1
|
Karkouri J, Watson W, Forner R, Weir-McCall JR, Horn T, Hill M, Hoole S, Klomp D, Rodgers CT. Regionally resolved cardiac metabolism using a dipole-loop array coil for 7 T 31P-MRSI. Magn Reson Med 2025. [PMID: 40123193 DOI: 10.1002/mrm.30492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE We introduce a novel commercial phosphorus-31 (31P) dipole-loop array coil, describing the coil hardware and testing its performance on phantoms. We used this coil to assess cardiac metabolism per region in healthy volunteers. METHODS B1 + field maps were simulated and compared to maps measured with a set of CSI sequences with varying voltages. Seventeen volunteers were scanned with 7 T phosphorus-31 magnetic resonance spectroscopic imaging (31P-MRSI). Reproducibility was assessed in nine of these volunteers. Strain was measured for six of these volunteers at 3 T. RESULTS Blood- and saturation-corrected Phosphocreatine/γ-adenosine triphosphate (PCr/ATP) ratios were measured for four regions of the left ventricle: 1.86 in septum, 2.25 in anterior wall, 1.41 in inferior wall, and 1.53 in lateral wall, respectively. These are in the expected range compared to previous studies. B1 + maps show good signal uniformity around the position of the heart (0.13 ± 0.06 μT/sqrt(W)). Intrasession and intersession coefficients of reproducibility were 0.22-0.88 and 0.29-0.79, respectively. Linear modeling shows that regional PCr/γATP correlates with circumferential strain but not radial strain. This requires corroboration by a larger study including patients with impaired function and energetics. CONCLUSION Dipole-loop array coils present a promising new approach for human cardiac 31P-MRSI at 7 T. Their favorable B1 + uniformity at depth and specific absorption rate over loop arrays and improved SNR when combined with loops for reception could be beneficial for further clinical studies measuring energetics by 31P-MRSI at 7 T. The new capability to assess PCr/γATP ratios across the whole left ventricle could enable clinical studies to investigate regional changes in cardiac energetics for the first time.
Collapse
Affiliation(s)
- Jabrane Karkouri
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Will Watson
- Department of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | | | - Jonathan R Weir-McCall
- Department of Radiology, University of Cambridge, Cambridge, UK
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| | - Tracy Horn
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Marion Hill
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Stephen Hoole
- Department of Radiology, University of Cambridge, Cambridge, UK
- Royal Papworth Hospital, Cambridge, UK
| | | | | |
Collapse
|
2
|
Rossi M, Fabris E, Sinagra G. Aficamten in non-obstructive hypertrophic cardiomyopathy: Unlocking new horizons? Eur J Heart Fail 2025; 27:456-458. [PMID: 39291300 DOI: 10.1002/ejhf.3461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Affiliation(s)
- Maddalena Rossi
- Cardiothoracovascular Department, Center for Diagnosis and Treatment of Cardiomyopathies, Azienda Sanitaria Universitaria Giuliano-Isontina, University of Trieste, Trieste, Italy
- European Reference Network for Rare, Low-Prevalence, and Complex Diseases of the Heart (ERN GUARD-Heart)
| | - Enrico Fabris
- Cardiothoracovascular Department, Center for Diagnosis and Treatment of Cardiomyopathies, Azienda Sanitaria Universitaria Giuliano-Isontina, University of Trieste, Trieste, Italy
- European Reference Network for Rare, Low-Prevalence, and Complex Diseases of the Heart (ERN GUARD-Heart)
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Center for Diagnosis and Treatment of Cardiomyopathies, Azienda Sanitaria Universitaria Giuliano-Isontina, University of Trieste, Trieste, Italy
- European Reference Network for Rare, Low-Prevalence, and Complex Diseases of the Heart (ERN GUARD-Heart)
| |
Collapse
|
3
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Maron MS, Mahmod M, Abd Samat AH, Choudhury L, Massera D, Phelan DMJ, Cresci S, Martinez MW, Masri A, Abraham TP, Adler E, Wever-Pinzon O, Nagueh SF, Lewis GD, Chamberlin P, Patel J, Yavari A, Dehbi HM, Sarwar R, Raman B, Valkovič L, Neubauer S, Udelson JE, Watkins H. Safety and Efficacy of Metabolic Modulation With Ninerafaxstat in Patients With Nonobstructive Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2024; 83:2037-2048. [PMID: 38599256 DOI: 10.1016/j.jacc.2024.03.387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND In nonobstructive hypertrophic cardiomyopathy (nHCM), there are no approved medical therapies. Impaired myocardial energetics is a potential cause of symptoms and exercise limitation. Ninerafaxstat, a novel cardiac mitotrope, enhances cardiac energetics. OBJECTIVES This study sought to evaluate the safety and efficacy of ninerafaxstat in nHCM. METHODS Patients with hypertrophic cardiomyopathy and left ventricular outflow tract gradient <30 mm Hg, ejection fraction ≥50%, and peak oxygen consumption <80% predicted were randomized to ninerafaxstat 200 mg twice daily or placebo (1:1) for 12 weeks. The primary endpoint was safety and tolerability, with efficacy outcomes also assessed as secondary endpoints. RESULTS A total of 67 patients with nHCM were enrolled at 12 centers (57 ± 11.8 years of age; 55% women). Serious adverse events occurred in 11.8% (n = 4 of 34) in the ninerafaxstat group and 6.1% (n = 2 of 33) of patients in the placebo group. From baseline to 12 weeks, ninerafaxstat was associated with significantly better VE/Vco2 (ventilatory efficiency) slope compared with placebo with a least-squares (LS) mean difference between the groups of -2.1 (95% CI: -3.6 to -0.6; P = 0.006), with no significant difference in peak VO2 (P = 0.90). The Kansas City Cardiomyopathy Questionnaire Clinical Summary Score was directionally, though not significantly, improved with ninerafaxstat vs placebo (LS mean 3.2; 95% CI: -2.9 to 9.2; P = 0.30); however, it was statistically significant when analyzed post hoc in the 35 patients with baseline Kansas City Cardiomyopathy Questionnaire Clinical Summary Score ≤80 (LS mean 9.4; 95% CI: 0.3-18.5; P = 0.04). CONCLUSIONS In symptomatic nHCM, novel drug therapy targeting myocardial energetics was safe and well tolerated and associated with better exercise performance and health status among those most symptomatically limited. The findings support assessing ninerafaxstat in a phase 3 study.
