1
|
Du XJ, She G, Wu W, Deng XL. Coupling of β-adrenergic and Hippo pathway signaling: Implications for heart failure pathophysiology and metabolic therapy. Mitochondrion 2024; 78:101941. [PMID: 39122227 DOI: 10.1016/j.mito.2024.101941] [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: 03/20/2024] [Revised: 07/17/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Activation of the sympatho-β-adrenergic receptor (βAR) system is the hallmark of heart disease with adverse consequences that facilitate the onset and progression of heart failure (HF). Use of β-blocking drugs has become the front-line therapy for HF. Last decade has witnessed progress in research demonstrating a pivotal role of Hippo pathway in cardiomyopathy and HF. Clinical studies have revealed myocardial Hippo pathway activation/YAP-TEAD1 inactivation in several types of human cardiomyopathy. Experimental activation of cardiac Hippo signaling or inhibition of YAP-TEAD1 have been shown to leads dilated cardiomyopathy with severe mitochondrial dysfunction and metabolic reprogramming. Studies have also convincingly shown that stimulation of βAR activates cardiac Hippo pathway with inactivation of the down-stream effector molecules YAP/TAZ. There is strong evidence for the adverse consequences of the βAR-Hippo signaling leading to HF. In addition to promoting cardiomyocyte death and fibrosis, recent progress is the demonstration of mitochondrial dysfunction and metabolic reprogramming mediated by βAR-Hippo pathway signaling. Activation of cardiac βAR-Hippo signaling is potent in downregulating a range of mitochondrial and metabolic genes, whereas expression of pro-inflammatory and pro-fibrotic factors are upregulated. Coupling of βAR-Hippo pathway signaling is mediated by several kinases, mechanotransduction and/or Ca2+ signaling, and can be blocked by β-antagonists. Demonstration of the converge of βAR signaling and Hippo pathway bears implications for a better understanding on the role of enhanced sympathetic nervous activity, efficacy of β-antagonists, and metabolic therapy targeting this pathway in HF. In this review we summarize the progress and discuss future research directions in this field.
Collapse
Affiliation(s)
- Xiao-Jun Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China; Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia,.
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Wei Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China; Department of Cardiology, Shaanxi Provincial Hospital and the Third Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
2
|
Rosengren S, Skibsted Clemmensen T, Hvitfeldt Poulsen S, Tolbod L, Harms HJ, Wikström G, Kero T, Thyrsted Ladefoged B, Sörensen J. Outcome prediction by myocardial external efficiency from 11 C-acetate positron emission tomography in cardiac amyloidosis. ESC Heart Fail 2024; 11:44-53. [PMID: 37806676 PMCID: PMC10804164 DOI: 10.1002/ehf2.14545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/02/2023] [Accepted: 09/15/2023] [Indexed: 10/10/2023] Open
Abstract
AIMS This study aimed to study the prognostic value of myocardial oxygen consumption (MVO2 ) and myocardial external efficiency (MEE) from 11 C-acetate positron emission tomography (PET) in cardiac amyloidosis (CA) patients. METHODS AND RESULTS Forty-eight CA patients, both transthyretin (ATTR) and immunoglobulin light chain (AL) amyloidosis, and 20 controls were included. All subjects were examined with 11 C-acetate PET and echocardiography. MVO2 , forward stroke volume (FSV), and left ventricular mass (LVM) were derived from 11 C-acetate PET and used to calculate MEE. CA patients were followed for survival and the prognostic impact of clinical, echocardiographic, and 11 C-acetate PET parameters was analysed. MVO2 and MEE were reduced in CA compared with controls, but without significant difference between deceased and surviving CA patients. The ratio of 11 C-acetate PET-derived FSV and LVM was also reduced in CA and significantly lowered in deceased patients compared with survivors. In univariate analysis, New York Heart Association class, N-terminal pro-brain natriuretic peptide, and the 11 C-acetate PET parameters FSV/LVM and MEE were the strongest prognostic factors. Of the 11 C-acetate PET parameters, FSV/LVM was the strongest survival predictor with hazard ratio of 0.56 per 0.1 mL/g (95% confidence interval 0.39-0.81, P = 0.002) and independently prognostic in a multivariate model. MEE significantly separated deceased from surviving CA patients with the cut-off of 15.7% (P = 0.032). Survival was significantly shorter with FSV/LVM below 0.27 mL/g (P < 0.001), also when separating AL- and ATTR-CA. CONCLUSIONS Reduced MEE was associated with shorter survival in CA patients, but FSV/LVM was the strongest survival predictor and the only independently prognostic 11 C-acetate PET parameter in multivariate analysis.
Collapse
Affiliation(s)
- Sara Rosengren
- Department of Medical Sciences, HaematologyUppsala UniversityIng 100, pl 2, Akademiska Hospital751 85UppsalaSweden
| | | | | | - Lars Tolbod
- Department of Nuclear Medicine and PETAarhus University HospitalAarhusDenmark
| | - Hendrik J. Harms
- Department of Nuclear Medicine and PETAarhus University HospitalAarhusDenmark
| | | | - Tanja Kero
- Department of Surgical Sciences, Nuclear Medicine and PETUppsala UniversityUppsalaSweden
| | | | - Jens Sörensen
- Department of Nuclear Medicine and PETAarhus University HospitalAarhusDenmark
- Department of Surgical Sciences, Nuclear Medicine and PETUppsala UniversityUppsalaSweden
| |
Collapse
|
3
|
Rivas García S, Álvarez-García J. Sacubitril/valsartan: Where mechanism meets evidence-based medicine. Eur J Heart Fail 2024; 26:127-129. [PMID: 38037688 DOI: 10.1002/ejhf.3100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023] Open
Affiliation(s)
- Sonia Rivas García
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), Department of Cardiology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Jesús Álvarez-García
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), Department of Cardiology, Ramón y Cajal University Hospital, Madrid, Spain
| |
Collapse
|
4
|
Nesterov SV, Räty J, Nammas W, Maaniitty T, Galloo X, Stassen J, Laurila S, Vasankari T, Huusko J, Bax JJ, Saraste A, Knuuti J. Short-term effects of sacubitril/valsartan therapy on myocardial oxygen consumption and energetic efficiency of cardiac work in heart failure with reduced ejection fraction: A randomized controlled study. Eur J Heart Fail 2024; 26:117-126. [PMID: 37905338 DOI: 10.1002/ejhf.3072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023] Open
Abstract
AIMS We sought to evaluate the mechanism of angiotensin receptor-neprilysin inhibitor (ARNI) sacubitril/valsartan therapy and compare it with a valsartan-only control group in patients with heart failure with reduced ejection fraction (HFrEF). METHODS AND RESULTS The study was a phase IV, prospective, randomized, double-blind, parallel-group study in patients with New York Heart Association class II-III heart failure and left ventricular ejection fraction (LVEF) ≤35%. During a 6-week run-in period, all patients received valsartan therapy, which was up-titrated to the highest tolerated dose level (80 mg bid or 160 mg bid) and then randomized to either valsartan or sacubitril/valsartan. Myocardial oxygen consumption, energetic efficiency of cardiac work, cardiac and systemic haemodynamics were quantified using echocardiography and 11 C-acetate positron emission tomography before and after 6 weeks of therapy (on stable dose) in 55 patients (ARNI group: n = 27, mean age 63 ± 10 years, LVEF 29.2 ± 10.4%; and valsartan-only control group: n = 28, mean age 64 ± 8 years, LVEF 29.0 ± 7.3%; all p = NS). The energetic efficiency of cardiac work remained unchanged in both treatment arms. However, both diastolic (-4.5 mmHg; p = 0.026) and systolic blood pressure (-9.8 mmHg; p = 0.0007), myocardial perfusion (-0.054 ml/g/min; p = 0.045), and left ventricular mechanical work (-296; p = 0.038) decreased significantly in the ARNI group compared to the control group. Although myocardial oxygen consumption decreased in the ARNI group (-5.4%) compared with the run-in period and remained unchanged in the control group (+0.5%), the between-treatment group difference was not significant (p = 0.088). CONCLUSIONS We found no differences in the energetic efficiency of cardiac work between ARNI and valsartan-only groups in HFrEF patients. However, ARNI appears to have haemodynamic and cardiac mechanical effects over valsartan in heart failure patients.
Collapse
Affiliation(s)
- Sergey V Nesterov
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Johanna Räty
- Department of Clinical Physiology, Nuclear Medicine and PET, Turku University Hospital, Turku, Finland
| | - Wail Nammas
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Teemu Maaniitty
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Clinical Physiology, Nuclear Medicine and PET, Turku University Hospital, Turku, Finland
| | - Xavier Galloo
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Stassen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sanna Laurila
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Tuija Vasankari
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | | | - Jeroen J Bax
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Clinical Physiology, Nuclear Medicine and PET, Turku University Hospital, Turku, Finland
| |
Collapse
|
5
|
Correale M, Tricarico L, Croella F, Alfieri S, Fioretti F, Brunetti ND, Inciardi RM, Nodari S. Novelties in the pharmacological approaches for chronic heart failure: new drugs and cardiovascular targets. Front Cardiovasc Med 2023; 10:1157472. [PMID: 37332581 PMCID: PMC10272855 DOI: 10.3389/fcvm.2023.1157472] [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: 02/02/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Despite recent advances in chronic heart failure (HF) management, the prognosis of HF patients is poor. This highlights the need for researching new drugs targeting, beyond neurohumoral and hemodynamic modulation approach, such as cardiomyocyte metabolism, myocardial interstitium, intracellular regulation and NO-sGC pathway. In this review we report main novelties on new possible pharmacological targets for HF therapy, mainly on new drugs acting on cardiac metabolism, GCs-cGMP pathway, mitochondrial function and intracellular calcium dysregulation.
Collapse
Affiliation(s)
- Michele Correale
- Department of Cardiothoracic, Policlinico Riuniti University Hospital, Foggia, Italy
| | - Lucia Tricarico
- Department of Cardiothoracic, Policlinico Riuniti University Hospital, Foggia, Italy
| | - Francesca Croella
- Department of Medical & Surgical Sciences, University of Foggia, Foggia, Italy
| | - Simona Alfieri
- Department of Medical & Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesco Fioretti
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili Hospital and University of Brescia, Brescia, Italy
| | | | - Riccardo M. Inciardi
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili Hospital and University of Brescia, Brescia, Italy
| | - Savina Nodari
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili Hospital and University of Brescia, Brescia, Italy
| |
Collapse
|
6
|
Wang T, Xiong T, Yang Y, Zuo B, Chen X, Wang D. Metabolic remodeling in takotsubo syndrome. Front Cardiovasc Med 2022; 9:1060070. [PMID: 36505375 PMCID: PMC9729286 DOI: 10.3389/fcvm.2022.1060070] [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: 10/02/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
The heart requires a large and constant supply of energy that is mainly the result of an efficient metabolic machinery that converges on mitochondrial oxidative metabolism to maintain its continuous mechanical work. Perturbations in these metabolic processes may therefore affect energy generation and contractile function directly. Metabolism characteristics in takotsubo syndrome (TTS) reveals several metabolic alterations called metabolic remodeling, including the hyperactivity of sympathetic metabolism, derangements of substrate utilization, effector subcellular dysfunction and systemic metabolic disorders, ultimately contributing to the progression of the disease and the development of a persistent and long-term heart failure (HF) phenotype. In this review, we explore the current literature investigating the pathological metabolic alterations in TTS. Although the metabolic dysfunction in takotsubo hearts is initially recognized as a myocardial metabolic inflexibility, we suggest that the widespread alterations of systemic metabolism with complex interplay between the heart and peripheral tissues rather than just cardiometabolic disorders per se account for long-term maladaptive metabolic, functional and structural impairment under this condition. Therapeutic strategies with the recent evidence from small clinical and animal researches, especially for targeting substrate utilization and/or oxidative stress, might be promising tools to improve the outcome of patients with TTS beyond that achieved with traditional sympathetic inhibition and symptomatic therapies.
