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Background A limited number of studies have investigated the effects of lifestyle interventions in hypertrophic cardiomyopathy (HCM). This study evaluated the effect of a novel lifestyle intervention incorporating physical activity (PA) and dietary nitrate supplementation on heart rate variability (HRV) and haemodynamic measures in HCM. Methods Twenty-eight individuals with HCM were randomised into either the intervention or control group. Frequency-domain HRV measures including low frequency power (LF), high frequency power (HF) and LF/HF were recorded at rest using bioimpedance. Non-invasive haemodynamic variables were recorded at rest using bioreactance. Participants in the intervention group consumed 6 mmol of nitrate daily (concentrated beetroot juice) and were instructed to increase and maintain daily PA by ≥ 2000 steps/day above baseline for 16 weeks. Control group participants retained their usual lifestyle and monitored daily step counts. Results There was a significant increase in post-intervention HF power (7.54 ± 2.14 vs 8.78 ± 1.60 ms2, P < .01) and LF power (6.89 ± 2.33 vs 8.17 ± 1.55, P < .01) in the intervention but not in the control group. Resting mean arterial blood pressure (MABP) in the intervention group significantly reduced at follow-up (108 ± 6 vs 102 ± 7 mmHg, P < .01). Conclusions A novel lifestyle intervention including PA and dietary nitrate supplementation enhanced parasympathetic activity and resting MABP in HCM.
Collapse
Affiliation(s)
- Alaa I. Alyahya
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
- Department of Cardiac Technology, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (AIA)
| | - Sarah J. Charman
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
| | - Nduka C. Okwose
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
- Faculty Research Centre (CHLS), Institute for Health and Wellbeing, Faculty of Health and Life Sciences, Coventry University, Coventry, UK (NCO, DGJ, ASF)
| | - Amy S. Fuller
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
| | - Christopher Eggett
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
| | - Peter Luke
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
| | - Kristian Bailey
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
| | - Guy A. MacGowan
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK (GAM)
| | - Djordje G. Jakovljevic
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, DGJ)
- Cardiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK (AIA, SJC, NCO, ASF, CE, PL, KB, GAM, DGJ)
- Faculty Research Centre (CHLS), Institute for Health and Wellbeing, Faculty of Health and Life Sciences, Coventry University, Coventry, UK (NCO, DGJ, ASF)
| |
Collapse
|
11
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
12
|
Heefner A, Simovic T, Mize K, Rodriguez-Miguelez P. The Role of Nutrition in the Development and Management of Chronic Obstructive Pulmonary Disease. Nutrients 2024; 16:1136. [PMID: 38674827 PMCID: PMC11053888 DOI: 10.3390/nu16081136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent lung condition associated with significant morbidity and mortality. The management of COPD classically involves pulmonary rehabilitation, bronchodilators, and corticosteroids. An aspect of COPD management that is currently lacking in the literature is nutritional management, despite the prevalence of inadequate nutritional status in patients with COPD. In addition, certain nutritional imbalances have been reported to increase the risk of COPD development. This review summarizes the current literature on the role diet and nutrients may play in the risk and management of COPD development.
Collapse
Affiliation(s)
- Allison Heefner
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Tijana Simovic
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Kasey Mize
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Paula Rodriguez-Miguelez
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
13
|
Bunsawat K, Nelson MD, Hearon CM, Wray DW. Exercise intolerance in heart failure with preserved ejection fraction: Causes, consequences and the journey towards a cure. Exp Physiol 2024; 109:502-512. [PMID: 38063130 PMCID: PMC10984794 DOI: 10.1113/ep090674] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/22/2023] [Indexed: 04/04/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for over 50% of all heart failure cases nationwide and continues to rise in its prevalence. The complex, multi-organ involvement of the HFpEF clinical syndrome requires clinicians and investigators to adopt an integrative approach that considers the contribution of both cardiac and non-cardiac function to HFpEF pathophysiology. Thus, this symposium review outlines the key points from presentations covering the contributions of disease-related changes in cardiac function, arterial stiffness, peripheral vascular function, and oxygen delivery and utilization to exercise tolerance in patients with HFpEF. While many aspects of HFpEF pathophysiology remain poorly understood, there is accumulating evidence for a decline in vascular health in this patient group that may be remediable through pharmacological and lifestyle interventions and could improve outcomes and clinical status in this ever-growing patient population.
Collapse
Affiliation(s)
- Kanokwan Bunsawat
- Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical CenterSalt Lake CityUtahUSA
- Department of Internal Medicine, Division of GeriatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Michael D. Nelson
- Department of KinesiologyUniversity of Texas at ArlingtonArlingtonTexasUSA
| | - Christopher M. Hearon
- Department of Applied Clinical ResearchThe University of Texas Southwestern Medical CenterDallasTexasUSA
| | - D. Walter Wray
- Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical CenterSalt Lake CityUtahUSA
- Department of Internal Medicine, Division of GeriatricsUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
14
|
Olas B. The Cardioprotective Role of Nitrate-Rich Vegetables. Foods 2024; 13:691. [PMID: 38472804 DOI: 10.3390/foods13050691] [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: 01/16/2024] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Nitric oxide (NO) is an inorganic radical produced by both the non-enzymatic nitrate (NO3-)-nitrite (NO2-)-NO pathway and enzymatic reactions catalyzed by nitric oxide synthase (NOS). Also, as nitrate and nitrite from dietary and other endogenous sources can be reduced back to nitric oxide in vivo, the endogenous NO level can be increased through the consumption of nitrate-rich vegetables. Ingestion of dietary NO3- has beneficial effects which have been attributed to a subsequent increase in NO: a signaling molecule that may regulate various systems, including the cardiovascular system. A diet rich in NO3- from green leafy and root vegetables has cardioprotective effects, with beetroot products being particularly good sources of NO3-. For example, various studies have demonstrated a significant increase in nitrite levels (regarded as markers of NO) in plasma after the intake of beetroot juice. The present review describes the current literature concerning the role of nitrate-rich vegetables (especially beetroot products) in the prophylaxis and treatment of cardiovascular diseases (CVDs). This review is based on studies identified in electronic databases, including PubMed, ScienceDirect, Web of Knowledge, Sci Finder, Web of Science, and SCOPUS.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
15
|
Dillon KN, Kang Y, Maharaj A, Martinez MA, Fischer SM, Figueroa A. L-Citrulline supplementation attenuates aortic pressure and pressure waves during metaboreflex activation in postmenopausal women. Br J Nutr 2024; 131:474-481. [PMID: 37664994 DOI: 10.1017/s000711452300199x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Postmenopausal women have augmented pressure wave responses to low-intensity isometric handgrip exercise (IHG) due to an overactive metaboreflex (postexercise muscle ischaemia, PEMI), contributing to increased aortic systolic blood pressure (SBP). Menopause-associated endothelial dysfunction via arginine (ARG) and nitric oxide deficiency may contribute to exaggerated exercise SBP responses. L-Citrulline supplementation (CIT) is an ARG precursor that decreases SBP, pulse pressure (PP) and pressure wave responses to cold exposure in older adults. We investigated the effects of CIT on aortic SBP, PP, and pressure of forward (Pf) and backward (Pb) waves during IHG and PEMI in twenty-two postmenopausal women. Participants were randomised to CIT (10 g/d) or placebo (PL) for 4 weeks. Aortic haemodynamics were assessed via applanation tonometry at rest, 2 min of IHG at 30 % of maximal strength, and 3 min of PEMI. Responses were analysed as change (Δ) from rest to IHG and PEMI at 0 and 4 weeks. CIT attenuated ΔSBP (−9 ± 2 v. −1 ± 1 mmHg, P = 0·006), ΔPP (−5 ± 2 v. 0 ± 1 mmHg, P = 0·03), ΔPf (−6 ± 2 v. −1 ± 1 mmHg, P = 0·01) and ΔPb (−3 ± 1 v. 0 ± 1 mmHg, P = 0·02) responses to PEMI v. PL. The ΔPP during PEMI was correlated with ΔPf (r = 0·743, P < 0·001) and ΔPb (r = 0·724, P < 0·001). Citrulline supplementation attenuates the increase in aortic pulsatile load induced by muscle metaboreflex activation via reductions in forward and backward pressure wave amplitudes in postmenopausal women.