Collapse
Affiliation(s)
- Martin S Maron
- Hypertrophic Cardiomyopathy Center, Lahey Hospital and Medical Center, Burlington, Massachusetts, USA.
| | - Masliza Mahmod
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Azlan Helmy Abd Samat
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Lubna Choudhury
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniele Massera
- Hypertrophic Cardiomyopathy Program, Leon H. Charney Division of Cardiology, NYU Langone Health, New York, New York, USA
| | - Dermot M J Phelan
- Sanger Heart and Vascular Institute, Atrium Health, Charlotte, North Carolina, USA
| | - Sharon Cresci
- Center for Cardiovascular Research, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Matthew W Martinez
- Division of Cardiology, Atlantic Health System, Morristown, New Jersey, USA
| | - Ahmad Masri
- Hypertrophic Cardiomyopathy Center, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Theodore P Abraham
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA
| | - Eric Adler
- Division of Cardiovascular Medicine, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Omar Wever-Pinzon
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Sherif F Nagueh
- Methodist DeBakey Heart and Vascular Center, Houston, Texas, USA
| | - Gregory D Lewis
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Jai Patel
- Imbria Pharmaceuticals, Boston, Massachusetts, USA
| | - Arash Yavari
- Imbria Pharmaceuticals, Boston, Massachusetts, USA
| | | | | | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - James E Udelson
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
Borrelli F, Lombardi R, Canciello G, Frisso G, Todde G, Esposito G, Losi MA. Mechano-energetic efficiency in patients with hypertrophic cardiomyopathy with and without sarcomeric mutations. J Cardiovasc Transl Res 2024; 17:458-466. [PMID: 37833437 DOI: 10.1007/s12265-023-10441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is mainly caused by sarcomeric mutations which may affect myocardial mechano-energetic efficiency (MEE). We investigated the effects of sarcomeric mutations on MEE. A non-invasive pressure/volume (P/V) analysis was performed. We included 49 genetically screened HCM patients. MEEi was calculated as the ratio between stroke volume and heart rate normalized by LV mass. Fifty-seven percent (57%) HCM patients carried a sarcomeric mutation. Patients with and without sarcomeric mutations had similar LV ejection fraction, heart rate, LV mass, and LV outflow gradient. Younger age at diagnosis, family history of HCM, and lower MEEi were associated with presence of sarcomeric mutation (p = 0.017; p = 0.001 and p = 0.0001, respectively). Lower MEEi in HCM with sarcomeric mutation is not related to significant differences on filling pressure as shown on P/V analysis. Sarcomeric mutations determine a reduction of the LV pump performance as estimated by MEEi in HCM. Lower MEEi may predict a positive genetic analysis.
Collapse
Affiliation(s)
- Felice Borrelli
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy
| | - Raffaella Lombardi
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy
| | - Grazia Canciello
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy
| | - Gaetano Todde
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy
| | - Maria-Angela Losi
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via S Pansini, 5, I-801313, Naples, Italy.
| |
Collapse
|
6
|
Coleman JA, Ashkir Z, Raman B, Bueno-Orovio A. Mechanisms and prognostic impact of myocardial ischaemia in hypertrophic cardiomyopathy. Int J Cardiovasc Imaging 2023; 39:1979-1996. [PMID: 37358707 PMCID: PMC10589194 DOI: 10.1007/s10554-023-02894-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/03/2023] [Indexed: 06/27/2023]
Abstract
Despite the progress made in risk stratification, sudden cardiac death and heart failure remain dreaded complications for hypertrophic cardiomyopathy (HCM) patients. Myocardial ischaemia is widely acknowledged as a contributor to cardiovascular events, but the assessment of ischaemia is not yet included in HCM clinical guidelines. This review aims to evaluate the HCM-specific pro-ischaemic mechanisms and the potential prognostic value of imaging for myocardial ischaemia in HCM. A literature review was performed using PubMed to identify studies with non-invasive imaging of ischaemia (cardiovascular magnetic resonance, echocardiography, and nuclear imaging) in HCM, prioritising studies published after the last major review in 2009. Other studies, including invasive ischaemia assessment and post-mortem histology, were also considered for mechanistic or prognostic relevance. Pro-ischaemic mechanisms in HCM reviewed included the effects of sarcomeric mutations, microvascular remodelling, hypertrophy, extravascular compressive forces and left ventricular outflow tract obstruction. The relationship between ischaemia and fibrosis was re-appraised by considering segment-wise analyses in multimodal imaging studies. The prognostic significance of myocardial ischaemia in HCM was evaluated using longitudinal studies with composite endpoints, and reports of ischaemia-arrhythmia associations were further considered. The high prevalence of ischaemia in HCM is explained by several micro- and macrostructural pathological features, alongside mutation-associated energetic impairment. Ischaemia on imaging identifies a subgroup of HCM patients at higher risk of adverse cardiovascular outcomes. Ischaemic HCM phenotypes are a high-risk subgroup associated with more advanced left ventricular remodelling, but further studies are required to evaluate the independent prognostic value of non-invasive imaging for ischaemia.