Collapse
Affiliation(s)
- Ti Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People’s Hospital), Taizhou, Jiangsu, China
| | - Ting Xiong
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuxue Yang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People’s Hospital), Taizhou, Jiangsu, China
| | - Bangyun Zuo
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People’s Hospital), Taizhou, Jiangsu, China
| | - Xiwei Chen
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People’s Hospital), Taizhou, Jiangsu, China
| | - Daxin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People’s Hospital), Taizhou, Jiangsu, China,*Correspondence: Daxin Wang, ,
| |
Collapse
|
7
|
Yurista SR, Chen S, Welsh A, Tang WHW, Nguyen CT. Targeting Myocardial Substrate Metabolism in the Failing Heart: Ready for Prime Time? Curr Heart Fail Rep 2022; 19:180-190. [PMID: 35567658 PMCID: PMC10950325 DOI: 10.1007/s11897-022-00554-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW We review the clinical benefits of altering myocardial substrate metabolism in heart failure. RECENT FINDINGS Modulation of cardiac substrates (fatty acid, glucose, or ketone metabolism) offers a wide range of therapeutic possibilities which may be applicable to heart failure. Augmenting ketone oxidation seems to offer great promise as a new therapeutic modality in heart failure. The heart has long been recognized as metabolic omnivore, meaning it can utilize a variety of energy substrates to maintain adequate ATP production. The adult heart uses fatty acid as a major fuel source, but it can also derive energy from other substrates including glucose and ketone, and to some extent pyruvate, lactate, and amino acids. However, cardiomyocytes of the failing heart endure remarkable metabolic remodeling including a shift in substrate utilization and reduced ATP production, which account for cardiac remodeling and dysfunction. Research to understand the implication of myocardial metabolic perturbation in heart failure has grown in recent years, and this has raised interest in targeting myocardial substrate metabolism for heart failure therapy. Due to the interdependency between different pathways, the main therapeutic metabolic approaches include inhibiting fatty acid uptake/fatty acid oxidation, reducing circulating fatty acid levels, increasing glucose oxidation, and augmenting ketone oxidation.
Collapse
Affiliation(s)
- Salva R Yurista
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Boston, MA, 02129, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Shi Chen
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Boston, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Aidan Welsh
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Boston, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - W H Wilson Tang
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
- Cardiovascular Innovation Research Center, Cleveland Clinic, Cleveland, OH, USA
| | - Christopher T Nguyen
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Boston, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
- Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Innovation Research Center, Cleveland Clinic, Cleveland, OH, USA
- Imaging Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
8
|
Lembo M, Trimarco V, Manzi MV, Mancusi C, Esposito G, Esposito S, Morisco C, Izzo R, Trimarco B. Determinants of improvement of left ventricular mechano-energetic efficiency in hypertensive patients. Front Cardiovasc Med 2022; 9:977657. [PMID: 35966525 PMCID: PMC9365966 DOI: 10.3389/fcvm.2022.977657] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Arterial hypertension, especially when coexisting with other cardiovascular risk factors, could determine an imbalance between myocardial energetic demand and altered efficiency, leading to an early left ventricular (LV) systolic dysfunction, even in terms of echo-derived mechano-energetic efficiency indexed for myocardial mass (MEEi). We aim to analyse an improvement in LV MEEi, if any, in a population of hypertensive patients with a long-term follow-up and to identify clinical, metabolic and therapeutic determinants of LV MEEi amelioration. Materials and methods In total, 7,052 hypertensive patients, followed-up for 5.3 ± 4.5 years, enrolled in the Campania Salute Network, underwent echocardiographic and clinical evaluation. LV MEEi was obtained as the ratio between stroke volume and heart rate and normalized per grams of LV mass and ΔMEEi was calculated as difference between follow-up and baseline MEEi. Patients in the highest ΔMEEi quartile (≥0.0454 mL/s/g) (group 1) were compared to the merged first, second and third quartiles (<0.0454 mL/s/g) (group 2). METS-IR (Metabolic Score for Insulin Resistance), an established index of insulin sensitivity, was also derived. Results Patients with MEEi improvement experienced a lower rate of major cardiovascular events (p = 0.02). After excluding patients experiencing cardiovascular events, patients in group 1 were younger (p < 0.0001), less often diabetic (p = 0.001) and obese (p = 0.035). Group 1 experienced more frequently LV mass index reduction, lower occurrence of LV ejection fraction reduction, and had a better metabolic control in terms of mean METS-IR during the follow-up (all p < 0.0001). Beta-blockers were more often used in group 1 (p < 0.0001) than group 2. A logistic regression analysis showed that younger age, lower mean METS-IR values, more frequent LV mass index reduction and therapy with beta-blockers were significantly associated with LV MEEi improvement, independently of presence of diabetes and obesity. Conclusion Metabolic control and therapy with beta-blockers could act in a synergic way, determining an improvement in LV MEEi in hypertensive patients over time, possibly confining cardiac damage and hampering progression toward heart failure.
Collapse
Affiliation(s)
- Maria Lembo
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Valentina Trimarco
- Department of Neurosciences, Federico II University of Naples, Naples, Italy
| | - Maria Virginia Manzi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Costantino Mancusi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Salvatore Esposito
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- *Correspondence: Raffaele Izzo,
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| |
Collapse
|
9
|
Harbo MB, Stokke MK, Sjaastad I, Espe EKS. One step closer to myocardial physiology: From PV loop analysis to state-of-the-art myocardial imaging. Acta Physiol (Oxf) 2022; 234:e13759. [PMID: 34978759 DOI: 10.1111/apha.13759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/14/2021] [Accepted: 01/01/2022] [Indexed: 11/29/2022]
Abstract
Recent advances in cardiac imaging have revitalized the assessment of fundamental physiological concepts. In the field of cardiac physiology, invasive measurements with pressure-volume (PV) loops have served as the gold standard methodology for the characterization of left ventricular (LV) function. From PV loop data, fundamental aspects of LV chamber function are derived such as work, efficiency, stiffness and contractility. However, the parametrization of these aspects is limited because of the need for invasive procedures. Through the utilization of recent advances in echocardiography, magnetic resonance imaging and positron emission tomography, it has become increasingly feasible to quantify these fundamental aspects of LV function non-invasively. Importantly, state-of-the-art imaging technology enables direct assessment of myocardial performance, thereby extending functional assessment from the net function of the LV chamber, as is done with PV loops, to the myocardium itself. With a strong coupling to underlying myocardial physiology, imaging measurements of myocardial work, efficiency, stiffness and contractility could represent the next generation of functional parameters. The purpose of this review is to discuss how the new imaging parameters of myocardial work, efficiency, stiffness and contractility can bring cardiac physiologists, researchers and clinicians alike one step closer to underlying myocardial physiology.
Collapse
Affiliation(s)
- Markus Borge Harbo
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
| | - Mathis Korseberg Stokke
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
- Department of Cardiology Oslo University Hospital Rikshospitalet Oslo Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
| | - Emil Knut Stenersen Espe
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
| |
Collapse
|
10
|
Stendahl JC, Sinusas AJ. 11C-acetate PET: A powerful tool to analyze metabolic and functional changes in the heart related to alcohol consumption. J Nucl Cardiol 2022; 29:289-292. [PMID: 32676907 PMCID: PMC7854759 DOI: 10.1007/s12350-020-02268-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 02/03/2023]
Affiliation(s)
- John C Stendahl
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Dana 3, P.O. Box 208017, New Haven, CT, 06520-8017, USA
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Dana 3, P.O. Box 208017, New Haven, CT, 06520-8017, USA.
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
11
|
Sorensen J. PET imaging of heart diseases by Acetate. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00209-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
12
|
Saraste A, Knuuti J. PET imaging in diabetic cardiomyopathy. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00051-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
13
|
Effects of Metformin in Heart Failure: From Pathophysiological Rationale to Clinical Evidence. Biomolecules 2021; 11:biom11121834. [PMID: 34944478 PMCID: PMC8698925 DOI: 10.3390/biom11121834] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a worldwide major health burden and heart failure (HF) is the most common cardiovascular (CV) complication in affected patients. Therefore, identifying the best pharmacological approach for glycemic control, which is also useful to prevent and ameliorate the prognosis of HF, represents a crucial issue. Currently, the choice is between the new drugs sodium/glucose co-transporter 2 inhibitors that have consistently shown in large CV outcome trials (CVOTs) to reduce the risk of HF-related outcomes in T2DM, and metformin, an old medicament that might end up relegated to the background while exerting interesting protective effects on multiple organs among which include heart failure. When compared with other antihyperglycemic medications, metformin has been demonstrated to be safe and to lower morbidity and mortality for HF, even if these results are difficult to interpret as they emerged mainly from observational studies. Meta-analyses of randomized controlled clinical trials have not produced positive results on the risk or clinical course of HF and sadly, large CV outcome trials are lacking. The point of force of metformin with respect to new diabetic drugs is the amount of data from experimental investigations that, for more than twenty years, still continues to provide mechanistic explanations of the several favorable actions in heart failure such as, the improvement of the myocardial energy metabolic status by modulation of glucose and lipid metabolism, the attenuation of oxidative stress and inflammation, and the inhibition of myocardial cell apoptosis, leading to reduced cardiac remodeling and preserved left ventricular function. In the hope that specific large-scale trials will be carried out to definitively establish the metformin benefit in terms of HF failure outcomes, we reviewed the literature in this field, summarizing the available evidence from experimental and clinical studies reporting on effects in heart metabolism, function, and structure, and the prominent pathophysiological mechanisms involved.