Collapse
Affiliation(s)
- Katherine N Dillon
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX79409, USA
| | - Yejin Kang
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX79409, USA
| | - Arun Maharaj
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX79409, USA
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mauricio A Martinez
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX79409, USA
| | - Stephen M Fischer
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX79409, USA
| | - Arturo Figueroa
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX79409, USA
| |
Collapse
|
16
|
Benjamim CJR, da Silva LSL, Sousa YBA, Rodrigues GDS, Pontes YMDM, Rebelo MA, Gonçalves LDS, Tavares SS, Guimarães CS, da Silva Sobrinho AC, Tanus-Santos JE, Gualano B, Bueno Júnior CR. Acute and short-term beetroot juice nitrate-rich ingestion enhances cardiovascular responses following aerobic exercise in postmenopausal women with arterial hypertension: A triple-blinded randomized controlled trial. Free Radic Biol Med 2024; 211:12-23. [PMID: 38092272 DOI: 10.1016/j.freeradbiomed.2023.11.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/19/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND The increase in blood pressure (BP) levels in the postmenopausal period can be partly explained by the decrease in nitric oxide synthases (NOS). OBJECTIVE To investigate the acute and one-week effects of beetroot juice nitrate-rich (BRJ-NO3-rich) ingestion on cardiovascular and autonomic performance in response to submaximal aerobic exercise in postmenopausal women with systemic arterial hypertension (SAH) who are physically inactive. METHODS Fourteen postmenopausal women with SAH [mean (SD) age: 59(4) y; BMI (kg/m2): 29.2(3.1)] completed submaximal aerobic exercise bouts after an acute and a one-week intervention with BRJ in a placebo-controlled, randomized, triple-blind, crossover design. Participants ingested either BRJ (800 mg of NO3-) or placebo acutely and drank either BRJ (400 mg of NO3-) or placebo every day for the next six days. After two and ½ hours, they performed a session of aerobic submaximal aerobic exercise, and their systolic BP (SBP) and diastolic BP (DBP), flow-mediated dilation (FMD), heart rate (HR) recovery, and HR variability were measured. RESULTS In the post-exercise recovery period, SBP dropped significantly in the BRJ-NO3-rich group (-9.28 mmHg [95%CI: -1.68 to -16.88] ES: -0.65, p = 0.019) compared to placebo after acute ingestion. The FMD values increased after acute BRJ-NO3-rich on post-exercise (3.18 % [0.36 to 5.99] ES: 0.87, p = 0.031). After the one-week intervention, FMD values were higher in the BRJ-NO3-rich group before (4.5 % [1.62 to 7.37] ES: 1.21, p = 0.005) and post-exercise measurements (4.2 % [1.52 to 6.87] ES: 1.22, p = 0.004) vs. placebo. HRV indices with remarkable parasympathetic modulation to heart recovered faster on the BRJ-NO3-rich group than placebo group. No between-group differences were identified in values of HR post-exercise recovery in the 30s, 60s, 120s, 180s, and 300s. CONCLUSIONS Acute and short-term BRJ-NO3-rich ingestion may enhance cardiovascular and autonomic behavior in response to aerobic exercise in postmenopausal women diagnosed with SAH. CLINICAL TRIAL REGISTRY NUMBER https://clinicaltrials.gov/ct2/show/NCT05384340.
Collapse
Affiliation(s)
- Cicero Jonas R Benjamim
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto, São Paulo, Brazil.
| | - Leonardo Santos L da Silva
- School of Physical Education of Ribeirão Preto, University of São Paulo (EEFERP/USP) Ribeirão Preto, Brazil
| | - Yaritza B Alves Sousa
- Department of Health Sciences, Ribeirao Preto Medical School, University of Sao Paulo, USP, Ribeirao Preto, Sao Paulo, Brazil
| | - Guilherme da Silva Rodrigues
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto, São Paulo, Brazil
| | | | - Macario Arosti Rebelo
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil
| | | | - Simone Sakagute Tavares
- School of Physical Education of Ribeirão Preto, University of São Paulo (EEFERP/USP) Ribeirão Preto, Brazil
| | - Carolina S Guimarães
- Ribeirao Preto Nursing School, University of São Paulo (EERP/USP), Ribeirão Preto, SP, Brazil
| | - Andressa C da Silva Sobrinho
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto, São Paulo, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto, SP, Brazil
| | - Bruno Gualano
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, University of São Paulo, Medical School (FMUSP), São Paulo, SP, Brazil
| | - Carlos R Bueno Júnior
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto, São Paulo, Brazil; School of Physical Education of Ribeirão Preto, University of São Paulo (EEFERP/USP) Ribeirão Preto, Brazil; Ribeirao Preto Nursing School, University of São Paulo (EERP/USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
17
|
Borlaug BA, Koepp KE, Reddy YNV, Obokata M, Sorimachi H, Freund M, Haberman D, Sweere K, Weber KL, Overholt EA, Safe BA, Omote K, Omar M, Popovic D, Acker NG, Gladwin MT, Olson TP, Carter RE. Inorganic Nitrite to Amplify the Benefits and Tolerability of Exercise Training in Heart Failure With Preserved Ejection Fraction: The INABLE-Training Trial. Mayo Clin Proc 2024; 99:206-217. [PMID: 38127015 PMCID: PMC10872737 DOI: 10.1016/j.mayocp.2023.08.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE To determine whether nitrite can enhance exercise training (ET) effects in heart failure with preserved ejection fraction (HFpEF). METHODS In this multicenter, double-blind, placebo-controlled, randomized trial conducted at 1 urban and 9 rural outreach centers between November 22, 2016, and December 9, 2021, patients with HFpEF underwent ET along with inorganic nitrite 40 mg or placebo 3 times daily. The primary end point was peak oxygen consumption (VO2). Secondary end points included Kansas City Cardiomyopathy Questionnaire overall summary score (KCCQ-OSS, range 0 to 100; higher scores reflect better health status), 6-minute walk distance, and actigraphy. RESULTS Of 92 patients randomized, 73 completed the trial because of protocol modifications necessitated by loss of drug availability. Most patients were older than 65 years (80%), were obese (75%), and lived in rural settings (63%). At baseline, median peak VO2 (14.1 mL·kg-1·min-1) and KCCQ-OSS (63.7) were severely reduced. Exercise training improved peak VO2 (+0.8 mL·kg-1·min-1; 95% CI, 0.3 to 1.2; P<.001) and KCCQ-OSS (+5.5; 95% CI, 2.5 to 8.6; P<.001). Nitrite was well tolerated, but treatment with nitrite did not affect the change in peak VO2 with ET (nitrite effect, -0.13; 95% CI, -1.03 to 0.76; P=.77) or KCCQ-OSS (-1.2; 95% CI, -7.2 to 4.9; P=.71). This pattern was consistent across other secondary outcomes. CONCLUSION For patients with HFpEF, ET administered for 12 weeks in a predominantly rural setting improved exercise capacity and health status, but compared with placebo, treatment with inorganic nitrite did not enhance the benefit from ET. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02713126.
Collapse
Affiliation(s)
- Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN.
| | - Katlyn E Koepp
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Yogesh N V Reddy
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Masaru Obokata
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Hidemi Sorimachi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Monique Freund
- Mayo Clinic Community Cardiology Southwest Wisconsin, La Crosse
| | - Doug Haberman
- Mayo Clinic Community Cardiology Southwest Wisconsin, La Crosse
| | - Kara Sweere
- Mayo Clinic Community Cardiology Southeast Minnesota, Albert Lea
| | - Kari L Weber
- Mayo Clinic Community Cardiology Southeast Minnesota, Austin
| | | | - Bethany A Safe
- Mayo Clinic Community Cardiology Southeast Minnesota, Red Wing
| | - Kazunori Omote
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Massar Omar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Dejana Popovic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Nancy G Acker
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Mark T Gladwin
- Department of Medicine, Maryland School of Medicine, Baltimore
| | - Thomas P Olson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Rickey E Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
18
|
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
|
19
|
Boulmpou A, Boutou AK, Pella E, Sarafidis P, Papadopoulos CE, Vassilikos V. Cardiopulmonary Exercise Testing in Heart Failure With Preserved Ejection Fraction: Technique Principles, Current Evidence, and Future Perspectives. Cardiol Rev 2023; 31:299-317. [PMID: 36723460 DOI: 10.1097/crd.0000000000000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifactorial clinical syndrome involving a rather complex pathophysiologic substrate and quite a challenging diagnosis. Exercise intolerance is a major feature of HFpEF, and in many cases, diagnosis is suspected in subjects presenting with exertional dyspnea. Cardiopulmonary exercise testing (CPET) is a noninvasive, dynamic technique that provides an integrative evaluation of cardiovascular, pulmonary, hematopoietic, neuropsychological, and metabolic functions during maximal or submaximal exercise. The assessment is based on the principle that system failure typically occurs when the system is under stress, and thus, CPET is currently considered to be the gold standard for identifying exercise intolerance, allowing the differential diagnosis of underlying causes. CPET is used in observational studies and clinical trials in HFpEF; however, in most cases, only a few from a wide variety of CPET parameters are examined, while the technique is largely underused in everyday cardiology practice. This article discusses the basic principles and methodology of CPET and studies that utilized CPET in patients with HFpEF, in an effort to increase awareness of CPET capabilities among practicing cardiologists.
Collapse
Affiliation(s)
- Aristi Boulmpou
- From the Third Department of Cardiology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Afroditi K Boutou
- Department of Respiratory Medicine, G. Papanikolaou Hospital, Thessaloniki, Greece
| | - Eva Pella
- Department of Nephrology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pantelis Sarafidis
- Department of Nephrology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christodoulos E Papadopoulos
- From the Third Department of Cardiology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vassilios Vassilikos
- From the Third Department of Cardiology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
20
|
Forsyth F, Mulrennan S, Burt J, Hartley P, Kuhn I, Lin H, Mant J, Tan S, Zhang R, Deaton C. What are the outcomes of dietary interventions in Heart Failure with preserved Ejection Fraction? A systematic review and meta-analysis. Eur J Cardiovasc Nurs 2023; 22:679-689. [PMID: 36453073 DOI: 10.1093/eurjcn/zvac114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 10/12/2023]
Abstract
AIMS To determine the efficacy of dietary interventions in Heart Failure with preserved Ejection Fraction (HFpEF). METHOD AND RESULTS Keyword searches were performed in five bibliographic databases to identify randomized or controlled studies of dietary interventions conducted in HFpEF or mixed heart failure (HF) samples published in the English language. Studies were appraised for bias and synthesized into seven categories based on the similarity of the intervention or targeted population. The quality of the body of evidence was assessed via the GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) framework. Twenty-five unique interventions were identified; 17 were considered for meta-analysis. Most studies were judged to be at high risk of bias. There was moderate-quality evidence that caloric restriction led to clinically meaningful improvements in blood pressure and body weight. There was moderate-quality evidence that carbohydrate restriction resulted in meaningful reductions in blood pressure. There was very low-quality evidence that protein supplementation improved blood pressure and body weight and moderate-quality evidence for clinically meaningful improvements in function. CONCLUSIONS While some types of dietary interventions appeared to deliver clinically meaningful change in critical outcomes; the study heterogeneity and overall quality of the evidence make it difficult to make firm recommendations. Greater transparency when reporting the nutritional composition of interventions would enhance the ability to pool studies. REGISTRATION PROSPERO CRD42019145388.