Collapse
Affiliation(s)
- James A Coleman
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Zakariye Ashkir
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | |
Collapse
|
7
|
Moore J, Ewoldt J, Venturini G, Pereira AC, Padilha K, Lawton M, Lin W, Goel R, Luptak I, Perissi V, Seidman CE, Seidman J, Chin MT, Chen C, Emili A. Multi-Omics Profiling of Hypertrophic Cardiomyopathy Reveals Altered Mechanisms in Mitochondrial Dynamics and Excitation-Contraction Coupling. Int J Mol Sci 2023; 24:4724. [PMID: 36902152 PMCID: PMC10002553 DOI: 10.3390/ijms24054724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Hypertrophic cardiomyopathy is one of the most common inherited cardiomyopathies and a leading cause of sudden cardiac death in young adults. Despite profound insights into the genetics, there is imperfect correlation between mutation and clinical prognosis, suggesting complex molecular cascades driving pathogenesis. To investigate this, we performed an integrated quantitative multi-omics (proteomic, phosphoproteomic, and metabolomic) analysis to illuminate the early and direct consequences of mutations in myosin heavy chain in engineered human induced pluripotent stem-cell-derived cardiomyocytes relative to late-stage disease using patient myectomies. We captured hundreds of differential features, which map to distinct molecular mechanisms modulating mitochondrial homeostasis at the earliest stages of pathobiology, as well as stage-specific metabolic and excitation-coupling maladaptation. Collectively, this study fills in gaps from previous studies by expanding knowledge of the initial responses to mutations that protect cells against the early stress prior to contractile dysfunction and overt disease.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jourdan Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
| | | | | | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Sao Paulo 05508-000, Brazil
| | - Matthew Lawton
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Weiwei Lin
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Raghuveera Goel
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ivan Luptak
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA 02118, USA
| | - Valentina Perissi
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02145, USA
| | - Christopher Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andrew Emili
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
8
|
Tsampasian V, Cameron D, Sobhan R, Bazoukis G, Vassiliou VS. Phosphorus Magnetic Resonance Spectroscopy ( 31P MRS) and Cardiovascular Disease: The Importance of Energy. Medicina (B Aires) 2023; 59:medicina59010174. [PMID: 36676798 PMCID: PMC9866867 DOI: 10.3390/medicina59010174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Background and Objectives: The heart is the organ with the highest metabolic demand in the body, and it relies on high ATP turnover and efficient energy substrate utilisation in order to function normally. The derangement of myocardial energetics may lead to abnormalities in cardiac metabolism, which herald the symptoms of heart failure (HF). In addition, phosphorus magnetic resonance spectroscopy (31P MRS) is the only available non-invasive method that allows clinicians and researchers to evaluate the myocardial metabolic state in vivo. This review summarises the importance of myocardial energetics and provides a systematic review of all the available research studies utilising 31P MRS to evaluate patients with a range of cardiac pathologies. Materials and Methods: We have performed a systematic review of all available studies that used 31P MRS for the investigation of myocardial energetics in cardiovascular disease. Results: A systematic search of the Medline database, the Cochrane library, and Web of Science yielded 1092 results, out of which 62 studies were included in the systematic review. The 31P MRS has been used in numerous studies and has demonstrated that impaired myocardial energetics is often the beginning of pathological processes in several cardiac pathologies. Conclusions: The 31P MRS has become a valuable tool in the understanding of myocardial metabolic changes and their impact on the diagnosis, risk stratification, and prognosis of patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Vasiliki Tsampasian
- Norwich Medical School, University of East Anglia, Bob Champion Research & Education Building, Research Park, Rosalind Franklin Rd, Norwich NR4 7UQ, UK
- Correspondence: (V.T.); (V.S.V.)
| | - Donnie Cameron
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Rashed Sobhan
- Norwich Medical School, University of East Anglia, Bob Champion Research & Education Building, Research Park, Rosalind Franklin Rd, Norwich NR4 7UQ, UK
| | - George Bazoukis
- Department of Cardiology, Larnaca General Hospital, Larnaca 6301, Cyprus
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia 2417, Cyprus
| | - Vassilios S. Vassiliou
- Norwich Medical School, University of East Anglia, Bob Champion Research & Education Building, Research Park, Rosalind Franklin Rd, Norwich NR4 7UQ, UK
- Correspondence: (V.T.); (V.S.V.)
| |
Collapse
|
9
|
Maron BA, Wang RS, Carnethon MR, Rowin EJ, Loscalzo J, Maron BJ, Maron MS. What Causes Hypertrophic Cardiomyopathy? Am J Cardiol 2022; 179:74-82. [PMID: 35843734 DOI: 10.1016/j.amjcard.2022.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 01/11/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a global and relatively common cause of patient morbidity and mortality and is among the first reported monogenic cardiac diseases. For 30 years, the basic etiology of HCM has been attributed largely to variants in individual genes encoding cardiac sarcomere proteins, with the implication that HCM is fundamentally a genetic disease. However, data from clinical and network medicine analyses, as well as contemporary genetic studies show that single gene variants do not fully explain the broad and diverse HCM clinical spectrum. These transformative advances place a new focus on possible novel interactions between acquired disease determinants and genetic context to produce complex HCM phenotypes, also offering a measure of caution against overemphasizing monogenics as the principal cause of this disease. These new perspectives in which HCM is not a uniformly genetic disease but likely explained by multifactorial etiology will also unavoidably impact how HCM is viewed by patients and families in the clinical practicing community going forward, including relevance to genetic counseling and access to healthcare insurance and psychosocial wellness.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine and Harvard Medical School, Boston, Massachusetts.
| | - Rui-Sheng Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mercedes R Carnethon
- Division of Pulmonology and Critical Care, Feinberg School of Medicine, Chicago, Illinois
| | - Ethan J Rowin
- HCM Center, Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine and Harvard Medical School, Boston, Massachusetts
| | - Barry J Maron
- HCM Center, Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Martin S Maron
- HCM Center, Lahey Hospital and Medical Center, Burlington, Massachusetts
| |
Collapse
|
10
|
Increased cardiac Pi/PCr in the diabetic heart observed using phosphorus magnetic resonance spectroscopy at 7T. PLoS One 2022; 17:e0269957. [PMID: 35709167 PMCID: PMC9202907 DOI: 10.1371/journal.pone.0269957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
Phosphorus magnetic resonance spectroscopy (31P-MRS) has previously demonstrated decreased energy reserves in the form of phosphocreatine to adenosine-tri-phosphate ratio (PCr/ATP) in the hearts of patients with type 2 diabetes (T2DM). Recent 31P-MRS techniques using 7T systems, e.g. long mixing time stimulated echo acquisition mode (STEAM), allow deeper insight into cardiac metabolism through assessment of inorganic phosphate (Pi) content and myocardial pH, which play pivotal roles in energy production in the heart. Therefore, we aimed to further explore the cardiac metabolic phenotype in T2DM using STEAM at 7T. Seventeen patients with T2DM and twenty-three healthy controls were recruited and their cardiac PCr/ATP, Pi/PCr and pH were assessed at 7T. Diastolic function of all patients with T2DM was assessed using echocardiography to investigate the relationship between diastolic dysfunction and cardiac metabolism. Mirroring the decreased PCr/ATP (1.70±0.31 vs. 2.07±0.39; p<0.01), the cardiac Pi/PCr was increased (0.13±0.07 vs. 0.10±0.03; p = 0.02) in T2DM patients in comparison to healthy controls. Myocardial pH was not significantly different between the groups (7.14±0.12 vs. 7.10±0.12; p = 0.31). There was a negative correlation between PCr/ATP and diastolic function (R2 = 0.33; p = 0.02) in T2DM. No correlation was observed between diastolic function and Pi/PCr and (R2 = 0.16; p = 0.21). In addition, we did not observe any correlation between cardiac PCr/ATP and Pi/PCr (p = 0.19). Using STEAM 31P-MRS at 7T we have for the first time explored Pi/PCr in the diabetic human heart and found it increased when compared to healthy controls. The lack of correlation between measured PCr/ATP and Pi/PCr suggests that independent mechanisms might contribute to these perturbations.