Collapse
|
14
|
Ahmadi A, Renaud JM, Promislow S, Burwash IG, Dwivedi G, Klein R, Zelt JGE, deKemp RA, Beanlands RS, Mielniczuk LM. Increased myocardial oxygen consumption rates are associated with maladaptive right ventricular remodeling and decreased event-free survival in heart failure patients. J Nucl Cardiol 2021; 28:2784-2795. [PMID: 32383088 DOI: 10.1007/s12350-020-02144-x] [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: 02/14/2019] [Accepted: 02/14/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Reduced left ventricular (LV) function is associated with increased myocardial oxygen consumption rate (MVO2) and altered sympathetic activity, the role of which is not well described in right ventricular (RV) dysfunction. METHODS AND RESULTS 33 patients with left heart failure were assessed for RV function/size using echocardiography. Positron emission tomography (PET) was used to measure 11C-acetate clearance rate (kmono), 11C-hydroxyephedrine (11C-HED) standardized uptake value (SUV), and retention rate. RV MVO2 was estimated from kmono. 11C-HED SUV and retention indicated sympathetic neuronal function. A composite clinical endpoint was defined as unplanned cardiac hospitalization within 5 years. Patients with (n = 10) or without (n = 23) RV dysfunction were comparable in terms of sex (male: 70.0 vs 69.5%), LV ejection fraction (39.6 ± 9.0 vs 38.6 ± 9.4%), and systemic hypertension (70.0 vs 78.3%). RV dysfunction patients were older (70.9 ± 13.5 vs 59.4 ± 11.5 years; P = .03) and had a higher prevalence of pulmonary hypertension (60.0% vs 13.0%; P = .01). RV dysfunction was associated with increased RV MVO2 (.106 ± .042 vs .068 ± .031 mL/min/g; P = .02) and decreased 11C-HED SUV and retention (6.05 ± .53 vs 7.40 ± 1.39 g/mL (P < .001) and .08 ± .02 vs .11 ± .03 mL/min/g (P < .001), respectively). Patients with an RV MVO2 above the median had a shorter event-free survival (hazard ratio = 5.47; P = .01). Patients who died within the 5-year follow-up period showed a trend (not statistically significant) for higher RV MVO2 (.120 ± .026 vs .074 ± .038 mL/min/g; P = .05). CONCLUSIONS RV dysfunction is associated with increased oxygen consumption (also characterized by a higher risk for cardiac events) and impaired RV sympathetic function.
Collapse
Affiliation(s)
- Ali Ahmadi
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada
| | - Jennifer M Renaud
- National Cardiac PET Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Steven Promislow
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada
| | - Ian G Burwash
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada
| | - Girish Dwivedi
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, Australia
| | - Ran Klein
- Division of Nuclear Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Jason G E Zelt
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Robert A deKemp
- National Cardiac PET Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Rob S Beanlands
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada
- National Cardiac PET Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Lisa M Mielniczuk
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada.
| |
Collapse
|
15
|
Hansen KB, Sörensen J, Hansson NH, Nielsen R, Larsen AH, Frøkiær J, Tolbod LP, Gormsen LC, Harms HJ, Wiggers H. Myocardial efficiency in patients with different aetiologies and stages of heart failure. Eur Heart J Cardiovasc Imaging 2021; 23:328-337. [PMID: 34751738 DOI: 10.1093/ehjci/jeab227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/15/2021] [Indexed: 11/12/2022] Open
Abstract
AIMS Myocardial external efficiency (MEE) is the ratio of cardiac work in relation with energy expenditure. We studied MEE in patients with different aetiologies and stages of heart failure (HF) to discover the role and causes of deranged MEE. In addition, we explored the impact of patient characteristics such as sex, body mass index (BMI), and age on myocardial energetics. METHODS AND RESULTS Cardiac energetic profiles were assessed with 11C-acetate positron emission tomography (PET) and left ventricular ejection fraction (LVEF) was acquired with echocardiography. MEE was studied in 121 participants: healthy controls (n = 20); HF patients with reduced (HFrEF; n = 25) and mildly reduced (HFmrEF; n = 23) LVEF; and patients with asymptomatic (AS-asymp; n = 38) and symptomatic (AS-symp; n = 15) aortic stenosis (AS). Reduced MEE coincided with symptoms of HF irrespective of aetiology and declined in tandem with deteriorating LVEF. Patients with AS-symp and HFmrEF had reduced MEE as compared with controls (22.2 ± 4.9%, P = 0.041 and 20.0 ± 4.2%, P < 0.001 vs. 26.1 ± 5.8% in controls) and a further decline was observed in patients with HFrEF (14.7 ± 6.3%, P < 0.001). Disproportionate left ventricular hypertrophy was a major cause of reduced MEE. Female sex (P < 0.001), a lower BMI (P = 0.001), and advanced age (P = 0.03) were associated with a lower MEE. CONCLUSION MEE was reduced in patients with HFrEF, HFmrEF, and HF due to pressure overload and MEE may therefore constitute a treatment target in HF. Patients with LVH, advanced age, female sex, and low BMI had more pronounced reduction in MEE and personalized treatment within these patient subgroups could be relevant.
Collapse
Affiliation(s)
- Kristoffer Berg Hansen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus N, Denmark
| | - Jens Sörensen
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus N, Denmark.,Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus N, Denmark.,Department of Surgical Sciences, Nuclear Medicine, Uppsala University, Uppsala, Sweden
| | - Nils Henrik Hansson
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Roni Nielsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Anders Hostrup Larsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Jørgen Frøkiær
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus N, Denmark
| | - Lars Poulsen Tolbod
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus N, Denmark
| | - Lars Christian Gormsen
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus N, Denmark.,Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus N, Denmark
| | - Hendrik Johannes Harms
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus N, Denmark
| | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
16
|
Tougaard RS, Laustsen C, Lassen TR, Qi H, Lindhardt JL, Schroeder M, Jespersen NR, Hansen ESS, Ringgaard S, Bøtker HE, Kim WY, Stødkilde-Jørgensen H, Wiggers H. Remodeling after myocardial infarction and effects of heart failure treatment investigated by hyperpolarized [1- 13 C]pyruvate magnetic resonance spectroscopy. Magn Reson Med 2021; 87:57-69. [PMID: 34378800 DOI: 10.1002/mrm.28964] [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: 02/06/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/10/2022]
Abstract
PURPOSE Hyperpolarized [1-13 C]pyruvate MRS can measure cardiac metabolism in vivo. We investigated whether [1-13 C]pyruvate MRS could predict left ventricular remodeling following myocardial infarction (MI), long-term left ventricular effects of heart failure medication, and could identify responders to treatment. METHODS Thirty-five rats were scanned with hyperpolarized [1-13 C]pyruvate MRS 3 days after MI or sham surgery. The animals were re-examined after 30 days of therapy with β-blockers and ACE-inhibitors (active group, n = 12), placebo treatment (placebo group, n = 13) or no treatment (sham group, n = 10). Furthermore, heart tissue mitochondrial respiratory capacity was assessed by high-resolution respirometry. Metabolic results were compared between groups, over time and correlated to functional MR data at each time point. RESULTS At 30 ± 0.5 days post MI, left ventricular ejection fraction (LVEF) differed between groups (sham, 77% ± 1%; placebo, 52% ± 3%; active, 63% ± 2%, P < .001). Cardiac metabolism, measured by both hyperpolarized [1-13 C]pyruvate MRS and respirometry, neither differed between groups nor between baseline and follow-up. Three days post MI, low bicarbonate + CO2 /pyruvate ratio was associated with low LVEF. At follow-up, in the active group, a poor recovery of LVEF was associated with high bicarbonate + CO2 /pyruvate ratio, as measured by hyperpolarized MRS. CONCLUSION In a rat model of moderate heart failure, medical treatment improved function, but did not on average influence [1-13 C]pyruvate flux as measured by MRS; however, responders to heart failure medication had reduced capacity for carbohydrate metabolism.
Collapse
Affiliation(s)
- Rasmus Stilling Tougaard
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | - Haiyun Qi
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Jakob Lykke Lindhardt
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Marie Schroeder
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | | | - Steffen Ringgaard
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Won Yong Kim
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
17
|
Saeed S, Holm H, Nilsson PM. Ventricular-arterial coupling: definition, pathophysiology and therapeutic targets in cardiovascular disease. Expert Rev Cardiovasc Ther 2021; 19:753-761. [PMID: 34252318 DOI: 10.1080/14779072.2021.1955351] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The heart and arterial system are equally affected by arteriosclerosis/atherosclerosis. There is a constant interaction between the left ventricular (LV) function and the arterial system, termed ventricular-arterial coupling (VAC), which reflects the global cardiovascular efficiency. VAC is traditionally assessed by echocardiography as the ratio of effective arterial elastance (Ea) over end-systolic elastance (Ees) (Ea/Ees). However, the concept of VAC is evolving and new methods have been proposed such as the ratio of pulse wave velocity (PWV) to global longitudinal strain (GLS) and myocardial work index. AREA COVERED This clinical review presents the hemodynamic background of VAC, its clinical implications and the impact of therapeutic interventions to normalize VAC. The review also summarizes the detrimental effects of cardio-metabolic risk factors on the aorta and LV, and provides an update on arterial load and its impact on LV function. The narrative review is based upon a systemic search of the bibliographic database PubMed for publications on VAC. EXPERT OPINION Newer methods such as PWV/GLS-ratio may be a superior marker of VAC than the traditional echocardiographic Ea/Ees in predicting target organ damage and its association with clinical outcomes. Novel anti-diabetic drugs and optimal antihypertensive treatment may normalize VAC in high-risk patients.
Collapse
Affiliation(s)
- Sahrai Saeed
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Hannes Holm
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden.,Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
18
|
Gullberg GT, Shrestha UM, Veress AI, Segars WP, Liu J, Ordovas K, Seo Y. Novel Methodology for Measuring Regional Myocardial Efficiency. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:1711-1725. [PMID: 33690114 PMCID: PMC8325923 DOI: 10.1109/tmi.2021.3065219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Our approach differs from the usual global measure of cardiac efficiency by using PET/MRI to measure efficiency of small pieces of cardiac tissue whose limiting size is equal to the spatial resolution of the PET scanner. We initiated a dynamic cardiac PET study immediately prior to the injection of 15.1 mCi of 11C-acetate acquiring data for 25 minutes while simultaneously acquiring MRI cine data. 1) A 3D finite element (FE) biomechanical model of the imaged heart was constructed by utilizing nonrigid deformable image registration to alter the Dassault Systèmes FE Living Heart Model (LHM) to fit the geometry in the cardiac MRI cine data. The patient specific FE cardiac model with estimates of stress, strain, and work was transformed into PET/MRI format. 2) A 1-tissue compartment model was used to calculate wash-in (K1) and the linear portion of the decay in the PET 11C-acetate time activity curve (TAC) was used to calculate the wash-out k2(mono) rate constant. K1 was used to calculate blood flow and k2(mono) was used to calculate myocardial volume oxygen consumption ( MVO2 ). 3) Estimates of stress and strain were used to calculate Myocardial Equivalent Minute Work ( MEMW ) and Cardiac Efficiency = MEMW/MVO2 was then calculated for 17 tissue segments of the left ventricle. The global MBF was 0.96 ± 0.15 ml/min/gm and MVO2 ranged from 8 to 17 ml/100gm/min. Six central slices of the MRI cine data provided a range of MEMW of 0.1 to 0.4 joules/gm/min and a range of Cardiac Efficiency of 6 to 18%.