Collapse
Affiliation(s)
- Faye Forsyth
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 OSR, UK
| | - Sandra Mulrennan
- Department of Cardiology, Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0AY, UK
| | - Jenni Burt
- The Healthcare Improvement Studies Institute, Department of Public Health & Primary Care, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 OSR, UK
| | - Peter Hartley
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 OSR, UK
- Physiotherapy Department, Cambridge University Hospital NHS Foundation Trust, Addenbrookes, Cambridge CB2 OQQ, UK
| | - Isla Kuhn
- Medical Library, University of Cambridge, Cambridge University Hospital NHS Foundation Trust, Addenbrookes, Cambridge CB2 OQQ, UK
| | - Helen Lin
- University of Cambridge School of Clinical Medicine, Cambridge University Hospital NHS Foundation Trust, Addenbrookes, Cambridge CB2 OQQ, UK
| | - Jonathan Mant
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 OSR, UK
| | - Sapphire Tan
- University of Cambridge School of Clinical Medicine, Cambridge University Hospital NHS Foundation Trust, Addenbrookes, Cambridge CB2 OQQ, UK
| | - Roy Zhang
- University of Cambridge School of Clinical Medicine, Cambridge University Hospital NHS Foundation Trust, Addenbrookes, Cambridge CB2 OQQ, UK
| | - Christi Deaton
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 OSR, UK
| |
Collapse
|
21
|
Mollace R, Scarano F, Bava I, Carresi C, Maiuolo J, Tavernese A, Gliozzi M, Musolino V, Muscoli S, Palma E, Muscoli C, Salvemini D, Federici M, Macrì R, Mollace V. Modulation of the nitric oxide/cGMP pathway in cardiac contraction and relaxation: Potential role in heart failure treatment. Pharmacol Res 2023; 196:106931. [PMID: 37722519 DOI: 10.1016/j.phrs.2023.106931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Evidence exists that heart failure (HF) has an overall impact of 1-2 % in the global population being often associated with comorbidities that contribute to increased disease prevalence, hospitalization, and mortality. Recent advances in pharmacological approaches have significantly improved clinical outcomes for patients with vascular injury and HF. Nevertheless, there remains an unmet need to clarify the crucial role of nitric oxide/cyclic guanosine 3',5'-monophosphate (NO/cGMP) signalling in cardiac contraction and relaxation, to better identify the key mechanisms involved in the pathophysiology of myocardial dysfunction both with reduced (HFrEF) as well as preserved ejection fraction (HFpEF). Indeed, NO signalling plays a crucial role in cardiovascular homeostasis and its dysregulation induces a significant increase in oxidative and nitrosative stress, producing anatomical and physiological cardiac alterations that can lead to heart failure. The present review aims to examine the molecular mechanisms involved in the bioavailability of NO and its modulation of downstream pathways. In particular, we focus on the main therapeutic targets and emphasize the recent evidence of preclinical and clinical studies, describing the different emerging therapeutic strategies developed to counteract NO impaired signalling and cardiovascular disease (CVD) development.
Collapse
Affiliation(s)
- Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Saverio Muscoli
- Division of Cardiology, Foundation PTV Polyclinic Tor Vergata, Rome 00133, Italy
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy.
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Renato Dulbecco Institute, Lamezia Terme, Catanzaro 88046, Italy.
| |
Collapse
|
22
|
Zhang Z, Zhang K, Sun Y, Yu B, Tan X, Lu Y, Wang Y, Xia F, Wang N. Sweetened beverages and incident heart failure. Eur J Prev Cardiol 2023; 30:1361-1370. [PMID: 37178176 DOI: 10.1093/eurjpc/zwad167] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/15/2023]
Abstract
AIMS Recent studies have demonstrated the associations of the consumption of different beverages with cardiometabolic diseases, whereas no studies have investigated such associations in heart failure (HF). Thus, this study aimed to explore the associations of the consumption of sugar-sweetened beverages (SSBs), artificially sweetened beverages (ASBs), and pure fruit/vegetable juices (PJs) with the risk of incident HF. METHODS AND RESULTS This prospective cohort study included 209 829 participants in the UK Biobank who completed at least one 24-h diet questionnaire and who were free of baseline HF. Cox proportional hazard models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs). During a median follow-up of 9.9 years, 4328 incident HF cases were recorded. Compared to corresponding non-consumers, individuals who consumed >2 L/week SSBs or ASBs had an increased risk of HF (HR: 1.22, 95% CI: 1.08-1.38 and HR: 1.30, 95% CI: 1.16-1.47, respectively) in the multivariate adjusted model. An inverse association was observed between the consumption of >0-1 L/week PJs and the risk of HF (HR, 0.90; 95% CI, 0.83-0.98). Additionally, a significant interaction was observed between PJ consumption and sleep duration on HF risk (P for interaction = 0.030). CONCLUSIONS Increased consumption of SSBs or ASBs may be an independent risk factor for HF, whereas moderate intake of PJs may have a protective effect on HF.
Collapse
Affiliation(s)
- Ziteng Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| | - Ying Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| | - Bowei Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| | - Xiao Tan
- School of Public Health, Zhejiang University, Hangzhou, China
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| | - Yu Wang
- Department of Cardiology, Shidong Hospital affiliated to University of Shanghai for Science and Technology, Shidong Hospital, Yangpu District, 999 Shiguang Road, Shanghai 200438, China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Zhizaoju Road No.639, Huangpu, Shanghai 200011, China
| |
Collapse
|
23
|
Gröne M, Duse DA, Kramser N, Ophoff N, Schweers H, Voß F, Quast C, Sansone R, Heiss C, Jung C, Kelm M, Erkens R. Cocoa flavanols improve peakVO 2 and exercise capacity in a randomized double blinded clinical trial in healthy elderly people. Food Funct 2023; 14:7562-7573. [PMID: 37526943 DOI: 10.1039/d3fo01737k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Background: Loss of functional capacity is one of the hallmarks in cardiovascular aging. Cocoa flavanols (CF) exert favorable effects on endothelial function, blood pressure, and inflammation. These cardiovascular health markers worsen with increasing age and limit functional exercise capacity. Aim: To investigate the effect of CF on cardiorespiratory-fitness in healthy elderly people. Methods: In a randomized, double-masked, placebo-controlled, parallel-group dietary intervention trial, 68 healthy elderly people (55-79 years, 28 female) received either 500 mg of CF or a nutrient-matched control capsule twice a day for 30 days. Primary endpoint was defined as peak oxygen consumption (VO2) in a cardiopulmonary exercise test (CPET). Secondary endpoints were oxygen pulse (VO2 per heart rate (HR)), resting blood pressure (BP), and resting vascular function. Results: After 30 days of CF intake peakVO2 increased by 190 ml min-1 (95% CI 1-371 ml min-1) and peakVO2 per kg by 2.5 ml (min kg)-1 (95% CI 0.30-4.2 ml (min kg)-1). O2-pulse increased by 1.7 ml (95% CI 0.29-3.2 ml) and max exercise capacity by 9.6 W (95% CI 2.1-17.7 W). CF decreased resting systolic and diastolic BP by 5.4 mmHg (95% CI -10.7 to -0.1 mmHg) and 2.9 mmHg (95% CI -5.5 to -0.4 mmHg), respectively. Flow-mediated vasodilation (FMD) increased by an absolute 1.3% (95% CI 0.76-1.79%) in the CF group. Indexes of pulmonary function were not affected. No changes for primary and secondary endpoints were detected in control. Conclusion: CF substantially improve markers of cardiorespiratory fitness in healthy elderly humans highlighting their potential to preserve cardiovascular health with increasing age.
Collapse
Affiliation(s)
- Michael Gröne
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Dragos Andrei Duse
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Nicolas Kramser
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Niklas Ophoff
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Hendrik Schweers
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Fabian Voß
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Christine Quast
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Roberto Sansone
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Christian Heiss
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, UK
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | - Ralf Erkens
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
24
|
Vaz-Salvador P, Adão R, Vasconcelos I, Leite-Moreira AF, Brás-Silva C. Heart Failure with Preserved Ejection Fraction: a Pharmacotherapeutic Update. Cardiovasc Drugs Ther 2023; 37:815-832. [PMID: 35098432 PMCID: PMC8801287 DOI: 10.1007/s10557-021-07306-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 02/06/2023]
Abstract
While guidelines for management of heart failure with reduced ejection fraction (HFrEF) are consensual and have led to improved survival, treatment options for heart failure with preserved ejection fraction (HFpEF) remain limited and aim primarily for symptom relief and improvement of quality of life. Due to the shortage of therapeutic options, several drugs have been investigated in multiple clinical trials. The majority of these trials have reported disappointing results and have suggested that HFpEF might not be as simply described by ejection fraction as previously though. In fact, HFpEF is a complex clinical syndrome with various comorbidities and overlapping distinct phenotypes that could benefit from personalized therapeutic approaches. This review summarizes the results from the most recent phase III clinical trials for HFpEF and the most promising drugs arising from phase II trials as well as the various challenges that are currently holding back the development of new pharmacotherapeutic options for these patients.