Collapse
|
11
|
Ibrahim ESH, Dennison J, Frank L, Stojanovska J. Diastolic Cardiac Function by MRI-Imaging Capabilities and Clinical Applications. Tomography 2021; 7:893-914. [PMID: 34941647 PMCID: PMC8706325 DOI: 10.3390/tomography7040075] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 02/05/2023] Open
Abstract
Most cardiac studies focus on evaluating left ventricular (LV) systolic function. However, the assessment of diastolic cardiac function is becoming more appreciated, especially with the increasing prevalence of pathologies associated with diastolic dysfunction like heart failure with preserved ejection fraction (HFpEF). Diastolic dysfunction is an indication of abnormal mechanical properties of the myocardium, characterized by slow or delayed myocardial relaxation, abnormal LV distensibility, and/or impaired LV filling. Diastolic dysfunction has been shown to be associated with age and other cardiovascular risk factors such as hypertension and diabetes mellitus. In this context, cardiac magnetic resonance imaging (MRI) has the capability for differentiating between normal and abnormal myocardial relaxation patterns, and therefore offers the prospect of early detection of diastolic dysfunction. Although diastolic cardiac function can be assessed from the ratio between early and atrial filling peaks (E/A ratio), measuring different parameters of heart contractility during diastole allows for evaluating spatial and temporal patterns of cardiac function with the potential for illustrating subtle changes related to age, gender, or other differences among different patient populations. In this article, we review different MRI techniques for evaluating diastolic function along with clinical applications and findings in different heart diseases.
Collapse
Affiliation(s)
- El-Sayed H. Ibrahim
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Correspondence:
| | - Jennifer Dennison
- Department of Medicine, Medical College of Wisconsin, Wausau, WI 54401, USA;
| | - Luba Frank
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | | |
Collapse
|
12
|
Zampieri M, Berteotti M, Ferrantini C, Tassetti L, Gabriele M, Tomberli B, Castelli G, Cappelli F, Stefàno P, Marchionni N, Coppini R, Olivotto I. Pathophysiology and Treatment of Hypertrophic Cardiomyopathy: New Perspectives. Curr Heart Fail Rep 2021; 18:169-179. [PMID: 34148184 DOI: 10.1007/s11897-021-00523-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW We provide a state of the art of therapeutic options in hypertrophic cardiomyopathy (HCM), focusing on recent advances in our understanding of the pathophysiology of sarcomeric disease. RECENT FINDINGS A wealth of novel information regarding the molecular mechanisms associated with the clinical phenotype and natural history of HCM have been developed over the last two decades. Such advances have only recently led to a number of controlled randomized studies, often limited in size and fortune. Recently, however, the allosteric inhibitors of cardiac myosin adenosine triphosphatase, countering the main pathophysiological abnormality associated with HCM-causing mutations, i.e. hypercontractility, have opened new management perspectives. Mavacamten is the first drug specifically developed for HCM used in a successful phase 3 trial, with the promise to reach symptomatic obstructive patients in the near future. In addition, the fine characterization of cardiomyocyte electrophysiological remodelling has recently highlighted relevant therapeutic targets. Current therapies for HCM focus on late disease manifestations without addressing the intrinsic pathological mechanisms. However, novel evidence-based approaches have opened the way for agents targeting HCM molecular substrates. The impact of these targeted interventions will hopefully alter the natural history of the disease in the near future.
Collapse
Affiliation(s)
- Mattia Zampieri
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy.
| | - Martina Berteotti
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luigi Tassetti
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Martina Gabriele
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Benedetta Tomberli
- Division of Interventional Structural Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - Gabriele Castelli
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Francesco Cappelli
- Division of Interventional Structural Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - Pierluigi Stefàno
- Division of Cardiac Surgery, Careggi University Hospital, Florence, Italy
| | - Niccolò Marchionni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Division of General Cardiology, Careggi University Hospital, Florence, Italy
| | | | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
13
|
Dynamic 31P-MRI and 31P-MRS of lower leg muscles in heart failure patients. Sci Rep 2021; 11:7412. [PMID: 33795721 PMCID: PMC8016929 DOI: 10.1038/s41598-021-86392-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/15/2021] [Indexed: 11/29/2022] Open
Abstract
Impaired oxidative metabolism is one of multi-variate factors leading to exercise intolerance in heart failure patients. The purpose of the study was to demonstrate the use of dynamic 31P magnetic resonance spectroscopy (MRS) and 31P magnetic resonance imaging (MRI) techniques to measure PCr resynthesis rate post-exercise as a biomarker for oxidative metabolism in skeletal muscle in HF patients and controls. In this prospective imaging study, we recruited six HF patients and five healthy controls. The imaging protocol included 31P-MRS, spectrally selective 3D turbo spin echo for 31P-MRI, and Dixon multi-echo GRE for fat–water imaging on a 3 T clinical MRI scanner. All the subjects were scanned pre-exercise, during plantar flexion exercise, and post-exercise recovery, with two rounds of exercise for 31P -MRS and 31P-MRI, respectively. Unpaired t-tests were used to compare 31P-MRS and 31P-MRI results between the HF and control cohorts. The results show that PCr resynthesis rate was significantly slower in the HF cohort compared to the controls using 31P-MRS (P = 0.0003) and 31P-MRI (P = 0.0014). 31P-MRI showed significant differences between the cohorts in muscle groups (soleus (P = 0.0018), gastrocnemius lateral (P = 0.0007) and gastrocnemius medial (P = 0.0054)). The results from this study suggest that 31P-MRS/31P-MRI may be used to quantify lower leg muscle oxidative metabolism in HF patients, with 31P-MRI giving an additional advantage of allowing further localization of oxidative metabolism deficits. Upon further validation, these techniques may serve as a potentially useful clinical imaging biomarker for staging and monitoring therapies in HF-patients.