Collapse
|
19
|
Perez DM. Targeting Adrenergic Receptors in Metabolic Therapies for Heart Failure. Int J Mol Sci 2021; 22:5783. [PMID: 34071350 PMCID: PMC8198887 DOI: 10.3390/ijms22115783] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
The heart has a reduced capacity to generate sufficient energy when failing, resulting in an energy-starved condition with diminished functions. Studies have identified numerous changes in metabolic pathways in the failing heart that result in reduced oxidation of both glucose and fatty acid substrates, defects in mitochondrial functions and oxidative phosphorylation, and inefficient substrate utilization for the ATP that is produced. Recent early-phase clinical studies indicate that inhibitors of fatty acid oxidation and antioxidants that target the mitochondria may improve heart function during failure by increasing compensatory glucose oxidation. Adrenergic receptors (α1 and β) are a key sympathetic nervous system regulator that controls cardiac function. β-AR blockers are an established treatment for heart failure and α1A-AR agonists have potential therapeutic benefit. Besides regulating inotropy and chronotropy, α1- and β-adrenergic receptors also regulate metabolic functions in the heart that underlie many cardiac benefits. This review will highlight recent studies that describe how adrenergic receptor-mediated metabolic pathways may be able to restore cardiac energetics to non-failing levels that may offer promising therapeutic strategies.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
20
|
Turgeon RD, Barry AR, Hawkins NM, Ellis UM. Pharmacotherapy for heart failure with reduced ejection fraction and health-related quality of life: a systematic review and meta-analysis. Eur J Heart Fail 2021; 23:578-589. [PMID: 33634543 DOI: 10.1002/ejhf.2141] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 01/27/2021] [Accepted: 02/23/2021] [Indexed: 01/08/2023] Open
Abstract
AIMS The aim of this study was to synthesize the evidence on the effect of heart failure with reduced ejection fraction (HFrEF) pharmacotherapy on health-related quality of life (HRQoL). METHODS AND RESULTS We searched MEDLINE, Embase, CENTRAL, CINAHL, ClinicalTrials.gov and the World Health Organization International Clinical Trials Registry Platform in June 2020. Randomized placebo-controlled trials evaluating contemporary HFrEF pharmacotherapy and reporting HRQoL as an outcome were included. Two reviewers independently assessed studies for eligibility, extracted data, and assessed risk of bias and GRADE certainty of evidence. The primary outcome was HRQoL at last available follow-up analysed using a random-effects model. We included 37 studies from 5770 identified articles. Risk of bias was low in 10 trials and high/unclear in 27 trials. High certainty evidence from meta-analyses demonstrated improved HRQoL over placebo with sodium-glucose co-transporter 2 (SGLT2) inhibitors [standardized mean difference (SMD) 0.16, 95% confidence interval (CI) 0.08-0.23] and intravenous iron (SMD 0.52, 95% CI 0.04-1.00). Furthermore, high certainty evidence from ≥1 landmark trial further supported improved HRQoL with angiotensin receptor blockers (ARBs) (SMD 0.09, 95% CI 0.02-0.17), ivabradine (SMD 0.14, 95% CI 0.04-0.23), hydralazine-nitrate (SMD 0.24, 95% CI 0.04-0.44) vs. placebo, and for angiotensin receptor-neprilysin inhibitor (ARNI) compared with an angiotensin-converting enzyme (ACE) inhibitor (SMD 0.09, 95% CI 0.02-0.17). Findings were inconclusive for ACE inhibitors, beta-blockers, digoxin, and oral iron based on low-to-moderate certainty evidence. CONCLUSION ARBs, ARNIs, SGLT2 inhibitors, ivabradine, hydralazine-nitrate, and intravenous iron improved HRQoL in patients with HFrEF. These findings can be incorporated into discussions with patients to enable shared decision-making.
Collapse
Affiliation(s)
- Ricky D Turgeon
- Greg Moore Professorship in Clinical & Community Cardiovascular Pharmacy, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, Vancouver, Canada
| | - Arden R Barry
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada.,Chilliwack General Hospital, Lower Mainland Pharmacy Services, Chilliwack, Canada
| | | | - Ursula M Ellis
- Woodward Library, University of British Columbia, Vancouver, Canada
| |
Collapse
|
21
|
Gropler RJ. Imaging Myocardial Metabolism. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00083-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
22
|
Congestive Heart Failure. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00050-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
23
|
Farber G, Boczar KE, Wiefels CC, Zelt JG, Guler EC, deKemp RA, Beanlands RS, Rotstein BH. The Future of Cardiac Molecular Imaging. Semin Nucl Med 2020; 50:367-385. [DOI: 10.1053/j.semnuclmed.2020.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Badolia R, Ramadurai DKA, Abel ED, Ferrin P, Taleb I, Shankar TS, Krokidi AT, Navankasattusas S, McKellar SH, Yin M, Kfoury AG, Wever-Pinzon O, Fang JC, Selzman CH, Chaudhuri D, Rutter J, Drakos SG. The Role of Nonglycolytic Glucose Metabolism in Myocardial Recovery Upon Mechanical Unloading and Circulatory Support in Chronic Heart Failure. Circulation 2020; 142:259-274. [PMID: 32351122 DOI: 10.1161/circulationaha.119.044452] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Significant improvements in myocardial structure and function have been reported in some patients with advanced heart failure (termed responders [R]) following left ventricular assist device (LVAD)-induced mechanical unloading. This therapeutic strategy may alter myocardial energy metabolism in a manner that reverses the deleterious metabolic adaptations of the failing heart. Specifically, our previous work demonstrated a post-LVAD dissociation of glycolysis and oxidative-phosphorylation characterized by induction of glycolysis without subsequent increase in pyruvate oxidation through the tricarboxylic acid cycle. The underlying mechanisms responsible for this dissociation are not well understood. We hypothesized that the accumulated glycolytic intermediates are channeled into cardioprotective and repair pathways, such as the pentose-phosphate pathway and 1-carbon metabolism, which may mediate myocardial recovery in R. METHODS We prospectively obtained paired left ventricular apical myocardial tissue from nonfailing donor hearts as well as R and nonresponders at LVAD implantation (pre-LVAD) and transplantation (post-LVAD). We conducted protein expression and metabolite profiling and evaluated mitochondrial structure using electron microscopy. RESULTS Western blot analysis shows significant increase in rate-limiting enzymes of pentose-phosphate pathway and 1-carbon metabolism in post-LVAD R (post-R) as compared with post-LVAD nonresponders (post-NR). The metabolite levels of these enzyme substrates, such as sedoheptulose-6-phosphate (pentose phosphate pathway) and serine and glycine (1-carbon metabolism) were also decreased in Post-R. Furthermore, post-R had significantly higher reduced nicotinamide adenine dinucleotide phosphate levels, reduced reactive oxygen species levels, improved mitochondrial density, and enhanced glycosylation of the extracellular matrix protein, α-dystroglycan, all consistent with enhanced pentose-phosphate pathway and 1-carbon metabolism that correlated with the observed myocardial recovery. CONCLUSIONS The recovering heart appears to direct glycolytic metabolites into pentose-phosphate pathway and 1-carbon metabolism, which could contribute to cardioprotection by generating reduced nicotinamide adenine dinucleotide phosphate to enhance biosynthesis and by reducing oxidative stress. These findings provide further insights into mechanisms responsible for the beneficial effect of glycolysis induction during the recovery of failing human hearts after mechanical unloading.
Collapse
Affiliation(s)
- Rachit Badolia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.).,Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Dinesh K A Ramadurai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.)
| | - E Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City (E.D.A.)
| | - Peter Ferrin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.)
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.).,Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.)
| | - Aspasia Thodou Krokidi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.)
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.)
| | - Stephen H McKellar
- Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Michael Yin
- Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Abdallah G Kfoury
- Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Omar Wever-Pinzon
- Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - James C Fang
- Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Craig H Selzman
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.).,Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.)
| | - Jared Rutter
- Department of Biochemistry, University of Utah and Howard Hughes Medical Institute, Salt Lake City (J.R.)
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (R.B., D.K.A.R., P.F., I.T., T.S.S., A.T.K., S.N., C.H.S., D.C., S.G.D.).,Utah Transplant Affiliated Hospitals Cardiac Transplant Program, University of Utah Healthcare and School of Medicine Intermountain Medical Center, Salt Lake VA Health Care System, Salt Lake City (R.B., I.T., S.H.M., M.Y., A.G.K., O.W.-P., J.C.F., C.H.S., S.G.D.)
| |
Collapse
|
25
|
Mancusi C, de Simone G, Brguljan Hitij J, Sudano I, Mahfoud F, Parati G, Kahan T, Barbato E, Pierard LA, Garbi M, Flachskampf FA, Gerdts E. Management of patients with combined arterial hypertension and aortic valve stenosis: a consensus document from the Council on Hypertension and Council on Valvular Heart Disease of the European Society of Cardiology, the European Association of Cardiovascular Imaging (EACVI), and the European Association of Percutaneous Cardiovascular Interventions (EAPCI). EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2020; 7:242-250. [PMID: 32353143 DOI: 10.1093/ehjcvp/pvaa040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/15/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
Aortic valve stenosis (AS) is the third most common cardiovascular disease. The prevalence of both AS and arterial hypertension increases with age, and the conditions therefore often co-exist. Co-existence of AS and arterial hypertension is associated with higher global left ventricular (LV) pressure overload, more abnormal LV geometry and function, and more adverse cardiovascular outcome. Arterial hypertension may also influence grading of AS, leading to underestimation of the true AS severity. Current guidelines suggest re-assessing patients once arterial hypertension is controlled. Management of arterial hypertension in AS has historically been associated with prudence and concerns, mainly related to potential adverse consequences of drug-induced peripheral vasodilatation combined with reduced stroke volume due to the fixed LV outflow obstruction. Current evidence suggests that patients should be treated with antihypertensive drugs blocking the renin-angiotensin-aldosterone system, adding further drug classes when required, to achieve similar target blood pressure (BP) values as in hypertensive patients without AS. The introduction of transcatheter aortic valve implantation has revolutionized the management of patients with AS, but requires proper BP management during and following valve replacement. The purpose of this document is to review the recent evidence and provide practical expert advice on management of hypertension in patients with AS.