Collapse
Affiliation(s)
- Pedro Vaz-Salvador
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Adão
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Inês Vasconcelos
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Rua Do Campo Alegre, 823 4150-180 Porto, Portugal
| |
Collapse
|
25
|
Nakamura M. Gut Microbiome: An Effector of Dietary Nitrate That Inhibits Cardiometabolic Disease? Diabetes 2023; 72:835-837. [PMID: 37339354 PMCID: PMC10281812 DOI: 10.2337/dbi23-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/04/2023] [Indexed: 06/22/2023]
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ
| |
Collapse
|
26
|
Wong SA, Drovandi A, Jones R, Golledge J. Effect of Dietary Supplements Which Upregulate Nitric Oxide on Walking and Quality of Life in Patients with Peripheral Artery Disease: A Meta-Analysis. Biomedicines 2023; 11:1859. [PMID: 37509499 PMCID: PMC10376856 DOI: 10.3390/biomedicines11071859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
This systematic review pooled evidence from randomised controlled trials (RCTs) on the effectiveness of dietary upregulators of nitric oxide (NO) in improving the walking and quality of life of patients with peripheral artery disease (PAD). RCTs examining the effect of dietary upregulators of NO in patients with PAD were included. The primary outcome was the maximum walking distance. Secondary outcomes were the initial claudication distance, the six-minute walking distance, quality of life, the ankle-brachial pressure index (ABI), adverse events and risk of mortality, revascularisation or amputation. Meta-analyses were performed using random effects models. The risk of bias was assessed using Cochrane's ROB-2 tool. Leave-one-out and subgroup analyses were conducted to assess the effect of individual studies, the risk of bias and intervention type on pooled estimates. Thirty-four RCTs involving 3472 participants were included. Seven trials tested NO donors, nineteen tested antioxidants, three tested NO synthase inducers and five tested enhancers of NO availability. Overall, the dietary supplements significantly improved the initial claudication (SMD 0.34; 95%CI 0.04, 0.64; p = 0.03) but not maximum walking (SMD 0.13; 95%CI -0.17, 0.43; p = 0.39) distances. Antioxidant supplements significantly increased both the maximum walking (SMD 0.36; 95%CI 0.14, 0.59; p = 0.001) and initial claudication (SMD 0.58; 95%CI 0.26, 0.90; p < 0.001) distances. The dietary interventions did not improve the physical function domain of the Short Form-36 (SMD -0.16; 95%CI -0.32, 0.00; p = 0.38), ABI or risk of adverse events, mortality, revascularisation or amputation. Dietary NO upregulators, especially antioxidants, appear to improve the initial claudication distance in patients with PAD. Larger high-quality RCTs are needed to fully examine the benefits and risks of these treatments. PROSPERO Registration: CRD42022256653.
Collapse
Affiliation(s)
- Shannon A Wong
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - Aaron Drovandi
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - Rhondda Jones
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
- The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD 4814, Australia
| |
Collapse
|
27
|
Forsyth F, Mulrennan S, Burt J, Hartley P, Kuhn I, Lin H, Mant J, Tan S, Zhang R, Deaton C. What dietary interventions have been tested in heart failure with preserved ejection fraction? A systematic scoping review. Eur J Cardiovasc Nurs 2023; 22:126-140. [PMID: 35816028 DOI: 10.1093/eurjcn/zvac062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022]
Abstract
AIMS To determine what dietary interventions have been tested in heart failure with preserved ejection fraction (HFpEF), the modulation method, and outcomes employed and to summarize any evidence for benefit. METHODS AND RESULTS We performed key word searches in five bibliographic databases from 2001 to 2021, to identify randomized or experimental dietary interventions tested in HFpEF or mixed heart failure (HF) samples. Study characteristics were summarized according to population, intervention, comparator, outcome categories and intervention complexity was assessed. Twenty-five clinical investigations were retrieved; only 10 (40%) were conducted exclusively in HFpEF; the remainder enrolled mixed HF samples. Most studies employed either highly tailored prescribed diets (n = 12, 48%) or dietary supplementation (n = 10, 40%) modalities. Dietary pattern interventions (n = 3, 12%) are less well represented in the literature. CONCLUSION Heterogeneity made pooling studies challenging. Better reporting of baseline characteristics and the use of standardized HF lexicon would ensure greater confidence in interpretation of studies involving mixed HF populations. The field would benefit greatly from explicit reporting of the biological mechanism of action (e.g. the causal pathway) that an intervention is designed to modulate so that studies can be synthesized via their underlying mechanism of action by which diet may affect HF. An extension of the current set of core outcomes proposed by the European Society of Cardiology Heart Failure Association would ensure dietary clinical endpoints are more consistently defined and measured. REGISTRATION PROSPERO: CRD42019145388.
Collapse
Affiliation(s)
- Faye Forsyth
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
| | - Sandra Mulrennan
- Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0AY, UK
| | - Jenni Burt
- The Healthcare Improvement Studies Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Peter Hartley
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
- Physiotherapy Department, Cambridge University Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Isla Kuhn
- Medical Library, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Helen Lin
- University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Jonathan Mant
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
| | - Sapphire Tan
- University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Roy Zhang
- University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Christi Deaton
- Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
| |
Collapse
|
28
|
Lv F, Zhang J, Tao Y. Efficacy and safety of inorganic nitrate/nitrite supplementary therapy in heart failure with preserved ejection fraction. Front Cardiovasc Med 2023; 10:1054666. [PMID: 36818337 PMCID: PMC9932197 DOI: 10.3389/fcvm.2023.1054666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
Background Approximately half of patients with heart failure have a preserved ejection fraction (HFpEF). To date, only SGLT-2i, ARNi, and MRAs treatments have been shown to be effective for HFpEF. Exercise intolerance is the primary clinical feature of HFpEF. The aim of this meta-analysis was to explore the effect of inorganic nitrate/nitrite supplementary therapy on the exercise capacity of HFpEF patients. Methods We searched PubMed, Embase, Cochrane Library, OVID, and Web of Science for eligible studies for this meta-analysis. The primary outcomes were peak oxygen consumption (peak VO2), exercise time, and respiratory exchange ratio (RER) during exercise. The secondary outcomes were cardiac output, heart rate, systolic blood pressure, diastolic blood pressure, mean arterial pressure, and systemic vascular resistance during rest and exercise, respectively. Results A total of eight randomized-controlled trials were enrolled for this meta-analysis. We found no benefit of inorganic nitrate/nitrite on exercise capacity in patients with HFpEF. Inorganic nitrate/nitrite compared to placebo, did not significantly increased peak VO2 (MD = 0.361, 95% CI = -0.17 to 0.89, p = 0.183), exercise time (MD = 9.74, 95% CI = -46.47 to 65.95, p = 0.734), and respiratory exchange ratio during exercise (MD = -0.003, 95% CI = -0.036 to 0.029, p = 0.834). Among the six diameters reflecting cardiac and artery hemodynamics, inorganic nitrate/nitrite can lower rest SBP, rest/exercise DBP, rest/exercise MAP, and exercise SVR, but has no effect in cardiac output and heart rate for HFpEF patients. Conclusion Our meta-analysis suggested that inorganic nitrate/nitrite supplementary therapy has no benefit in improving the exercise capacity of patients with HFpEF, but can yield a blood pressure lowering effect, especially during exercise.
Collapse
Affiliation(s)
- Feng Lv
- Department of Cardiology, Shengzhou People’s Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou City, Zhejiang Province, China
| | - Junyi Zhang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| | - Yuan Tao
- Department of Cardiology, Shengzhou People’s Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou City, Zhejiang Province, China,*Correspondence: Yuan Tao,
| |
Collapse
|
29
|
Ratchford SM, Bunsawat K, Alpenglow JK, Zhao J, Wright JB, Ryan JJ, Wray DW. Improved vascular function and functional capacity following l-citrulline administration in patients with heart failure with preserved ejection fraction: a single-arm, open-label, prospective pilot study. J Appl Physiol (1985) 2023; 134:328-338. [PMID: 36476159 PMCID: PMC9886346 DOI: 10.1152/japplphysiol.00445.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
There is accumulating evidence for both peripheral vascular dysfunction and impaired functional capacity in patients with heart failure with a preserved ejection fraction (HFpEF). Although derangements in the l-arginine-nitric oxide (l-Arg-NO) pathway are likely to contribute to these aspects of HFpEF pathophysiology, the impact of increased NO substrate on vascular health and physical capacity has not been evaluated in this patient population. Thus, using a single-arm study design, we evaluated the impact of enteral l-citrulline (l-Cit, 6 g/day for 7 days), a precursor for l-Arg biosynthesis, on vascular function [flow-mediated dilation (FMD), reactive hyperemia (RH), and passive limb movement (PLM)], functional capacity [6-min walk test (6MWT)], and biomarkers of l-Arg-NO signaling in 14 patients with HFpEF (n = 14, 4 M/10 F, 70 ± 10 yr, EF: 66 ± 7%). Compared with baseline (0d), 7 days of l-Cit administration improved FMD (0d: 2.5 ± 1.6%, 7d: 4.5 ± 2.9%), RH (0d: 468 ± 167 mL, 7d: 577 ± 199 mL), PLM blood flow area-under-the-curve (0d: 139 ± 130 mL, 7d: 198 ± 115 mL), and 6MWT distance (0d: 377 ± 27 m, 7d: 397 ± 27 m) (P < 0.05). An increase in plasma l-Cit (0d: 42 ± 11 µM/L, 7d: 369 ± 201 µM/L), l-Arg (0d: 65 ± 8 µM/L, 7d: 257 ± 25 µM/L), and the ratio of l-Arg to asymmetric dimethylarginine (ADMA) (0d: 136 ± 13 AU, 7d: 481 ± 49 AU) (P < 0.05) was also observed. Though preliminary in nature, these functional and biomarker assessments demonstrate a potential benefit of l-Cit administration in patients with HFpEF, findings that provide new insight into the mechanisms that govern vascular and physical dysfunction in this patient group.NEW & NOTEWORTHY The current investigation has demonstrated that l-Cit administration may improve brachial artery endothelium-dependent vasodilation, upper and lower limb microvascular function, and physical capacity in patients with HFpEF, highlighting the potential therapeutic potential of interventions targeting the l-Arg-NO signaling cascade to improve outcomes in this patient group.