Collapse
|
14
|
Fernandez-Caggiano M, Kamynina A, Francois AA, Prysyazhna O, Eykyn TR, Krasemann S, Crespo-Leiro MG, Vieites MG, Bianchi K, Morales V, Domenech N, Eaton P. Mitochondrial pyruvate carrier abundance mediates pathological cardiac hypertrophy. Nat Metab 2020; 2:1223-1231. [PMID: 33106688 PMCID: PMC7610404 DOI: 10.1038/s42255-020-00276-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Cardiomyocytes rely on metabolic substrates, not only to fuel cardiac output, but also for growth and remodelling during stress. Here we show that mitochondrial pyruvate carrier (MPC) abundance mediates pathological cardiac hypertrophy. MPC abundance was reduced in failing hypertrophic human hearts, as well as in the myocardium of mice induced to fail by angiotensin II or through transverse aortic constriction. Constitutive knockout of cardiomyocyte MPC1/2 in mice resulted in cardiac hypertrophy and reduced survival, while tamoxifen-induced cardiomyocyte-specific reduction of MPC1/2 to the attenuated levels observed during pressure overload was sufficient to induce hypertrophy with impaired cardiac function. Failing hearts from cardiomyocyte-restricted knockout mice displayed increased abundance of anabolic metabolites, including amino acids and pentose phosphate pathway intermediates and reducing cofactors. These hearts showed a concomitant decrease in carbon flux into mitochondrial tricarboxylic acid cycle intermediates, as corroborated by complementary 1,2-[13C2]glucose tracer studies. In contrast, inducible cardiomyocyte overexpression of MPC1/2 resulted in increased tricarboxylic acid cycle intermediates, and sustained carrier expression during transverse aortic constriction protected against cardiac hypertrophy and failure. Collectively, our findings demonstrate that loss of the MPC1/2 causally mediates adverse cardiac remodelling.
Collapse
Affiliation(s)
- Mariana Fernandez-Caggiano
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Alisa Kamynina
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asvi A Francois
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Oleksandra Prysyazhna
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Thomas R Eykyn
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Susanne Krasemann
- University Medical Center Hamburg Eppendorf UKE, Institute for Neuropathology, Hamburg, Germany
| | - Maria G Crespo-Leiro
- Unidad de Cirugia Cardiaca y Trasplante, Servicio de Cardiología, Complejo Hospitalario Universitario de A Coruña (CHUAC), Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Maria Garcia Vieites
- Unidad de Cirugia Cardiaca y Trasplante, Servicio de Cardiología, Complejo Hospitalario Universitario de A Coruña (CHUAC), Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Katiuscia Bianchi
- Barts Cancer Institute, Queen Mary, John Vane Science Centre, University of London, London, UK
| | - Valle Morales
- Barts Cancer Institute, Queen Mary, John Vane Science Centre, University of London, London, UK
| | - Nieves Domenech
- Unidad de Cirugia Cardiaca y Trasplante, Servicio de Cardiología, Complejo Hospitalario Universitario de A Coruña (CHUAC), Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Philip Eaton
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
15
|
Valkovič L, Lau JYC, Abdesselam I, Rider OJ, Frollo I, Tyler DJ, Rodgers CT, Miller JJJ. Effects of contrast agents on relaxation properties of 31P metabolites. Magn Reson Med 2020; 85:1805-1813. [PMID: 33090502 DOI: 10.1002/mrm.28541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/22/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022]
Abstract
PURPOSE Phosphorous MR spectroscopy (31P-MRS) forms a powerful, non-invasive research tool to quantify the energetics of the heart in diverse patient populations. 31P-MRS is frequently applied alongside other radiological examinations, many of which use various contrast agents that shorten relaxation times of water in conventional proton MR, for a better characterisation of cardiac function, or following prior computed tomography (CT). It is, however, unknown whether these agents confound 31P-MRS signals, for example, 2,3-diphosphoglycerate (2,3-DPG). METHODS In this work, we quantitatively assess the impact of non-ionic, low osmolar iodinated CT contrast agent (iopamidol/Niopam), gadolinium chelates (linear gadopentetic acid dimeglumine/Magnevist and macrocyclic gadoterate meglumine/Dotarem) and superparamagnetic iron oxide nanoparticles (ferumoxytol/Feraheme) on the nuclear T1 and T2 of 31P metabolites (ie, 2,3-DPG), and 1H in water in live human blood and saline phantoms at 11.7 T. RESULTS Addition of all contrast agents led to significant shortening of all relaxation times in both 1H and 31P saline phantoms. On the contrary, the T1 relaxation time of 2,3-DPG in blood was significantly shortened only by Magnevist (P = .03). Similarly, the only contrast agent that influenced the T2 relaxation times of 2,3-DPG in blood samples was ferumoxytol (P = .02). CONCLUSION Our results show that, unlike conventional proton MR, phosphorus MRS is unconfounded in patients who have had prior CT with contrast, not all gadolinium-based contrast agents influence 31P-MRS data in vivo, and that ferumoxytol is a promising contrast agent for the reduction in 31P-MRS blood-pool signal.