Collapse
Affiliation(s)
- Costantino Mancusi
- Hypertension Research Center, Federico II University Hospital, Naples, Italy.,Department of Advanced Biomedical Science, Federico II University of Naples, Naples, Italy
| | - Giovanni de Simone
- Hypertension Research Center, Federico II University Hospital, Naples, Italy.,Department of Advanced Biomedical Science, Federico II University of Naples, Naples, Italy
| | - Jana Brguljan Hitij
- Hypertension Division, Department of Internal Medicine, University Medical Centre Ljubljana, Medical Faculty Ljubljana, Ljubljana, Slovenia
| | - Isabella Sudano
- Department of Cardiology, University Heart Center Zurich, University Hospital of Zürich, Zürich, Switzerland
| | - Felix Mahfoud
- Department for Cardiology, Angiology, Intensive Medicine, Saarland University Hospital, Homburg/Saar, Germany
| | - Gianfranco Parati
- Department of Medicine and Surgery, University of Milano-Bicocca & Istituto Auxologico Italiano, IRCCS, Cardiology Unit, Milan, Italy
| | - Thomas Kahan
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Emanuele Barbato
- Hypertension Research Center, Federico II University Hospital, Naples, Italy.,Department of Advanced Biomedical Science, Federico II University of Naples, Naples, Italy
| | - Luc A Pierard
- Department of Cardiology, Heart Valve Clinic, University Hospital Sart-Tilman, Liège, Belgium
| | - Madalina Garbi
- Royal Papworth Hospital NHS Foundation Trust Papworth Road, Cambridge Biomedical Campus, Cambridge, UK
| | - Frank A Flachskampf
- Department of Medical Sciences, Uppsala University, and Clinical Physiology and Cardiology, Akademiska, Uppsala, Sweden
| | - Eva Gerdts
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
26
|
Hwang IC. Myocardial Efficiency: A Reliable Load-independent Parameter of Cardiac Performance? J Cardiovasc Imaging 2020; 28:279-282. [PMID: 33086444 PMCID: PMC7572257 DOI: 10.4250/jcvi.2020.0118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 07/29/2020] [Indexed: 11/22/2022] Open
Affiliation(s)
- In Chang Hwang
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
27
|
Larsen AH, Jessen N, Nørrelund H, Tolbod LP, Harms HJ, Feddersen S, Nielsen F, Brøsen K, Hansson NH, Frøkiær J, Poulsen SH, Sörensen J, Wiggers H. A randomised, double‐blind, placebo‐controlled trial of metformin on myocardial efficiency in insulin‐resistant chronic heart failure patients without diabetes. Eur J Heart Fail 2019; 22:1628-1637. [DOI: 10.1002/ejhf.1656] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/04/2019] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Affiliation(s)
| | - Niels Jessen
- Department of Clinical Pharmacology Aarhus University Hospital Aarhus Denmark
- Department of Biomedicine Aarhus University Aarhus Denmark
- Steno Diabetes Centre Aarhus Aarhus University Hospital Aarhus Denmark
| | - Helene Nørrelund
- Department of Clinical Epidemiology, Institute of Clinical Medicine Aarhus University Hospital Aarhus Denmark
| | - Lars Poulsen Tolbod
- Department of Nuclear Medicine and PET Centre Aarhus University Hospital Aarhus Denmark
| | | | - Søren Feddersen
- Department of Clinical Biochemistry and Pharmacology Odense University Hospital Odense Denmark
| | - Flemming Nielsen
- Department of Clinical Biochemistry and Pharmacology Odense University Hospital Odense Denmark
| | - Kim Brøsen
- Department of Clinical Biochemistry and Pharmacology Odense University Hospital Odense Denmark
| | | | - Jørgen Frøkiær
- Department of Nuclear Medicine and PET Centre Aarhus University Hospital Aarhus Denmark
| | | | - Jens Sörensen
- Department of Nuclear Medicine and PET Centre Aarhus University Hospital Aarhus Denmark
- Department of Medical Sciences, Uppsala Clinical Research Centre Uppsala University Uppsala Sweden
| | - Henrik Wiggers
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
28
|
Sörensen J, Harms HJ, Aalen JM, Baron T, Smiseth OA, Flachskampf FA. Myocardial Efficiency: A Fundamental Physiological Concept on the Verge of Clinical Impact. JACC Cardiovasc Imaging 2019; 13:1564-1576. [PMID: 31864979 DOI: 10.1016/j.jcmg.2019.08.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/26/2019] [Accepted: 08/21/2019] [Indexed: 02/08/2023]
Abstract
Myocardial external efficiency is the relation of mechanical energy generated by the left (or right) ventricle to the consumed chemical energy from aerobic metabolism. Efficiency can be calculated invasively, and, more importantly, noninvasively by using positron emission tomography, providing a single parameter by which to judge the adequacy of myocardial metabolism to generated mechanical output. This parameter has been found to be impaired in heart failure of myocardial or valvular etiology, and it changes in a characteristic manner with medical or interventional cardiac therapy. The authors discuss the concept, strengths, and limitations, known applications, and future perspectives of the use of myocardial efficiency.
Collapse
Affiliation(s)
- Jens Sörensen
- Department of Nuclear Medicine and PET, Surgical Sciences, Uppsala University, Sweden; Department of Nuclear Medicine and PET, Clinical Institute, Aarhus University, Aarhus, Denmark
| | - Hendrik Johannes Harms
- Department of Nuclear Medicine and PET, Clinical Institute, Aarhus University, Aarhus, Denmark
| | - John M Aalen
- Institute for Surgical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; Center for Cardiological Innovation, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Department of Clinical Physiology, Akademiska University Hospital, Uppsala, Sweden
| | - Tomasz Baron
- Department of Medical Sciences, Uppsala University and Akademiska Hospital, Uppsala, Sweden; Department of Radiology, Uppsala University and Akademiska Hospital, Uppsala, Sweden; Department of Cardiology, Uppsala University and Akademiska Hospital, Uppsala, Sweden
| | - Otto Armin Smiseth
- Institute for Surgical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; Center for Cardiological Innovation, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Department of Radiology, Uppsala University and Akademiska Hospital, Uppsala, Sweden
| | - Frank A Flachskampf
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Department of Clinical Physiology, Akademiska University Hospital, Uppsala, Sweden.
| |
Collapse
|
29
|
Ishida M, Miyagawa S, Saito A, Fukushima S, Harada A, Ito E, Ohashi F, Watabe T, Hatazawa J, Matsuura K, Sawa Y. Transplantation of Human-induced Pluripotent Stem Cell-derived Cardiomyocytes Is Superior to Somatic Stem Cell Therapy for Restoring Cardiac Function and Oxygen Consumption in a Porcine Model of Myocardial Infarction. Transplantation 2019; 103:291-298. [PMID: 30119058 PMCID: PMC6365242 DOI: 10.1097/tp.0000000000002384] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Somatic stem cell (SC) therapy can improve cardiac performance following ischemic injury. In this study, we investigated whether induced pluripotent SC-derived cardiomyocytes (iPS-CMs) are more effective than somatic SCs, such as skeletal myoblasts (SM) and mesenchymal (M)SCs, in promoting functional recovery upon transplantation in a porcine model of myocardial infarction. Methods Myocardial injury was induced by ameroid ring placement in immunosuppressed female mini pigs; after 1 month, epicardial cell transplantation was performed with iPS-CMs (n = 7), SMs (n = 7), and MSCs (n = 7). Control pigs underwent sham operation (n = 8). Results Cell therapy improved functional recovery 2 months after myocardial infarction, as evidenced by increased ejection fraction (iPS-CM, +7.3% ± 2.2% and SM, +5.8% ± 5.4% vs control, −4.4% ± 3.8%; P < 0.05). The analysis of regional contractile function in the infarcted zone revealed an increase in transverse peak strain (iPS-CM, +4.6% ± 2.2% vs control, −3.8% ± 4.7%; P < 0.05). The C-11 acetate kinetic analysis by positron emission tomography showed that the work-metabolic cardiac energy efficacy increased by the transplantation of iPS-CMs, but was reduced by the other cell types. This was accompanied by decreased myocardial wall stress in the infarcted zone (iPS-CM, −27.6 ± 32.3 Pa and SM, −12.8 ± 27 Pa vs control, +40.5 ± 33.9 Pa; P < 0.05). Conclusions The iPS-CM is superior to other somatic cell sources in terms of improving regional contractile function and cardiac bioenergetic efficiency, suggesting greater clinical benefits in severely damaged myocardium. The authors compare the therapeutic effects among human induced pluripotent stem cell-derived cardiomyocytes, skeletal myoblasts stem cells and mesenchymal stem cells on promoting cardiac functional recovery including regional contractile function and cardiac bioenergetic efficiency in a porcine model of myocardial infarction. Supplemental digital content is available in the text.
Collapse
Affiliation(s)
- Masaru Ishida
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Miyagawa
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsuhiro Saito
- Medical Center for Translational Research, Osaka University Hospital, Osaka, Japan
| | - Satsuki Fukushima
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akima Harada
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Emiko Ito
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumiya Ohashi
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Hatazawa
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Yoshiki Sawa
- Division of Surgery, Department of Cardiovascular Surgery, Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
30
|
Grande D, Iacoviello M, Aspromonte N. The effects of heart rate control in chronic heart failure with reduced ejection fraction. Heart Fail Rev 2019; 23:527-535. [PMID: 29687317 DOI: 10.1007/s10741-018-9704-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Elevated heart rate has been associated with worse prognosis both in the general population and in patients with heart failure. Heart rate is finely modulated by neurohormonal signals and it reflects the balance between the sympathetic and the parasympathetic limbs of the autonomic nervous system. For this reason, elevated heart rate in heart failure has been considered an epiphenomenon of the sympathetic hyperactivation during heart failure. However, experimental and clinical evidence suggests that high heart rate could have a direct pathogenetic role. Consequently, heart rate might act as a pathophysiological mediator of heart failure as well as a marker of adverse outcome. This hypothesis has been supported by the observation that the positive effect of beta-blockade could be linked to the degree of heart rate reduction. In addition, the selective heart rate control with ivabradine has recently been demonstrated to be beneficial in patients with heart failure and left ventricular systolic dysfunction. The objective of this review is to examine the pathophysiological implications of elevated heart rate in chronic heart failure and explore the mechanisms underlying the effects of pharmacological heart rate control.