Collapse
Affiliation(s)
- Stephen M Ratchford
- Geriatric Research, Education, and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Kanokwan Bunsawat
- Geriatric Research, Education, and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Jeremy K Alpenglow
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Jia Zhao
- Geriatric Research, Education, and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Josephine B Wright
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - John J Ryan
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - D Walter Wray
- Geriatric Research, Education, and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
30
|
Targeting mitochondrial impairment for the treatment of cardiovascular diseases: From hypertension to ischemia-reperfusion injury, searching for new pharmacological targets. Biochem Pharmacol 2023; 208:115405. [PMID: 36603686 DOI: 10.1016/j.bcp.2022.115405] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Mitochondria and mitochondrial proteins represent a group of promising pharmacological target candidates in the search of new molecular targets and drugs to counteract the onset of hypertension and more in general cardiovascular diseases (CVDs). Indeed, several mitochondrial pathways result impaired in CVDs, showing ATP depletion and ROS production as common traits of cardiac tissue degeneration. Thus, targeting mitochondrial dysfunction in cardiomyocytes can represent a successful strategy to prevent heart failure. In this context, the identification of new pharmacological targets among mitochondrial proteins paves the way for the design of new selective drugs. Thanks to the advances in omics approaches, to a greater availability of mitochondrial crystallized protein structures and to the development of new computational approaches for protein 3D-modelling and drug design, it is now possible to investigate in detail impaired mitochondrial pathways in CVDs. Furthermore, it is possible to design new powerful drugs able to hit the selected pharmacological targets in a highly selective way to rescue mitochondrial dysfunction and prevent cardiac tissue degeneration. The role of mitochondrial dysfunction in the onset of CVDs appears increasingly evident, as reflected by the impairment of proteins involved in lipid peroxidation, mitochondrial dynamics, respiratory chain complexes, and membrane polarization maintenance in CVD patients. Conversely, little is known about proteins responsible for the cross-talk between mitochondria and cytoplasm in cardiomyocytes. Mitochondrial transporters of the SLC25A family, in particular, are responsible for the translocation of nucleotides (e.g., ATP), amino acids (e.g., aspartate, glutamate, ornithine), organic acids (e.g. malate and 2-oxoglutarate), and other cofactors (e.g., inorganic phosphate, NAD+, FAD, carnitine, CoA derivatives) between the mitochondrial and cytosolic compartments. Thus, mitochondrial transporters play a key role in the mitochondria-cytosol cross-talk by leading metabolic pathways such as the malate/aspartate shuttle, the carnitine shuttle, the ATP export from mitochondria, and the regulation of permeability transition pore opening. Since all these pathways are crucial for maintaining healthy cardiomyocytes, mitochondrial carriers emerge as an interesting class of new possible pharmacological targets for CVD treatments.
Collapse
|
31
|
Cai Z, Wu C, Xu Y, Cai J, Zhao M, Zu L. The NO-cGMP-PKG Axis in HFpEF: From Pathological Mechanisms to Potential Therapies. Aging Dis 2023; 14:46-62. [PMID: 36818566 PMCID: PMC9937694 DOI: 10.14336/ad.2022.0523] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for almost half of all heart failure (HF) cases worldwide. Unfortunately, its incidence is expected to continue to rise, and effective therapy to improve clinical outcomes is lacking. Numerous efforts currently directed towards the pathophysiology of human HFpEF are uncovering signal transduction pathways and novel therapeutic targets. The nitric oxide-cyclic guanosine phosphate-protein kinase G (NO-cGMP-PKG) axis has been described as an important regulator of cardiac function. Suppression of the NO-cGMP-PKG signalling pathway is involved in the progression of HFpEF. Therefore, the NO-cGMP-PKG signalling pathway is a potential therapeutic target for HFpEF. In this review, we aim to explore the mechanism of NO-cGMP-PKG in the progression of HFpEF and to summarize potential therapeutic drugs that target this signalling pathway.
Collapse
Affiliation(s)
- Zhulan Cai
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Cencen Wu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Yuan Xu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Jiageng Cai
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Menglin Zhao
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Lingyun Zu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, China.
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
32
|
Buber J, Robertson HT. Cardiopulmonary exercise testing for heart failure: pathophysiology and predictive markers. Heart 2023; 109:256-263. [PMID: 35410893 DOI: 10.1136/heartjnl-2021-319617] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the numerous recent advancements in therapy, heart failure (HF) remains a principle cause of both morbidity and mortality. HF with preserved ejection fraction (HFpEF), a condition that shares the prevalence and adverse outcomes of HF with reduced ejection fraction, remains poorly recognised in its initial manifestations. Cardiopulmonary exercise testing (CPET), defined as a progressive work exercise test that includes non-invasive continuous measurement of cardiovascular and respiratory parameters, provides a reliable mode to evaluate for early features and for the assessment of prognostic features of both forms of HF. While CPET measurements are standard of care for advanced HF and transplant programmes, they merit a broader clinical application in the early diagnosis and assessment of patients with HFpEF. In this review, we provide an overview of the pathophysiology of exercise intolerance in HF and discuss key findings in CPETs used to evaluate both severity of impairment and the prognostic implications.
Collapse
Affiliation(s)
- Jonathan Buber
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - H Thomas Robertson
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
33
|
Turpin VRG, Lovoy GM, Parr SK, Hammond ST, Post HK, Caldwell JT, Banister HR, Scheuermann BC, Colburn TD, Ade CJ. Inorganic nitrate supplementation may improve diastolic function and the O 2 cost of exercise in cancer survivors: a pilot study. Support Care Cancer 2022; 31:63. [PMID: 36534177 DOI: 10.1007/s00520-022-07520-6] [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: 10/22/2021] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
In non-cancer populations, inorganic dietary nitrate (NO3-) supplementation is associated with enhanced cardiorespiratory function but remains untested in patients with a history of cancer. Therefore, this pilot study sought to determine if oral NO3- supplementation, as a supportive care strategy, increases left ventricular (LV) function and exercise performance in survivors of cancer treated with anticancer therapy while simultaneously evaluating the feasibility of the methods and procedures required for future large-scale randomized trials. Two cohorts of patients with a history of cancer treated with anticancer chemotherapy were recruited. Patients in cohort 1 (n = 7) completed a randomized, double-blind, crossover study with 7 days of NO3- or placebo (PL) supplementation, with echocardiography. Similarly, patients in cohort 2 (n = 6) received a single, acute dose of NO3- supplementation or PL. Pulmonary oxygen uptake (VO2), arterial blood pressure, and stroke volume were assessed during exercise. In cohort 1, NO3- improved LV strain rate in early filling (mean difference (MD) [95% CI]: - 0.3 1/s [- 0.6 to 0.06]; P = 0.04) and early mitral septal wall annular velocity (MD [95% CI]: 0.1 m/s [- 0.01 to - 0.001]; P = 0.02) compared to placebo. In cohort 2, NO3- decreased the O2 cost of low-intensity steady-state exercise (MD [95% CI]: - 0.5 ml/kg/min [- 0.9 to - 0.09]; P = 0.01). Resting and steady-state arterial blood pressure and stroke volume were not different between conditions. No differences between conditions for peak VO2 (MD [95% CI]: - 0.7 ml/kg/min [- 3.0 to 1.6]; P = 0.23) were observed. The findings from this pilot study warrant further investigation in larger clinical trials targeting the use of long-term inorganic dietary NO3- supplementation as a possible integrative supportive care strategy in patients following anticancer therapy.
Collapse
Affiliation(s)
- Vanessa-Rose G Turpin
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Garrett M Lovoy
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Shannon K Parr
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Stephen T Hammond
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Hunter K Post
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Jacob T Caldwell
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Heather R Banister
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Britton C Scheuermann
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Trenton D Colburn
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA
| | - Carl J Ade
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, USA. .,Physician Assistant Studies, College of Health and Human Sciences, Kansas State University, Manhattan, USA. .,Johnson Cancer Research Center, Kansas State University, Manhattan, USA. .,Clinical Integrative Physiology Laboratory, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
34
|
Peters AE, DeVore AD. Pharmacologic Therapy for Heart Failure with Preserved Ejection Fraction. Cardiol Clin 2022; 40:473-489. [DOI: 10.1016/j.ccl.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Bienvenu LA, Bell JR, Weeks KL, Delbridge LMD, Young MJ. New Perspectives on Sex Steroid and Mineralocorticoid Receptor Signaling in Cardiac Ischemic Injury. Front Physiol 2022; 13:896425. [PMID: 35846011 PMCID: PMC9277457 DOI: 10.3389/fphys.2022.896425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
The global burden of ischemic heart disease is burgeoning for both men and women. Although advances have been made, the need for new sex-specific therapies targeting key differences in cardiovascular disease outcomes in men and women remains. Mineralocorticoid receptor directed treatments have been successfully used for blood pressure control and heart failure management and represent a potentially valuable therapeutic option for ischemic cardiac events. Clinical and experimental data indicate that mineralocorticoid excess or inappropriate mineralocorticoid receptor (MR) activation exacerbates ischemic damage, and many of the intracellular response pathways activated in ischemia and subsequent reperfusion are regulated by MR. In experimental contexts, where MR are abrogated genetically or mineralocorticoid signaling is suppressed pharmacologically, ischemic injury is alleviated, and reperfusion recovery is enhanced. In the chronic setting, mineralocorticoid signaling induces fibrosis, oxidative stress, and inflammation, which can predispose to ischemic events and exacerbate post-myocardial infarct pathologies. Whilst a range of cardiac cell types are involved in mineralocorticoid-mediated regulation of cardiac function, cardiomyocyte-specific MR signaling pathways are key. Selective inhibition of cardiomyocyte MR signaling improves electromechanical resilience during ischemia and enhances contractile recovery in reperfusion. Emerging evidence suggests that the MR also contribute to sex-specific aspects of ischemic vulnerability. Indeed, MR interactions with sex steroid receptors may differentially regulate myocardial nitric oxide bioavailability in males and females, potentially determining sex-specific post-ischemic outcomes. There is hence considerable impetus for exploration of MR directed, cell specific therapies for both women and men in order to improve ischemic heart disease outcomes.