Collapse
Affiliation(s)
- Ladislav Valkovič
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Justin Y C Lau
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Ines Abdesselam
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Ivan Frollo
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Damian J Tyler
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Christopher T Rodgers
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- The Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jack J J Miller
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Santini L, Palandri C, Nediani C, Cerbai E, Coppini R. Modelling genetic diseases for drug development: Hypertrophic cardiomyopathy. Pharmacol Res 2020; 160:105176. [DOI: 10.1016/j.phrs.2020.105176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
|
17
|
Apps A, Valkovič L, Peterzan M, Lau JYC, Hundertmark M, Clarke W, Tunnicliffe EM, Ellis J, Tyler DJ, Neubauer S, Rider OJ, Rodgers CT, Schmid AI. Quantifying the effect of dobutamine stress on myocardial Pi and pH in healthy volunteers: A 31 P MRS study at 7T. Magn Reson Med 2020; 85:1147-1159. [PMID: 32929770 PMCID: PMC8239988 DOI: 10.1002/mrm.28494] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 07/07/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Purpose Phosphorus spectroscopy (31P‐MRS) is a proven method to probe cardiac energetics. Studies typically report the phosphocreatine (PCr) to adenosine triphosphate (ATP) ratio. We focus on another 31P signal: inorganic phosphate (Pi), whose chemical shift allows computation of myocardial pH, with Pi/PCr providing additional insight into cardiac energetics. Pi is often obscured by signals from blood 2,3‐diphosphoglycerate (2,3‐DPG). We introduce a method to quantify Pi in 14 min without hindrance from 2,3‐DPG. Methods Using a 31P stimulated echo acquisition mode (STEAM) sequence at 7 Tesla that inherently suppresses signal from 2,3‐DPG, the Pi peak was cleanly resolved. Resting state UTE‐chemical shift imaging (PCr/ATP) and STEAM 31P‐MRS (Pi/PCr, pH) were undertaken in 23 healthy controls; pH and Pi/PCr were subsequently recorded during dobutamine infusion. Results We achieved a clean Pi signal both at rest and stress with good 2,3‐DPG suppression. Repeatability coefficient (8 subjects) for Pi/PCr was 0.036 and 0.12 for pH. We report myocardial Pi/PCr and pH at rest and during catecholamine stress in healthy controls. Pi/PCr was maintained during stress (0.098 ± 0.031 [rest] vs. 0.098 ± 0.031 [stress] P = .95); similarly, pH did not change (7.09 ± 0.07 [rest] vs. 7.08 ± 0.11 [stress] P = .81). Feasibility for patient studies was subsequently successfully demonstrated in a patient with cardiomyopathy. Conclusion We introduced a method that can resolve Pi using 7 Tesla STEAM 31P‐MRS. We demonstrate the stability of Pi/PCr and myocardial pH in volunteers at rest and during catecholamine stress. This protocol is feasible in patients and potentially of use for studying pathological myocardial energetics.
Collapse
Affiliation(s)
- Andrew Apps
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.,Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Mark Peterzan
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Justin Y C Lau
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Moritz Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - William Clarke
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Elizabeth M Tunnicliffe
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jane Ellis
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Damian J Tyler
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Oliver J Rider
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Christopher T Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.,Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Albrecht Ingo Schmid
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.,High Field MR Center, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Solomon T, Filipovska A, Hool L, Viola H. Preventative therapeutic approaches for hypertrophic cardiomyopathy. J Physiol 2020; 599:3495-3512. [PMID: 32822065 PMCID: PMC8359240 DOI: 10.1113/jp279410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/06/2020] [Indexed: 11/08/2022] Open
Abstract
Sarcomeric gene mutations are associated with the development of hypertrophic cardiomyopathy (HCM). Current drug therapeutics for HCM patients are effective in relieving symptoms, but do not prevent or reverse disease progression. Moreover, due to heterogeneity in the clinical manifestations of the disease, patients experience variable outcomes in response to therapeutics. Mechanistically, alterations in calcium handling, sarcomeric disorganization, energy metabolism and contractility participate in HCM disease progression. While some similarities exist, each mutation appears to lead to mutation‐specific pathophysiology. Furthermore, these alterations may precede or proceed development of the pathology. This review assesses the efficacy of HCM therapeutics from studies performed in animal models of HCM and human clinical trials. Evidence suggests that a preventative rather than corrective therapeutic approach may be more efficacious in the treatment of HCM. In addition, a clear understanding of mutation‐specific mechanisms may assist in informing the most effective therapeutic mode of action.
![]()
Collapse
Affiliation(s)
- Tanya Solomon
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Aleksandra Filipovska
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia.,ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, Western Australia, Australia.,Centre for Medical Research, University of Western Australia, QEII Medical Centre, Nedlands, Western Australia, Australia.,Telethon Kids Institute, Perth Children's Hospital, Nedlands, Western Australia, Australia.,School of Molecular Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Livia Hool
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia.,Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Helena Viola
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
19
|
Difference in Metabolomic Response to Exercise between Patients with and without Hypertrophic Cardiomyopathy. J Cardiovasc Transl Res 2020; 14:246-255. [PMID: 32594362 DOI: 10.1007/s12265-020-10051-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/14/2020] [Indexed: 10/24/2022]
Abstract
It is unclear how hypertrophic cardiomyopathy (HCM) affects cardiac metabolic pathways at rest and with exercise. This case-control study compared 15 cases with HCM to 2 control groups without HCM. Metabolomic profiling of 210 metabolites was carried out at rest and at peak exercise. The 50 most discriminant metabolites differentially regulated during exercise were selected using partial least squares discriminant analysis. Pathway enrichment analysis was also performed. At rest, no significant difference was observed in metabolomic profiling of HCM cases as compared to controls. By contrast, there were significant differences in metabolomic profiling in response to exercise (p < 0.05) in the following metabolic pathways: the aminoacyl-tRNA biosynthesis pathway; the nitrogen metabolism pathway; the glycine, serine, and threonine metabolism pathway; and the arginine and proline metabolism pathway. The present study demonstrates differential regulation of several metabolic pathways in patients with HCM in the setting of exercise stress.
Collapse
|
20
|
Mitochondrial Energetics and Ca2 +-Activated ATPase in Obstructive Hypertrophic Cardiomyopathy. J Clin Med 2020; 9:jcm9061799. [PMID: 32527005 PMCID: PMC7356244 DOI: 10.3390/jcm9061799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 11/29/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common genetic disease of the myocardium associated to mutations in sarcomeric genes, but the link between genotype and phenotype remains poorly understood. Magnetic resonance spectroscopy studies have demonstrated impaired cardiac energetics in patients with HCM, and altered mitochondria were described in biopsies, but little is known about possible perturbations of mitochondrial function and adenosine triphosphate (ATP) production/consumption. The aim of this study was to investigate possible abnormalities in mitochondrial enzymes generating/scavenging reactive oxygen species, and changes in the Ca2+-activated ATPases in myocardial tissue from patients with obstructive HCM undergoing surgical myectomy compared to unused donor hearts (CTRL). Methods and Results: Both the amount and activity of mitochondrial Complex I (nicotinamide adenine dinucleotide -reduced form, NADH, dehydrogenase) were upregulated in HCM vs. CTRL, whilst the activity of Complex V (ATP synthase) was not reduced and ATP levels were significantly higher in HCM vs. CTRL. Antioxidant Mn-activated superoxide dismutase (SOD2) and (m)-aconitase activities were increased in HCM vs. CTRL. The Cu/Zn-activated superoxide dismutase (SOD1) amount and mtDNA copy number were unaltered in HCM. Total Ca2+-activated ATPase activity and absolute amount were not different HCM vs. CTRL, but the ratio between ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting type 2 (ATP2A2) and type 1 (ATP2A1), ATP2A2/ATP2A1, was increased in HCM in favor of the slow isoform (ATP2A2). Conclusion: HCM is characterized by mitochondrial Complex I hyperactivity and preserved Ca2+-activated ATPase activity with a partial switch towards slow ATP2A2. This data may give insight into the abnormal cellular energetics observed in HCM cardiomyopathy but other studies would need to be performed to confirm the observations described here.