Collapse
Affiliation(s)
- Dario Grande
- School of Cardiology, University of Bari, Bari, Italy
| | - Massimo Iacoviello
- Cardiology Unit, Cardiothoracic Department, Policlinic University Hospital, Bari, Italy
| | - Nadia Aspromonte
- Department of Cardiovascular Medicine, Foundation Policlinico Gemelli, Rome, Italy.
| |
Collapse
|
31
|
Hansson NH, Harms HJ, Kim WY, Nielsen R, Tolbod LP, Frøkiær J, Bouchelouche K, Poulsen SH, Wiggers H, Parner ET, Sörensen J. Test-retest repeatability of myocardial oxidative metabolism and efficiency using standalone dynamic 11C-acetate PET and multimodality approaches in healthy controls. J Nucl Cardiol 2018; 25:1929-1936. [PMID: 29855984 DOI: 10.1007/s12350-018-1302-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/25/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Myocardial efficiency measured by 11C-acetate positron emission tomography (PET) has successfully been used in clinical research to quantify mechanoenergetic coupling. The objective of this study was to establish the repeatability of myocardial external efficiency (MEE) and work metabolic index (WMI) by non-invasive concepts. METHODS AND RESULTS Ten healthy volunteers (63 ± 4 years) were examined twice, one week apart, using 11C-acetate PET, cardiovascular magnetic resonance (CMR), and echocardiography. Myocardial oxygen consumption from PET was combined with stroke work data from CMR, echocardiography, or PET to obtain MEE and WMI for each modality. Repeatability was estimated as the coefficient of variation (CV) between test and retest. MEECMR, MEEEcho, and MEEPET values were 21.9 ± 2.7%, 16.4 ± 3.7%, and 23.8 ± 4.9%, respectively, P < .001. WMICMR, WMIEcho, and WMIPET values were 4.42 ± 0.90, 4.07 ± 0.63, and 4.58 ± 1.13 mmHg × mL/m2 × 106, respectively, P = .45. Repeatability for MEECMR was superior compared with MEEEcho but did not differ significantly compared with MEEPET (6.3% vs 12.9% and 9.4%, P = .04 and .25). CV values for WMICMR, WMIEcho, and WMIPET were 10.0%, 14.8%, and 12.0%, respectively, (P = .53). CONCLUSIONS Non-invasive measurements of MEE using 11C-acetate PET are highly repeatable. A PET-only approach did not differ significantly from CMR/PET and might facilitate further clinical research due to lower costs and broader applicability.
Collapse
Affiliation(s)
| | - Hendrik Johannes Harms
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | - Won Yong Kim
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Roni Nielsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars P Tolbod
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Frøkiær
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | - Kirsten Bouchelouche
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | | | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Erik Thorlund Parner
- Section for Biostatistics, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Jens Sörensen
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
32
|
Wu KY, Dinculescu V, Renaud JM, Chen SY, Burwash IG, Mielniczuk LM, Beanlands RSB, deKemp RA. Repeatable and reproducible measurements of myocardial oxidative metabolism, blood flow and external efficiency using 11C-acetate PET. J Nucl Cardiol 2018; 25:1912-1925. [PMID: 29453603 DOI: 10.1007/s12350-018-1206-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Non-invasive approaches to investigate myocardial efficiency can help track the progression of heart failure (HF). This study evaluates the repeatability and reproducibility of 11C-acetate positron emission tomography (PET) imaging of oxidative metabolism. METHODS AND RESULTS Dynamic 11C-acetate PET scans were performed at baseline and followup (47 ± 22 days apart) in 20 patients with stable HF with reduced ejection fraction. Two observers blinded to patients' clinical data used FlowQuant® to evaluate test-retest repeatability, as well as intra- and inter-observer reproducibility of 11C-acetate tracer uptake and clearance rates, for the measurement of myocardial oxygen consumption (MVO2), myocardial external efficiency (MEE), work metabolic index (WMI), and myocardial blood flow. Reproducibility and repeatability were evaluated using intra-class-correlation (ICC) and Bland-Altman coefficient-of-repeatability (CR). Test-retest correlations and repeatability were better for MEE and WMI compared to MVO2. All intra- and inter-observer correlations were excellent (ICC = 0.95-0.99) and the reproducibility values (CR = 3%-6%) were significantly lower than the test-retest repeatability values (22%-54%, P < 0.001). Repeatability was improved for all parameters using a newer PET-computed tomography (CT) scanner compared to older PET-only instrumentation. CONCLUSION 11C-acetate PET measurements of WMI and MEE exhibited excellent test-retest repeatability and operator reproducibility. Newer PET-CT scanners may be preferred for longitudinal tracking of cardiac efficiency.
Collapse
Affiliation(s)
- Kai Yi Wu
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Vincent Dinculescu
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Jennifer M Renaud
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Shin-Yee Chen
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Ian G Burwash
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Lisa M Mielniczuk
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Rob S B Beanlands
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada
| | - Robert A deKemp
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y4W7, Canada.
| |
Collapse
|
33
|
Harms HJ, Hansson NHS, Kero T, Baron T, Tolbod LP, Kim WY, Frøkiær J, Flachskampf FA, Wiggers H, Sörensen J. Automatic calculation of myocardial external efficiency using a single 11C-acetate PET scan. J Nucl Cardiol 2018; 25:1937-1944. [PMID: 29946824 PMCID: PMC6280778 DOI: 10.1007/s12350-018-1338-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/25/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Myocardial external efficiency (MEE) is defined as the ratio of kinetic energy associated with cardiac work [forward cardiac output (FCO)*mean systemic pressure] and the chemical energy from oxygen consumed (MVO2) by the left ventricular mass (LVM). We developed a fully automated method for estimating MEE based on a single 11C-acetate PET scan without ECG-gating. METHODS AND RESULTS Ten healthy controls, 34 patients with aortic valve stenosis (AVS), and 20 patients with mitral valve regurgitation (MVR) were recruited in a dual-center study. MVO2 was calculated using washout of 11C -acetate activity. FCO and LVM were calculated automatically using dynamic PET and parametric image formation. FCO and LVM were also obtained using cardiac magnetic resonance (CMR) in all subjects. The correlation between MEEPET-CMR and MEEPET was high (r = 0.85, P < 0.001) without significant bias. MEEPET was 23.6 ± 4.2% for controls and was lowered in AVS (17.2 ± 4.3%, P < 0.001) and in MVR (18.0 ± 5.2%, P = 0.004). MEEPET was strongly associated with both NYHA class (P < 0.001) and the magnitude of valvular dysfunction (mean aortic gradient: P < 0.001, regurgitant fraction: P = 0.009). CONCLUSION A single 11C-acetate PET yields accurate and automated MEE results on different scanners. MEE might provide an unbiased measurement of the phenotypic response to valvular disease.
Collapse
Affiliation(s)
- Hendrik J. Harms
- Department of Nuclear Medicine, & PET Center, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | | | - Tanja Kero
- Department of Nuclear Medicine & PET, Uppsala University Hospital, Uppsala, Sweden
| | - Tomasz Baron
- Department of Medical Sciences, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Lars P. Tolbod
- Department of Nuclear Medicine, & PET Center, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Won Y. Kim
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Frøkiær
- Department of Nuclear Medicine, & PET Center, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Frank A. Flachskampf
- Department of Medical Sciences, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Sörensen
- Department of Nuclear Medicine, & PET Center, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- Department of Nuclear Medicine & PET, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
34
|
Gewirtz H. Tight software, methodological/physiological variation, less so. J Nucl Cardiol 2018; 25:1926-1928. [PMID: 29453602 DOI: 10.1007/s12350-018-1209-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 12/14/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Henry Gewirtz
- Department of Medicine (Cardiology Division), Massachusetts General Hospital, Harvard Medical School, Yawkey 5E (Nuclear Cardiology), 55 Fruit St, Boston, MA, 02114, USA.
| |
Collapse
|
35
|
Bengel FM. PET-based myocardial efficiency: Powerful yet under-utilized-now simpler than ever. J Nucl Cardiol 2018; 25:1945-1947. [PMID: 30097851 DOI: 10.1007/s12350-018-1400-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
36
|
Manabe O, Kikuchi T, Scholte AJHA, El Mahdiui M, Nishii R, Zhang MR, Suzuki E, Yoshinaga K. Radiopharmaceutical tracers for cardiac imaging. J Nucl Cardiol 2018; 25:1204-1236. [PMID: 29196910 PMCID: PMC6133155 DOI: 10.1007/s12350-017-1131-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death and disease burden worldwide. Nuclear myocardial perfusion imaging with either single-photon emission computed tomography or positron emission tomography has been used extensively to perform diagnosis, monitor therapies, and predict cardiovascular events. Several radiopharmaceutical tracers have recently been developed to evaluate CVD by targeting myocardial perfusion, metabolism, innervation, and inflammation. This article reviews old and newer used in nuclear cardiac imaging.
Collapse
Affiliation(s)
- Osamu Manabe
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tatsuya Kikuchi
- Department of Radiopharmaceutical Development, National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Arthur J H A Scholte
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mohammed El Mahdiui
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ryuichi Nishii
- Diagnostic and Therapeutic Nuclear Medicine, National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-Ku, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Radiopharmaceutical Development, National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Eriko Suzuki
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Keiichiro Yoshinaga
- Diagnostic and Therapeutic Nuclear Medicine, National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-Ku, Chiba, 263-8555, Japan.
| |
Collapse
|
37
|
Pituskin E, Mackey JR, Koshman S, Jassal D, Pitz M, Haykowsky MJ, Pagano JJ, Chow K, Thompson RB, Vos LJ, Ghosh S, Oudit GY, Ezekowitz JA, Paterson DI. Multidisciplinary Approach to Novel Therapies in Cardio-Oncology Research (MANTICORE 101-Breast): A Randomized Trial for the Prevention of Trastuzumab-Associated Cardiotoxicity. J Clin Oncol 2016; 35:870-877. [PMID: 27893331 DOI: 10.1200/jco.2016.68.7830] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose The primary toxicity of trastuzumab therapy for human epidermal growth factor receptor 2-overexpressing (HER2-positive) breast cancer is dose-independent cardiac dysfunction. Angiotensin-converting enzyme inhibitors and β-blockers are recommended first-line agents for heart failure. We hypothesized that angiotensin-converting enzyme inhibitors and β-blockers could prevent trastuzumab-related cardiotoxicity. Patients and Methods In this double-blinded, placebo-controlled trial, patients with HER2-positive early breast cancer were randomly assigned to receive treatment with perindopril, bisoprolol, or placebo (1:1:1) for the duration of trastuzumab adjuvant therapy. Patients underwent cardiac magnetic resonance imaging at baseline and post-cycle 17 for the determination of left ventricular volumes and left ventricular ejection fraction (LVEF). Cardiotoxicity was evaluated as the change in indexed left ventricular end diastolic volume and LVEF. Results Thirty-three patients received perindopril, 31 received bisoprolol, and 30 received placebo. Baseline demographic, cancer, and cardiovascular profiles were similar between groups. Study drugs were well tolerated with no serious adverse events. After 17 cycles of trastuzumab, indexed left ventricular end diastolic volume increased in patients treated with perindopril (+7 ± 14 mL/m2), bisoprolol (+8 mL ± 9 mL/m2), and placebo (+4 ± 11 mL/m2; P = .36). In secondary analyses, trastuzumab-mediated decline in LVEF was attenuated in bisoprolol-treated patients (-1 ± 5%) relative to the perindopril (-3 ± 4%) and placebo (-5 ± 5%) groups ( P = .001). Perindopril and bisoprolol use were independent predictors of maintained LVEF on multivariable analysis. Conclusion Perindopril and bisoprolol were well tolerated in patients with HER2-positive early breast cancer who received trastuzumab and protected against cancer therapy-related declines in LVEF; however, trastuzumab-mediated left ventricular remodeling-the primary outcome-was not prevented by these pharmacotherapies.