Collapse
Affiliation(s)
- Laura A. Bienvenu
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne VIC, Melbourne, VIC, Australia
| | - James R. Bell
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
- *Correspondence: James R. Bell,
| | - Kate L. Weeks
- Baker Department of Cardiometabolic Health, University of Melbourne VIC, Melbourne, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Lea M. D. Delbridge
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Morag J. Young
- Baker Department of Cardiometabolic Health, University of Melbourne VIC, Melbourne, VIC, Australia
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
36
|
Bayes-Genis A, Cediel G, Domingo M, Codina P, Santiago E, Lupón J. Biomarkers in Heart Failure with Preserved Ejection Fraction. Card Fail Rev 2022; 8:e20. [PMID: 35815256 PMCID: PMC9253965 DOI: 10.15420/cfr.2021.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous disorder developing from multiple aetiologies with overlapping pathophysiological mechanisms. HFpEF diagnosis may be challenging, as neither cardiac imaging nor physical examination are sensitive in this situation. Here, we review biomarkers of HFpEF, of which the best supported are related to myocardial stretch and injury, including natriuretic peptides and cardiac troponins. An overview of biomarkers of inflammation, extracellular matrix derangements and fibrosis, senescence, vascular dysfunction, anaemia/iron deficiency and obesity is also provided. Finally, novel biomarkers from -omics technologies, including plasma metabolites and circulating microRNAs, are outlined briefly. A cardiac-centred approach to HFpEF diagnosis using natriuretic peptides seems reasonable at present in clinical practice. A holistic approach including biomarkers that provide information on the non-cardiac components of the HFpEF syndrome may enrich our understanding of the disease and may be useful in classifying HFpEF phenotypes or endotypes that may guide patient selection in HFpEF trials.
Collapse
Affiliation(s)
- Antoni Bayes-Genis
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; Department of Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Germán Cediel
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Domingo
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Pau Codina
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Evelyn Santiago
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Lupón
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
37
|
Shannon OM, Clifford T, Seals DR, Craighead DH, Rossman MJ. Nitric oxide, aging and aerobic exercise: Sedentary individuals to Master's athletes. Nitric Oxide 2022; 125-126:31-39. [PMID: 35705144 DOI: 10.1016/j.niox.2022.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022]
Abstract
Aging is associated with a decline in physiological function and exercise performance. These effects are mediated, at least in part, by an age-related decrease in the bioavailability of nitric oxide (NO), a ubiquitous gasotransmitter and regulator of myriad physiological processes. The decrease in NO bioavailability with aging is especially apparent in sedentary individuals, whereas older, physically active individuals maintain higher levels of NO with advancing age. Strategies which enhance NO bioavailability (including nutritional supplementation) have been proposed as a potential means of reducing the age-related decrease in physiological function and enhancing exercise performance and may be of interest to a range of older individuals including those taking part in competitive sport. In this brief review we discuss the effects of aging on physiological function and endurance exercise performance, and the potential role of changes in NO bioavailability in these processes. We also provide a summary of current evidence for dietary supplementation with substrates for NO production - including inorganic nitrate and nitrite, l-arginine and l-citrulline - for improving exercise capacity/performance in older adults. Additionally, we discuss the (limited) evidence on the effects of (poly)phenols and other dietary antioxidants on NO bioavailability in older individuals. Finally, we provide suggestions for future research.
Collapse
Affiliation(s)
- Oliver M Shannon
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Tom Clifford
- School of Sport, Exercise and Health Science, Loughborough University, Loughborough, UK
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Daniel H Craighead
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
38
|
Proctor DN, Neely KA, Mookerjee S, Tucker J, Somani YB, Flanagan M, Kim-Shapiro DB, Basu S, Muller MD, Jin-Kwang Kim D. Inorganic nitrate supplementation and blood flow restricted exercise tolerance in post-menopausal women. Nitric Oxide 2022; 122-123:26-34. [PMID: 35240317 PMCID: PMC9062890 DOI: 10.1016/j.niox.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/12/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022]
Abstract
Exercise tolerance appears to benefit most from dietary nitrate (NO3-) supplementation when muscle oxygen (O2) availability is low. Using a double-blind, randomized cross-over design, we tested the hypothesis that acute NO3- supplementation would improve blood flow restricted exercise duration in post-menopausal women, a population with reduced endogenous nitric oxide bioavailability. Thirteen women (57-76 yr) performed rhythmic isometric handgrip contractions (10% MVC, 30 per min) during progressive forearm blood flow restriction (upper arm cuff gradually inflated 20 mmHg each min) on three study visits, with 7-10 days between visits. Approximately one week following the first (familiarization) visit, participants consumed 140 ml of NO3- concentrated (9.7 mmol, 0.6 gm NO3-) or NO3-depleted beetroot juice (placebo) on separate days (≥7 days apart), with handgrip exercise beginning 100 min post-consumption. Handgrip force recordings were analyzed to determine if NO3- supplementation enhanced force development as blood flow restriction progressed. Nitrate supplementation increased plasma NO3- (16.2-fold) and NO2- (4.2-fold) and time to volitional fatigue (61.8 ± 56.5 s longer duration vs. placebo visit; p = 0.03). Nitrate supplementation increased the rate of force development as forearm muscle ischemia progressed (p = 0.023 between 50 and 75% of time to fatigue) with non-significant effects thereafter (p = 0.052). No effects of nitrate supplementation were observed for mean duration of contraction or relaxation rates (all p > 0.150). These results suggest that acute NO3- supplementation prolongs time-to-fatigue and speeds grip force development during progressive forearm muscle ischemia in postmenopausal women.
Collapse
Affiliation(s)
- David N Proctor
- Penn State University, University Park, PA, USA; Penn State College of Medicine, Hershey, PA, USA.
| | | | | | | | | | - Michael Flanagan
- Penn State College of Medicine, Hershey, PA, USA; Penn State Health Family and Community Medicine, University Park, PA, USA
| | | | - Swati Basu
- Wake Forest University, Winston-Salem, NC, USA
| | - Matthew D Muller
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Danielle Jin-Kwang Kim
- Penn State University, University Park, PA, USA; Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
39
|
Weerts J, Mourmans SGJ, Barandiarán Aizpurua A, Schroen BLM, Knackstedt C, Eringa E, Houben AJHM, van Empel VPM. The Role of Systemic Microvascular Dysfunction in Heart Failure with Preserved Ejection Fraction. Biomolecules 2022; 12:biom12020278. [PMID: 35204779 PMCID: PMC8961612 DOI: 10.3390/biom12020278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a condition with increasing incidence, leading to a health care problem of epidemic proportions for which no curative treatments exist. Consequently, an urge exists to better understand the pathophysiology of HFpEF. Accumulating evidence suggests a key pathophysiological role for coronary microvascular dysfunction (MVD), with an underlying mechanism of low-grade pro-inflammatory state caused by systemic comorbidities. The systemic entity of comorbidities and inflammation in HFpEF imply that patients develop HFpEF due to systemic mechanisms causing coronary MVD, or systemic MVD. The absence or presence of peripheral MVD in HFpEF would reflect HFpEF being predominantly a cardiac or a systemic disease. Here, we will review the current state of the art of cardiac and systemic microvascular dysfunction in HFpEF (Graphical Abstract), resulting in future perspectives on new diagnostic modalities and therapeutic strategies.
Collapse
Affiliation(s)
- Jerremy Weerts
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
- Correspondence: ; Tel.: +31-43-387-7097
| | - Sanne G. J. Mourmans
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Arantxa Barandiarán Aizpurua
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Blanche L. M. Schroen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Christian Knackstedt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Etto Eringa
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6211 LK Maastricht, The Netherlands;
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Alfons J. H. M. Houben
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Vanessa P. M. van Empel
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| |
Collapse
|
40
|
Poole DC, Ferguson SK, Musch TI, Porcelli S. Role of nitric oxide in convective and diffusive skeletal microvascular oxygen kinetics. Nitric Oxide 2022; 121:34-44. [PMID: 35123062 DOI: 10.1016/j.niox.2022.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/29/2021] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
Progress in understanding physiological mechanisms often consists of discrete discoveries made across different models and species. Accordingly, understanding the mechanistic bases for how altering nitric oxide (NO) bioavailability impacts exercise tolerance (or not) depends on integrating information from cellular energetics and contractile regulation through microvascular/vascular control of O2 transport and pulmonary gas exchange. This review adopts state-of-the-art concepts including the intramyocyte power grid, the Wagner conflation of perfusive and diffusive O2 conductances, and the Critical Power/Critical Speed model of exercise tolerance to address how altered NO bioavailability may, or may not, affect physical performance. This question is germane from the elite athlete to the recreational exerciser and particularly the burgeoning heart failure (and other clinical) populations for whom elevating O2 transport and/or exercise capacity translates directly to improved life quality and reduced morbidity and mortality. The dearth of studies in females is also highlighted, and areas of uncertainty and questions for future research are identified.