Collapse
|
21
|
Peterzan MA, Lewis AJM, Neubauer S, Rider OJ. Non-invasive investigation of myocardial energetics in cardiac disease using 31P magnetic resonance spectroscopy. Cardiovasc Diagn Ther 2020; 10:625-635. [PMID: 32695642 DOI: 10.21037/cdt-20-275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiac metabolism and function are intrinsically linked. High-energy phosphates occupy a central and obligate position in cardiac metabolism, coupling oxygen and substrate fuel delivery to the myocardium with external work. This insight underlies the widespread clinical use of ischaemia testing. However, other deficits in high-energy phosphate metabolism (not secondary to supply-demand mismatch of oxygen and substrate fuels) may also be documented, and are of particular interest when found in the context of structural heart disease. This review introduces the scope of deficits in high-energy phosphate metabolism that may be observed in the myocardium, how to assess for them, and how they might be interpreted.
Collapse
Affiliation(s)
- Mark A Peterzan
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J M Lewis
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Parbhudayal RY, Harms HJ, Michels M, van Rossum AC, Germans T, van der Velden J. Increased Myocardial Oxygen Consumption Precedes Contractile Dysfunction in Hypertrophic Cardiomyopathy Caused by Pathogenic TNNT2 Gene Variants. J Am Heart Assoc 2020; 9:e015316. [PMID: 32290750 PMCID: PMC7428531 DOI: 10.1161/jaha.119.015316] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Hypertrophic cardiomyopathy is caused by pathogenic sarcomere gene variants. Individuals with a thin‐filament variant present with milder hypertrophy than carriers of thick‐filament variants, although prognosis is poorer. Herein, we defined if decreased energetic status of the heart is an early pathomechanism in TNNT2 (troponin T gene) variant carriers. Methods and Results Fourteen individuals with TNNT2 variants (genotype positive), without left ventricular hypertrophy (G+/LVH−; n=6) and with LVH (G+/LVH+; n=8) and 14 healthy controls were included. All participants underwent cardiac magnetic resonance and [11C]‐acetate positron emission tomography imaging to assess LV myocardial oxygen consumption, contractile parameters and myocardial external efficiency. Cardiac efficiency was significantly reduced compared with controls in G+/LVH− and G+/LVH+. Lower myocardial external efficiency in G+/LVH− is explained by higher global and regional oxygen consumption compared with controls without changes in contractile parameters. Reduced myocardial external efficiency in G+/LVH+ is explained by the increase in LV mass and higher oxygen consumption. Septal oxygen consumption was significantly lower in G+/LVH+ compared with G+/LVH−. Although LV ejection fraction was higher in G+/LVH+, both systolic and diastolic strain parameters were lower compared with controls, which was most evident in the hypertrophied septal wall. Conclusions Using cardiac magnetic resonance and [11C]‐acetate positron emission tomography imaging, we show that G+/LVH− have an initial increase in oxygen consumption preceding contractile dysfunction and cardiac hypertrophy, followed by a decline in oxygen consumption in G+/LVH+. This suggests that high oxygen consumption and reduced myocardial external efficiency characterize the early gene variant–mediated disease mechanisms that may be used for early diagnosis and development of preventive treatments.
Collapse
Affiliation(s)
- Rahana Y Parbhudayal
- Department of Cardiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands.,Department of Physiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands.,The Netherlands Heart Institute Utrecht the Netherlands
| | - Hendrik J Harms
- Department of Nuclear Medicine and PET Center Aarhus University Aarhus Denmark
| | - Michelle Michels
- Department of Cardiology Erasmus Medical Center Rotterdam the Netherlands
| | - Albert C van Rossum
- Department of Cardiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands
| | - Tjeerd Germans
- Department of Cardiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands
| | - Jolanda van der Velden
- Department of Physiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands.,The Netherlands Heart Institute Utrecht the Netherlands
| |
Collapse
|
23
|
The relation between sarcomere energetics and the rate of isometric tension relaxation in healthy and diseased cardiac muscle. J Muscle Res Cell Motil 2019; 42:47-57. [PMID: 31745760 PMCID: PMC7932984 DOI: 10.1007/s10974-019-09566-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022]
Abstract
Full muscle relaxation happens when [Ca2+] falls below the threshold for force activation. Several experimental models, from whole muscle organs and intact muscle down to skinned fibers, have been used to explore the cascade of kinetic events leading to mechanical relaxation. The use of single myofibrils together with fast solution switching techniques, has provided new information about the role of cross-bridge (CB) dissociation in the time course of isometric force decay. Myofibril’s relaxation is biphasic starting with a slow seemingly linear phase, with a rate constant, slow kREL, followed by a fast mono-exponential phase. Sarcomeres remain isometric during the slow force decay that reflects CB detachment under isometric conditions while the final fast relaxation phase begins with a sudden give of few sarcomeres and is then dominated by intersarcomere dynamics. Based on a simple two-state model of the CB cycle, myofibril slow kREL represents the apparent forward rate with which CBs leave force generating states (gapp) under isometric conditions and correlates with the energy cost of tension generation (ATPase/tension ratio); in short slow kREL ~ gapp ~ tension cost. The validation of this relationship is obtained by simultaneously measuring maximal isometric force and ATP consumption in skinned myocardial strips that provide an unambiguous determination of the relation between contractile and energetic properties of the sarcomere. Thus, combining kinetic experiments in isolated myofibrils and mechanical and energetic measurements in multicellular cardiac strips, we are able to provide direct evidence for a positive linear correlation between myofibril isometric relaxation kinetics (slow kREL) and the energy cost of force production both measured in preparations from the same cardiac sample. This correlation remains true among different types of muscles with different ATPase activities and also when CB kinetics are altered by cardiomyopathy-related mutations. Sarcomeric mutations associated to hypertrophic cardiomyopathy (HCM), a primary cardiac disorder caused by mutations in genes encoding sarcomeric proteins, have been often found to accelerate CB turnover rate and increase the energy cost of myocardial contraction. Here we review data showing that faster CB detachment results in a proportional increase in the energetic cost of tension generation in heart samples from both HCM patients and mouse models of the disease.