Collapse
Affiliation(s)
- Edith Pituskin
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John R Mackey
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sheri Koshman
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder Jassal
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marshall Pitz
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mark J Haykowsky
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Joseph J Pagano
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kelvin Chow
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard B Thompson
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Larissa J Vos
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sunita Ghosh
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gavin Y Oudit
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Justin A Ezekowitz
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| | - D Ian Paterson
- Edith Pituskin John R. Mackey, Sheri Koshman, Mark J. Haykowsky, Joseph J. Pagano, Kelvin Chow, Richard B. Thompson, Larissa J. Vos, Sunita Ghosh, Gavin Y. Oudit, Justin A. Ezekowitz, and D. Ian Paterson, University of Alberta, Edmonton, Alberta; and Davinder Jassal and Marshall Pitz, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
38
|
Jatain S, Kapoor A, Sinha A, Khanna R, Kumar S, Garg N, Tewari S, Goel P. Metabolic manipulation in dilated cardiomyopathy: Assessing the role of trimetazidine. Indian Heart J 2016; 68:803-808. [PMID: 27931551 PMCID: PMC5143816 DOI: 10.1016/j.ihj.2016.04.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 04/26/2016] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVES To study the role of metabolic modulator (trimetazidine: TMZ) in dilated cardiomyopathy (DCM). Optimizing altered substrate metabolism in heart failure (HF) with metabolic modulators allows more efficacious energy production from glucose than from free fatty acids. METHODS 100 patients of DCM (47.7 years, NYHA class 2.17, LVEF 27.3%) were randomized to TMZ (20mg tid, n=50) vs conventional therapy (n=50). Functional status, BNP and various echocardiographic parameters were assessed at 3-6 months. RESULTS At 3 months, TMZ group had significantly improved NYHA class (2.25 vs 1.85), 6min walk test (349.7 vs 402m), LVD-36 score (25.5 vs 21) and BNP (744.7 vs 248.3pg/ml), all p 0.001. Significant improvement was also seen in LV end-systolic (LVESV, 87.1±27.5 vs 78.5±24.9ml/m2, p 0.001), LV end-diastolic volumes (LVEDV, 117.6±29.3 vs 110.9±27.4ml/m2, p 0.001), LVEF (27 vs 30.9%, p 0.001) and LV wall stress (90.2±18.9 vs 71.1±13.2dyn/cm2, p 0.0001). The % change in LVESV, LVEDV, LVEF and LV wall stress was -9.5%, -5.4%, +8.4% and -21.8%. Other echo parameters also improved after 3 months of TMZ (E/A ratio 1.9 vs 1.2, p=0.001, E/A VTI 2.7 vs 1.6, p=0.001, myocardial performance index, MPI 0.8 vs 0.7, p=0.0001), Tissue Doppler parameters (E/E' septal (19.7 vs 12.5, p=0.001) and E/E' lateral (13.3 vs 9.4, p=0.0001)). Patients in control group had no change in NYHA class, LVD-36 scores, LV volumes or LVEF at 3 months although BNP and LV wall stress reduced to a slight extent. Patients on TMZ had further improvement in NYHA class, walk test, BNP levels and echocardiographic parameters at 6 months. CONCLUSIONS Metabolic modulators (TMZ) may help in improving LV function in DCM. In this study, benefit was noted by 3 months with further improvement at 6 months.
Collapse
Affiliation(s)
- Suman Jatain
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| | - Aditya Kapoor
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India.
| | - Archana Sinha
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| | - Roopali Khanna
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| | - Sudeep Kumar
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| | - Naveen Garg
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| | - Satyendra Tewari
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| | - Pravin Goel
- Department of Cardiology, Sanjay Gandhi PGIMS, Lucknow 226014, India
| |
Collapse
|
39
|
van Campen JSJA, de Boer K, van de Veerdonk MC, van der Bruggen CEE, Allaart CP, Raijmakers PG, Heymans MW, Marcus JT, Harms HJ, Handoko ML, de Man FS, Vonk Noordegraaf A, Bogaard HJ. Bisoprolol in idiopathic pulmonary arterial hypertension: an explorative study. Eur Respir J 2016; 48:787-96. [PMID: 27390285 DOI: 10.1183/13993003.00090-2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 05/21/2016] [Indexed: 01/08/2023]
Abstract
While beta-blockers are considered contraindicated in pulmonary arterial hypertension (PAH), the prognostic significance of sympathetic nervous system over-activity suggests a potential benefit of beta-blocker therapy. The aim of this randomised, placebo-controlled, crossover, single centre study was to determine the effects of bisoprolol on right ventricular ejection fraction (RVEF) in idiopathic PAH (iPAH) patients. Additional efficacy and safety parameters were explored.Patients with optimally treated, stable iPAH (New York Heart Association functional class II/III) were randomised to placebo or bisoprolol. Imaging and functional measurements were performed at baseline, crossover and end of study.18 iPAH patients were included, because inclusion faltered before enrolment of the targeted 25 patients. 17 patients completed 6 months of bisoprolol, 15 tolerated bisoprolol, one patient required intravenous diuretics. Bisoprolol was associated with a lower heart rate (17 beats per minute, p=0.0001) but RVEF remained unchanged. A drop in cardiac index (0.5 L·min(-1)·m(-2), p=0.015) was observed, along with a trend towards a decreased 6-min walking distance (6MWD).Although careful up-titration of bisoprolol was tolerated by most patients and resulted in a decreased heart rate, no benefit of bisoprolol in iPAH was demonstrated. Decreases in cardiac index and 6MWD suggest a deteriorated cardiac function. The results do not favour the use of bisoprolol in iPAH patients.
Collapse
Affiliation(s)
- Jasmijn S J A van Campen
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands Both authors contributed equally
| | - Karin de Boer
- Dept of cardiology, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands Both authors contributed equally
| | - Mariëlle C van de Veerdonk
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands Dept of cardiology, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| | - Cathelijne E E van der Bruggen
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| | - Cor P Allaart
- Dept of cardiology, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| | - Pieter G Raijmakers
- Dept of nuclear medicine and PET-research, VU University medical center, Amsterdam, The Netherlands
| | - Martijn W Heymans
- Dept of epidemiology, VU University medical center, Amsterdam, The Netherlands
| | - J Tim Marcus
- Dept of physics and medical technology, VU University medical center, Amsterdam, The Netherlands
| | - Hendrik J Harms
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands Dept of nuclear medicine and PET-research, VU University medical center, Amsterdam, The Netherlands
| | - M Louis Handoko
- Dept of cardiology, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| | - Frances S de Man
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands Dept of physiology, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| | - Anton Vonk Noordegraaf
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| | - Harm-Jan Bogaard
- Dept of pulmonary medicine, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands Dept of physiology, Institute for cardiovascular research, VU University medical center, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Masuda A, Yoshinaga K, Naya M, Manabe O, Yamada S, Iwano H, Okada T, Katoh C, Takeishi Y, Tsutsui H, Tamaki N. Accelerated (99m)Tc-sestamibi clearance associated with mitochondrial dysfunction and regional left ventricular dysfunction in reperfused myocardium in patients with acute coronary syndrome. EJNMMI Res 2016; 6:41. [PMID: 27169534 PMCID: PMC4864798 DOI: 10.1186/s13550-016-0196-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/28/2016] [Indexed: 12/02/2022] Open
Abstract
Background Accelerated clearance of 99mtechnetium-sestamibi (MIBI) has been observed after reperfusion therapy in patients with acute coronary syndrome (ACS), but the mechanisms have not been fully investigated. MIBI retention may depend on mitochondrial function. The clearance rate of 11carbon-acetate reflects such mitochondrial functions as oxidative metabolism. The purpose of this study was to examine the mechanisms of accelerated MIBI clearance in ACS. We therefore compared it to oxidative metabolism estimated using 11C-acetate positron emission tomography (PET). Methods Eighteen patients [mean age 69.2 ± 8.7 years, 10 males (56 %)] with reperfused ACS underwent MIBI single-photon emission computed tomography (SPECT), echocardiography, and 11C-acetate PET within 3 weeks of the onset of ACS. MIBI images were obtained 30 min and 3 h after MIBI administration. Regional left ventricular (LV) function was evaluated by echocardiography. The measurement of oxidative metabolism was obtained through the mono-exponential fitting of the 11C-acetate time-activity curve (kmono). Results Among 95 segments of reperfused myocardium, MIBI SPECT showed 64 normal segments (group N), 14 segments with accelerated MIBI clearance (group AC), and 17 segments with fixed defect (group F). Group AC showed lower kmono than group N (0.041 ± 0.009 vs 0.049 ± 0.010, p = 0.02). Group F showed lower kmono than group N (0.039 ± 0.012 vs 0.049 ± 0.010, p = 0.01). However, kmono was similar in group AC and group F (p = 0.99). Conclusions Segments with accelerated MIBI clearance showed reduced oxidative metabolism in ACS. Loss of MIBI retention may be associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Atsuro Masuda
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan.,Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Keiichiro Yoshinaga
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan. .,Molecular Imaging Research Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-Ku, Chiba, 263-8555, Japan.
| | - Masanao Naya
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Osamu Manabe
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Yamada
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroyuki Iwano
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tatsuya Okada
- Department of Natural Sciences, Fukushima Medical University, Fukushima, Japan
| | - Chietsugu Katoh
- Department of Health Sciences, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
41
|
Nguyen PK, Wu JC. Large Animal Models of Ischemic Cardiomyopathy: Are They Enough to Bridge the Translational Gap? J Nucl Cardiol 2015; 22:666-72. [PMID: 25777782 DOI: 10.1007/s12350-015-0078-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Patricia K Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Grant Building S140, Stanford, CA, 94305-5111, USA,
| | | |
Collapse
|
42
|
Kundu BK, Zhong M, Sen S, Davogustto G, Keller SR, Taegtmeyer H. Remodeling of glucose metabolism precedes pressure overload-induced left ventricular hypertrophy: review of a hypothesis. Cardiology 2015; 130:211-20. [PMID: 25791172 DOI: 10.1159/000369782] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/10/2014] [Indexed: 12/14/2022]
Abstract
When subjected to pressure overload, the ventricular myocardium shifts from fatty acids to glucose as its main source for energy provision and frequently increases its mass. Here, we review the evidence in support of the concept that metabolic remodeling, measured as an increased myocardial glucose uptake using dynamic positron emission tomography (PET) with the glucose analogue 2-deoxy-2-[(18)F]fluoro-D-glucose (FDG), precedes the onset of left ventricular hypertrophy (LVH) and heart failure. Consistent with this, early intervention with propranolol, which attenuates glucose uptake, prevents the maladaptive metabolic response and preserves cardiac function in vivo. We also review ex vivo studies suggesting a link between dysregulated myocardial glucose metabolism, intracellular accumulation of glucose 6-phosphate (G6P) and contractile dysfunction of the heart. G6P levels correlate with activation of mTOR (mechanistic target of rapamycin) and endoplasmic reticulum stress. This sequence of events could be prevented by pretreatment with rapamycin (mTOR inhibition) or metformin (enzyme 5'-AMP-activated protein kinase activation). In conclusion, we propose that metabolic imaging with FDG PET may provide a novel approach to guide the treatment of patients with hypertension-induced LVH.