Collapse
Affiliation(s)
- David C Poole
- Departments of Kinesiology and Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Scott K Ferguson
- Department of Kinesiology and Exercise Science, University of Hawaii, Hilo, HI, 96720, USA
| | - Timothy I Musch
- Departments of Kinesiology and Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Simone Porcelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
41
|
Boulmpou A, Theodorakopoulou MP, Alexandrou ME, Boutou AK, Papadopoulos CE, Pella E, Sarafidis P, Vassilikos V. Meta-analysis addressing the impact of cardiovascular-acting medication on peak oxygen uptake of patients with HFpEF. Heart Fail Rev 2022; 27:609-623. [DOI: 10.1007/s10741-021-10207-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 11/28/2022]
|
42
|
Dudhe R, Dudhe AC, Raut SD. Significance of Inorganic Nitrate Supplement in Cardiovascular Health. Cardiovasc Hematol Agents Med Chem 2022; 20:83-89. [PMID: 33906593 DOI: 10.2174/1871525719666210427130511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/27/2021] [Accepted: 03/29/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND & OBJECTIVES Nitric Oxide (NO) is frequently produced by the enzyme nitric oxide synthase (NOS) and is crucial for the control and effectiveness of the cardiovascular system. However, there is a substantial reduction in NOS activity with aging that can lead to the development of hypertension and other cardiovascular obstacles. Fortunately, NO can also be produced by sequential reduction of inorganic nitrates supplementation. This proves that NO from inorganic nitrate supplements can compensate for inadequate NOS activity, thus providing cardio protective benefits. DISCUSSION This review focuses on the general information about nitrous oxide, its types and mechanism of action and the fact that overview of inadequate NOS activity for cardio protective benefits was often studied for cardiovascular treatments. CONCLUSION We concluded that the natural plant NO is essential for cardiovascular activity to target site with desired concentration. Moreover, the researchers focused on evidence that suggested that nitrate supplementation could help regulate blood pressure, limit progression of atherosclerosis, and improve myocardial contractility in both healthy individuals and those with cardiovascular diseases.
Collapse
Affiliation(s)
- Rupesh Dudhe
- School of Pharmacy, G H Raisoni University, Saikheda, Chhindwara-480337, M.P., India
| | - Anshu C Dudhe
- School of Pharmacy, G H Raisoni University, Saikheda, Chhindwara-480337, M.P., India
| | - Shravan D Raut
- School of Pharmacy, G H Raisoni University, Saikheda, Chhindwara-480337, M.P., India
| |
Collapse
|
43
|
Moreira LDSG, Fanton S, Cardozo L, Borges NA, Combet E, Shiels PG, Stenvinkel P, Mafra D. Pink pressure: beetroot (Beta vulgaris rubra) as a possible novel medical therapy for chronic kidney disease. Nutr Rev 2021; 80:1041-1061. [PMID: 34613396 DOI: 10.1093/nutrit/nuab074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) manifests with systemic inflammation, oxidative stress, and gut dysbiosis, resulting in metabolic disorders and elevated rates of cardiovascular disease-associated death. These all correlate with a high economic cost to healthcare systems. Growing evidence indicates that diet is an indispensable ally in the prevention and management of CKD and its complications. In this context, the root vegetable beetroot (Beta vulgaris rubra) deserves special attention because it is a source of several bioactive compounds, such as nitrate, betaine, and betalain, and has shown beneficial effects in CKD, including reduction of blood pressure, anti-inflammatory effects, and antioxidant actions by scavenging radical oxidative species, as observed in preclinical studies. Beetroot consumption as a possible therapeutic strategy to improve the clinical treatment of patients with CKD and future directions for clinical studies are addressed in this narrative review.
Collapse
Affiliation(s)
- Laís de Souza Gouveia Moreira
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Susane Fanton
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ludmila Cardozo
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Natalia A Borges
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Emilie Combet
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Paul G Shiels
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Stenvinkel
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Denise Mafra
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
44
|
Pandey A, Shah SJ, Butler J, Kellogg DL, Lewis GD, Forman DE, Mentz RJ, Borlaug BA, Simon MA, Chirinos JA, Fielding RA, Volpi E, Molina AJA, Haykowsky MJ, Sam F, Goodpaster BH, Bertoni AG, Justice JN, White JP, Ding J, Hummel SL, LeBrasseur NK, Taffet GE, Pipinos II, Kitzman D. Exercise Intolerance in Older Adults With Heart Failure With Preserved Ejection Fraction: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 78:1166-1187. [PMID: 34503685 PMCID: PMC8525886 DOI: 10.1016/j.jacc.2021.07.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/16/2022]
Abstract
Exercise intolerance (EI) is the primary manifestation of chronic heart failure with preserved ejection fraction (HFpEF), the most common form of heart failure among older individuals. The recent recognition that HFpEF is likely a systemic, multiorgan disorder that shares characteristics with other common, difficult-to-treat, aging-related disorders suggests that novel insights may be gained from combining knowledge and concepts from aging and cardiovascular disease disciplines. This state-of-the-art review is based on the outcomes of a National Institute of Aging-sponsored working group meeting on aging and EI in HFpEF. We discuss aging-related and extracardiac contributors to EI in HFpEF and provide the rationale for a transdisciplinary, "gero-centric" approach to advance our understanding of EI in HFpEF and identify promising new therapeutic targets. We also provide a framework for prioritizing future research, including developing a uniform, comprehensive approach to phenotypic characterization of HFpEF, elucidating key geroscience targets for treatment, and conducting proof-of-concept trials to modify these targets.
Collapse
Affiliation(s)
- Ambarish Pandey
- University of Texas Southwestern Medical Center, Dallas, Texas, USA. https://twitter.com/ambarish4786
| | - Sanjiv J Shah
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Dean L Kellogg
- University of Texas Health Science Center and GRECC, South Texas Veterans Affairs Health System, San Antonio, Texas, USA
| | | | - Daniel E Forman
- University of Pittsburgh and VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Robert J Mentz
- Duke Clinical Research Center, Durham, North Carolina, USA
| | | | - Marc A Simon
- University of Pittsburgh and VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | | | | | - Elena Volpi
- University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | | | | | - Flora Sam
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Bret H Goodpaster
- Advent Health Translational Research Institute, Orlando, Florida, USA
| | - Alain G Bertoni
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jamie N Justice
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Jingzhone Ding
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Scott L Hummel
- University of Michigan and the VA Ann Arbor Health System, Ann Arbor, Michigan, USA
| | | | | | | | - Dalane Kitzman
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
45
|
Alsulayyim AS, Alasmari AM, Alghamdi SM, Polkey MI, Hopkinson NS. Impact of dietary nitrate supplementation on exercise capacity and cardiovascular parameters in chronic respiratory disease: a systematic review and meta-analysis. BMJ Open Respir Res 2021; 8:8/1/e000948. [PMID: 34489239 PMCID: PMC8422488 DOI: 10.1136/bmjresp-2021-000948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022] Open
Abstract
Background Dietary nitrate supplementation, usually in the form of beetroot juice, may improve exercise performance and endothelial function. We undertook a systematic review and meta-analysis to establish whether this approach has beneficial effects in people with respiratory disease. Methods A systematic search of records up to March 2021 was performed on PubMed, CINAHL, MEDLINE (Ovid), Cochrane and Embase to retrieve clinical trials that evaluated the efficacy of dietary nitrate supplementation on cardiovascular parameters and exercise capacity in chronic respiratory conditions. Two authors independently screened titles, abstracts and full texts of potential studies and performed the data extraction. Results After full-text review of 67 papers, eleven (two randomised controlled trials and nine crossover trials) involving 282 participants met the inclusion criteria. Three were single dose; seven short term; and one, the largest (n=122), done in the context of pulmonary rehabilitation. Pooled analysis showed that dietary nitrate supplementation reduced systolic blood pressure (BP), diastolic BP and mean arterial pressure (mean difference (95% CI), −3.39 mm Hg (−6.79 to 0.01); p=0.05 and –2.20 mm Hg (−4.36 to −0.03); p=0.05 and −4.40 mm Hg (−7.49 to −1.30); p=0.005, respectively). It was associated with increased walk distance in the context of pulmonary rehabilitation (standardised mean difference (95% CI), 0.47 (0.11 to 0.83), p=0.01), but no effect was identified in short-term studies (0.08 (−0.32 to 0.49). Conclusion Dietary nitrate supplementation may have a beneficial effect on BP and augment the effect of pulmonary rehabilitation on exercise capacity. Short-term studies do not suggest a consistent benefit on exercise capacity. PROSPERO registration number CRD42019130123.