Collapse
Affiliation(s)
- Bijoy K Kundu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Va., USA
| | | | | | | | | | | |
Collapse
|
43
|
Ahmadi A, Ohira H, Mielniczuk LM. FDG PET Imaging for Identifying Pulmonary Hypertension and Right Heart Failure. Curr Cardiol Rep 2014; 17:555. [DOI: 10.1007/s11886-014-0555-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
44
|
Treatment with inotropes and related prognosis in acute heart failure: Contemporary data from the Italian Network on Heart Failure (IN-HF) Outcome registry. J Heart Lung Transplant 2014; 33:1056-65. [DOI: 10.1016/j.healun.2014.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 05/13/2014] [Accepted: 05/28/2014] [Indexed: 01/11/2023] Open
|
45
|
Bell SP, Adkisson DW, Lawson MA, Wang L, Ooi H, Sawyer DB, Kronenberg MW. Antifailure therapy including spironolactone improves left ventricular energy supply-demand relations in nonischemic dilated cardiomyopathy. J Am Heart Assoc 2014; 3:e000883. [PMID: 25164945 PMCID: PMC4310370 DOI: 10.1161/jaha.114.000883] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 08/04/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Left ventricular (LV) energy supply-demand imbalance is postulated to cause "energy starvation" and contribute to heart failure (HF) in nonischemic dilated cardiomyopathy (NIDCM). Using cardiac magnetic resonance (CMR) and [(11)C] acetate positron emission tomography (PET), we evaluated LV perfusion and oxidative metabolism in NIDCM and the effects of spironolactone on LV supply-demand relations. METHODS AND RESULTS Twelve patients with NIDCM underwent CMR and PET at baseline and after ≥6 months of spironolactone therapy added to a standard HF regimen. The myocardial perfusion reserve index (MPRI) was calculated after gadolinium injection during adenosine, as compared to rest. The monoexponential clearance rate of [(11)C] acetate (kmono) was used to calculate the work metabolic index (WMI), an index of LV mechanical efficiency, and kmono/RPP (rate-pressure product), an index of energy supply/demand. At baseline, the subendocardium was hypoperfused versus the subepicardium (median MPRI, 1.63 vs. 1.80; P<0.001), but improved to 1.80 (P<0.001) after spironolactone. The WMI increased (P=0.001), as did kmono/RPP (P=0.003). These improvements were associated with reverse remodeling, increased LV ejection fraction, and decreases in LV mass and systolic wall stress (all P<0.002). CONCLUSIONS NIDCM is associated with subendocardial hypoperfusion and impaired myocardial oxidative metabolism, consistent with energy starvation. Antifailure therapy improves parameters of energy starvation and is associated with augmented LV performance. CLINICAL TRIAL REGISTRATION URL http://www.clinicaltrials.gov/ Unique identifier: ID NCT00574119.
Collapse
Affiliation(s)
- Susan P. Bell
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN (S.P.B., D.W.A., M.A.L., H.O., D.B.S., M.W.K.)
| | - Douglas W. Adkisson
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN (S.P.B., D.W.A., M.A.L., H.O., D.B.S., M.W.K.)
| | - Mark A. Lawson
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN (S.P.B., D.W.A., M.A.L., H.O., D.B.S., M.W.K.)
| | - Li Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN (L.W.)
| | - Henry Ooi
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN (S.P.B., D.W.A., M.A.L., H.O., D.B.S., M.W.K.)
- Cardiology Section, VA Tennessee Valley Healthcare System, Nashville, TN (H.O., M.W.K.)
| | - Douglas B. Sawyer
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN (S.P.B., D.W.A., M.A.L., H.O., D.B.S., M.W.K.)
| | - Marvin W. Kronenberg
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN (S.P.B., D.W.A., M.A.L., H.O., D.B.S., M.W.K.)
- Cardiology Section, VA Tennessee Valley Healthcare System, Nashville, TN (H.O., M.W.K.)
| |
Collapse
|
46
|
Kadkhodayan A, Coggan AR, Peterson LR. A "PET" area of interest: myocardial metabolism in human systolic heart failure. Heart Fail Rev 2014. [PMID: 23180281 DOI: 10.1007/s10741-012-9360-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Myocardial substrate metabolism provides the energy needed for cardiac contraction and relaxation. The normal adult heart uses predominantly fatty acids (FAs) as its primary fuel source. However, the heart can switch and use glucose (and to a lesser extent, ketones, lactate, as well as endogenous triglycerides and glycogen), depending on the metabolic milieu and superimposed conditions. FAs are not a wholly better fuel than glucose, but they do provide more energy per mole than glucose. Conversely, glucose is the more oxygen-efficient fuel. Studies in animal models of heart failure (HF) fairly consistently demonstrate a shift away from myocardial fatty acid metabolism and toward glucose metabolism. Studies in humans are less consistent. Some show the same metabolic switch away from FA metabolism but not all. This may be due to differences in the etiology of HF, sex-related differences, or other mitigating factors. For example, obesity, insulin resistance, and diabetes are all related to an increased risk of HF and may complicate or contribute to its development. However, these conditions are associated with increased FA metabolism. This review will discuss aspects of human heart metabolism in systolic dysfunction as measured by the noninvasive, quantitative method-positron emission tomography. Continued research in this area is vital if we are to ameliorate HF by manipulating heart metabolism with the aim of increasing energy production and/or efficiency.
Collapse
Affiliation(s)
- Ana Kadkhodayan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
47
|
Yoshinaga K, Ohira H, Tsujino I, Oyama-Manabe N, Mielniczuk L, Beanlands RSB, Katoh C, Kasai K, Manabe O, Sato T, Fujii S, Ito YM, Tomiyama Y, Nishimura M, Tamaki N. Attenuated right ventricular energetics evaluated using ¹¹C-acetate PET in patients with pulmonary hypertension. Eur J Nucl Med Mol Imaging 2014; 41:1240-50. [PMID: 24615469 DOI: 10.1007/s00259-014-2736-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/17/2014] [Indexed: 12/18/2022]
Abstract
PURPOSE The right ventricle (RV) has a high capacity to adapt to pressure or volume overload before failing. However, the mechanisms of RV adaptation, in particular RV energetics, in patients with pulmonary hypertension (PH) are still not well understood. We aimed to evaluate RV energetics including RV oxidative metabolism, power and efficiency to adapt to increasing pressure overload in patients with PH using (11)C-acetate PET. METHODS In this prospective study, 27 patients with WHO functional class II/III PH (mean pulmonary arterial pressure 39.8 ± 13.5 mmHg) and 9 healthy individuals underwent (11)C-acetate PET. (11)C-acetate PET was used to simultaneously measure oxidative metabolism (k mono) for the left ventricle (LV) and RV. LV and RV efficiency were also calculated. RESULTS The RV ejection fraction in PH patients was lower than in controls (p = 0.0054). There was no statistically significant difference in LV k mono (p = 0.09). In contrast, PH patients showed higher RV k mono than did controls (0.050 ± 0.009 min(-1) vs. 0.030 ± 0.006 min(-1), p < 0.0001). PH patients exhibited significantly increased RV power (p < 0.001) and hence increased RV efficiency compared to controls (0.40 ± 0.14 vs. 0.017 ± 0.12 mmHg·mL·min/g, p = 0.001). CONCLUSION The RV oxidative metabolic rate was increased in patients with PH. Patients with WHO functional class II/III PH also had increased RV power and efficiency. These findings may indicate a myocardial energetics adaptation response to increasing pulmonary arterial pressure.
Collapse
Affiliation(s)
- Keiichiro Yoshinaga
- Department of Molecular Imaging, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-Ku, Sapporo, Hokkaido, Japan, 060-8638,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Takada T, Sakata Y, Miyata S, Takahashi J, Nochioka K, Miura M, Tadaki S, Shimokawa H. Impact of elevated heart rate on clinical outcomes in patients with heart failure with reduced and preserved ejection fraction: a report from the CHART-2 Study. Eur J Heart Fail 2013; 16:309-16. [DOI: 10.1002/ejhf.22] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/13/2013] [Accepted: 10/18/2013] [Indexed: 01/13/2023] Open
Affiliation(s)
- Tsuyoshi Takada
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Yasuhiko Sakata
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Satoshi Miyata
- Department of Evidenced-based Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Kotaro Nochioka
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Masanobu Miura
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Soichiro Tadaki
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
- Department of Evidenced-based Cardiovascular Medicine; Tohoku University Graduate School of Medicine; Sendai 980-8574 Japan
| | | |
Collapse
|
49
|
Abstract
Abnormalities in myocardial substrate metabolism play a central role in the manifestations of most forms of cardiac disease such as ischemic heart disease, heart failure, hypertensive heart disease, and the cardiomyopathy due to either obesity or diabetes mellitus. Their importance is exemplified by both the development of numerous imaging tools designed to detect the specific metabolic perturbations or signatures related to these different diseases, and the vigorous efforts in drug discovery/development targeting various aspects of myocardial metabolism. Since the prior review in 2005, we have gained new insights into how perturbations in myocardial metabolism contribute to various forms of cardiac disease. For example, the application of advanced molecular biologic techniques and the development of elegant genetic models have highlighted the pleiotropic actions of cellular metabolism on energy transfer, signal transduction, cardiac growth, gene expression, and viability. In parallel, there have been significant advances in instrumentation, radiopharmaceutical design, and small animal imaging, which now permit a near completion of the translational pathway linking in-vitro measurements of metabolism with the human condition. In this review, most of the key advances in metabolic imaging will be described, their contribution to cardiovascular research highlighted, and potential new clinical applications proposed.
Collapse
Affiliation(s)
- Robert J Gropler
- Division of Radiological Sciences, Cardiovascular Imaging Laboratory, Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway, St. Louis, MO, 63110, USA,
| |
Collapse
|
50
|
Bayeva M, Sawicki KT, Ardehali H. Taking diabetes to heart--deregulation of myocardial lipid metabolism in diabetic cardiomyopathy. J Am Heart Assoc 2013; 2:e000433. [PMID: 24275630 PMCID: PMC3886738 DOI: 10.1161/jaha.113.000433] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Marina Bayeva
- Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL
| | | | | |
Collapse
|