Collapse
Affiliation(s)
- Abdullah S Alsulayyim
- National Heart and Lung Institute, Imperial College London, London, UK.,Faculty of Applied Medical Sciences, Respiratory Therapy Department, Jazan University, Jazan, Saudi Arabia
| | - Ali M Alasmari
- National Heart and Lung Institute, Imperial College London, London, UK.,Faculty of Medical Rehabilitation Sciences, Taibah University, Madinah, Saudi Arabia
| | - Saeed M Alghamdi
- National Heart and Lung Institute, Imperial College London, London, UK.,Clinical Technology Department, Umm Al-Qura University, College of Applied Medical Science, Makkah, Saudi Arabia
| | - Michael I Polkey
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
46
|
Leite S, Moreira-Costa L, Cerqueira R, Sousa-Mendes C, Angélico-Gonçalves A, Fontoura D, Vasques-Nóvoa F, Leite-Moreira AF, Lourenço AP. Chronic Sildenafil Therapy in the ZSF1 Obese Rat Model of Metabolic Syndrome and Heart Failure With Preserved Ejection Fraction. J Cardiovasc Pharmacol Ther 2021; 26:690-701. [PMID: 34328815 DOI: 10.1177/10742484211034253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although decreased protein kinase G (PKG) activity was proposed as potential therapeutic target in heart failure with preserved ejection fraction (HFpEF), randomized clinical trials (RCTs) with type-5 phosphodiesterase inhibitors (PDE5i) showed neutral results. Whether specific subgroups of HFpEF patients may benefit from PDE5i remains to be defined. Our aim was to test chronic sildenafil therapy in the young male ZSF1 obese rat model of HFpEF with severe hypertension and metabolic syndrome. Sixteen-week-old ZSF1 obese rats were randomly assigned to receive sildenafil 100 mg·Kg-1·d-1 dissolved in drinking water (ZSF1 Ob SIL, n = 8), or placebo (ZSF1 Ob PL, n = 8). A group of Wistar-Kyoto rats served as control (WKY, n = 8). Four weeks later animals underwent effort tests, glucose metabolism studies, hemodynamic evaluation, and samples were collected for aortic ring preparation, left ventricular (LV) myocardial adenosine triphosphate (ATP) quantification, immunoblotting and histology. ZSF1 Ob PL rats showed systemic hypertension, aortic stiffening, impaired LV relaxation and increased LV stiffness, with preserved ejection fraction and cardiac index. Their endurance capacity was decreased as assessed by maximum workload and peak oxygen consumption (V˙O2) and respiratory quotient were increased, denoting more reliance on anaerobic metabolism. Additionally, ATP levels were decreased. Chronic sildenafil treatment attenuated hypertension and decreased LV stiffness, modestly enhancing effort tolerance with a concomitant increase in peak, ATP levels and VASP phosphorylation. Chronic sildenafil therapy in this model of HFpEF of the young male with extensive and poorly controlled comorbidities has beneficial cardiovascular effects which support RCTs in HFpEF patient subgroups with similar features.
Collapse
Affiliation(s)
- Sara Leite
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal.,Anta Family Health Unit, Espinho/Gaia Healthcare Centre, Espinho, Portugal
| | - Liliana Moreira-Costa
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal
| | - Rui Cerqueira
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal.,Department of Cardiothoracic Surgery, São João Hospital Centre, Porto, Portugal
| | - Cláudia Sousa-Mendes
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal
| | | | - Dulce Fontoura
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal
| | - Francisco Vasques-Nóvoa
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal.,Department of Internal Medicine, São João Hospital Centre, Porto, Portugal
| | - Adelino F Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal.,Department of Cardiothoracic Surgery, São João Hospital Centre, Porto, Portugal
| | - André P Lourenço
- Department of Surgery and Physiology, Faculty of Medicine, 26705University of Porto, Porto, Portugal.,Department of Anesthesiology, São João Hospital Centre, Porto, Portugal
| |
Collapse
|
47
|
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
|
48
|
Acute Effects of Dietary Nitrate on Exercise Tolerance, Muscle Oxygenation, and Cardiovascular Function in Patients With Peripheral Arterial Disease. Int J Sport Nutr Exerc Metab 2021; 31:385-396. [PMID: 34284348 DOI: 10.1123/ijsnem.2021-0054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 11/18/2022]
Abstract
Previous studies have used supplements to increase dietary nitrate intake in clinical populations. Little is known about whether effects can also be induced through vegetable consumption. Therefore, the aim of this study was to assess the impact of dietary nitrate, through nitrate-rich vegetables (NRV) and beetroot juice (BRJ) supplementation, on plasma nitrate and nitrite concentrations, exercise tolerance, muscle oxygenation, and cardiovascular function in patients with peripheral arterial disease. In a randomized crossover design, 18 patients with peripheral arterial disease (age: 73 ± 8 years) followed a nitrate intake protocol (∼6.5 mmol) through the consumption of NRV, BRJ, and nitrate-depleted BRJ (placebo). Blood samples were taken, blood pressure and arterial stiffness were measured in fasted state and 150 min after intervention. Each intervention was followed by a maximal walking exercise test to determine claudication onset time and peak walking time. Gastrocnemius oxygenation was measured by near-infrared spectroscopy. Blood samples were taken and blood pressure was measured 10 min after exercise. Mean plasma nitrate and nitrite concentrations increased (nitrate; Time × Intervention interaction; p < .001), with the highest concentrations after BRJ (494 ± 110 μmol/L) compared with NRV (202 ± 89 μmol/L) and placebo (80 ± 19 μmol/L; p < .001). Mean claudication onset time and peak walking time did not differ between NRV (413 ± 187 s and 745 ± 220 s, respectively), BRJ (392 ± 154 s and 746 ± 176 s), and placebo (403 ± 176 s and 696 ± 222 s) (p = .762 and p = .165, respectively). Gastrocnemius oxygenation, blood pressure, and arterial stiffness were not affected by the intervention. NRV and BRJ intake markedly increase plasma nitrate and nitrite, but this does not translate to improved exercise tolerance, muscle oxygenation, and/or cardiovascular function.
Collapse
|
49
|
Bahrami LS, Arabi SM, Feizy Z, Rezvani R. The effect of beetroot inorganic nitrate supplementation on cardiovascular risk factors: A systematic review and meta-regression of randomized controlled trials. Nitric Oxide 2021; 115:8-22. [PMID: 34119659 DOI: 10.1016/j.niox.2021.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Inorganic nitrate is one of the most effective compounds in beetroot for improving cardiovascular function due to its conversion to nitric oxide in the body. This review and meta-analysis aimed to investigate the role of beetroot inorganic nitrate supplementation on adults' cardiovascular risk factors. METHODS We conducted a systematic literature review of articles published without time limitation until November 2020 in PubMed, Embase, ISI Web of Science, Scopus, Cochrane Library, and gray literature databases. We included the original randomized clinical trials (RCTs) in which the effect of beetroot inorganic nitrate supplementation on endothelial function, arterial stiffness, and blood pressure was studied. RESULTS 43 studies were included for qualitative synthesis, out of which 27 were eligible for meta-analysis. Beetroot inorganic nitrate supplementation significantly decreased Arterial Stiffness (Pulse Wave Velocity (-0.27 m/s, p = 0.04)) and increased Endothelial function (Flow Mediated Dilation: 0.62%, p = 0.002) but did not change other parameters (p > 0.05). CONCLUSION Beetroot inorganic nitrate supplementation might have a beneficial effect on cardiovascular risk factors. Further high-quality investigations will be needed to provide sufficient evidence.
Collapse
Affiliation(s)
- Leila Sadat Bahrami
- Metabolic Syndrome Research Center, Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyyed Mostafa Arabi
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Zahra Feizy
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX79414, USA.
| | - Reza Rezvani
- Metabolic Syndrome Research Center, Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
50
|
Shannon OM, Easton C, Shepherd AI, Siervo M, Bailey SJ, Clifford T. Dietary nitrate and population health: a narrative review of the translational potential of existing laboratory studies. BMC Sports Sci Med Rehabil 2021; 13:65. [PMID: 34099037 PMCID: PMC8186051 DOI: 10.1186/s13102-021-00292-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dietary inorganic nitrate (NO3-) is a polyatomic ion, which is present in large quantities in green leafy vegetables and beetroot, and has attracted considerable attention in recent years as a potential health-promoting dietary compound. Numerous small, well-controlled laboratory studies have reported beneficial health effects of inorganic NO3- consumption on blood pressure, endothelial function, cerebrovascular blood flow, cognitive function, and exercise performance. Translating the findings from small laboratory studies into 'real-world' applications requires careful consideration. MAIN BODY This article provides a brief overview of the existing empirical evidence basis for the purported health-promoting effects of dietary NO3- consumption. Key areas for future research are then proposed to evaluate whether promising findings observed in small animal and human laboratory studies can effectively translate into clinically relevant improvements in population health. These proposals include: 1) conducting large-scale, longer duration trials with hard clinical endpoints (e.g. cardiovascular disease incidence); 2) exploring the feasibility and acceptability of different strategies to facilitate a prolonged increase in dietary NO3- intake; 3) exploitation of existing cohort studies to explore associations between NO3- intake and health outcomes, a research approach allowing larger samples sizes and longer duration follow up than is feasible in randomised controlled trials; 4) identifying factors which might account for individual differences in the response to inorganic NO3- (e.g. sex, genetics, habitual diet) and could assist with targeted/personalised nutritional interventions; 5) exploring the influence of oral health and medication on the therapeutic potential of NO3- supplementation; and 6) examining potential risk of adverse events with long term high- NO3- diets. CONCLUSION The salutary effects of dietary NO3- are well established in small, well-controlled laboratory studies. Much less is known about the feasibility and efficacy of long-term dietary NO3- enrichment for promoting health, and the factors which might explain the variable responsiveness to dietary NO3- supplementation between individuals. Future research focussing on the translation of laboratory data will provide valuable insight into the potential applications of dietary NO3- supplementation to improve population health.
Collapse
Affiliation(s)
- Oliver M Shannon
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Chris Easton
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Blantyre, Scotland, UK
| | - Anthony I Shepherd
- School of Sport, Health & Exercise Science, University of Portsmouth, Portsmouth, UK
| | - Mario Siervo
- School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Tom Clifford
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| |
Collapse
|