Collapse
|
24
|
Valkovič L, Clarke WT, Schmid AI, Raman B, Ellis J, Watkins H, Robson MD, Neubauer S, Rodgers CT. Measuring inorganic phosphate and intracellular pH in the healthy and hypertrophic cardiomyopathy hearts by in vivo 7T 31P-cardiovascular magnetic resonance spectroscopy. J Cardiovasc Magn Reson 2019; 21:19. [PMID: 30871562 PMCID: PMC6419336 DOI: 10.1186/s12968-019-0529-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 02/14/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cardiovascular phosphorus MR spectroscopy (31P-CMRS) is a powerful tool for probing energetics in the human heart, through quantification of phosphocreatine (PCr) to adenosine triphosphate (ATP) ratio. In principle, 31P-CMRS can also measure cardiac intracellular pH (pHi) and the free energy of ATP hydrolysis (ΔGATP). However, these require determination of the inorganic phosphate (Pi) signal frequency and amplitude that are currently not robustly accessible because blood signals often obscure the Pi resonance. Typical cardiac 31P-CMRS protocols use low (e.g. 30°) flip-angles and short repetition time (TR) to maximise signal-to-noise ratio (SNR) within hardware limits. Unfortunately, this causes saturation of Pi with negligible saturation of the flowing blood pool. We aimed to show that an adiabatic 90° excitation, long-TR, 7T 31P-CMRS protocol will reverse this balance, allowing robust cardiac pHi measurements in healthy subjects and patients with hypertrophic cardiomyopathy (HCM). METHODS The cardiac Pi T1 was first measured by the dual TR technique in seven healthy subjects. Next, ten healthy subjects and three HCM patients were scanned with 7T 31P-MRS using long (6 s) TR protocol and adiabatic excitation. Spectra were fitted for cardiac metabolites including Pi. RESULTS The measured Pi T1 was 5.0 ± 0.3 s in myocardium and 6.4 ± 0.6 s in skeletal muscle. Myocardial pH was 7.12 ± 0.04 and Pi/PCr ratio was 0.11 ± 0.02. The coefficients of repeatability were 0.052 for pH and 0.027 for Pi/PCr quantification. The pH in HCM patients did not differ (p = 0.508) from volunteers. However, Pi/PCr was higher (0.24 ± 0.09 vs. 0.11 ± 0.02; p = 0.001); Pi/ATP was higher (0.44 ± 0.14 vs. 0.24 ± 0.05; p = 0.002); and PCr/ATP was lower (1.78 ± 0.07 vs. 2.10 ± 0.20; p = 0.020), in HCM patients, which is in agreement with previous reports. CONCLUSION A 7T 31P-CMRS protocol with adiabatic 90° excitation and long (6 s) TR gives sufficient SNR for Pi and low enough blood signal to permit robust quantification of cardiac Pi and hence pHi. Pi was detectable in every subject scanned for this study, both in healthy subjects and HCM patients. Cardiac pHi was unchanged in HCM patients, but both Pi/PCr and Pi/ATP increased that indicate an energetic impairment in HCM. This work provides a robust technique to quantify cardiac Pi and pHi.
Collapse
Affiliation(s)
- Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - William T. Clarke
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Albrecht I. Schmid
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- High-Field MR Centre, Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Jane Ellis
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Matthew D. Robson
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Christopher T. Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- The Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
25
|
Piroddi N, Witjas-Paalberends ER, Ferrara C, Ferrantini C, Vitale G, Scellini B, Wijnker PJM, Sequiera V, Dooijes D, Dos Remedios C, Schlossarek S, Leung MC, Messer A, Ward DG, Biggeri A, Tesi C, Carrier L, Redwood CS, Marston SB, van der Velden J, Poggesi C. The homozygous K280N troponin T mutation alters cross-bridge kinetics and energetics in human HCM. J Gen Physiol 2018; 151:18-29. [PMID: 30578328 PMCID: PMC6314385 DOI: 10.1085/jgp.201812160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/29/2018] [Indexed: 01/24/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomeric proteins, but the pathogenic mechanism is unclear. Piroddi et al. find impairment of cross-bridge kinetics and energetics in human sarcomeres with a TNNT2 mutation, suggesting that HCM involves inefficient ATP utilization. Hypertrophic cardiomyopathy (HCM) is a genetic form of left ventricular hypertrophy, primarily caused by mutations in sarcomere proteins. The cardiac remodeling that occurs as the disease develops can mask the pathogenic impact of the mutation. Here, to discriminate between mutation-induced and disease-related changes in myofilament function, we investigate the pathogenic mechanisms underlying HCM in a patient carrying a homozygous mutation (K280N) in the cardiac troponin T gene (TNNT2), which results in 100% mutant cardiac troponin T. We examine sarcomere mechanics and energetics in K280N-isolated myofibrils and demembranated muscle strips, before and after replacement of the endogenous troponin. We also compare these data to those of control preparations from donor hearts, aortic stenosis patients (LVHao), and HCM patients negative for sarcomeric protein mutations (HCMsmn). The rate constant of tension generation following maximal Ca2+ activation (kACT) and the rate constant of isometric relaxation (slow kREL) are markedly faster in K280N myofibrils than in all control groups. Simultaneous measurements of maximal isometric ATPase activity and Ca2+-activated tension in demembranated muscle strips also demonstrate that the energy cost of tension generation is higher in the K280N than in all controls. Replacement of mutant protein by exchange with wild-type troponin in the K280N preparations reduces kACT, slow kREL, and tension cost close to control values. In donor myofibrils and HCMsmn demembranated strips, replacement of endogenous troponin with troponin containing the K280N mutant increases kACT, slow kREL, and tension cost. The K280N TNNT2 mutation directly alters the apparent cross-bridge kinetics and impairs sarcomere energetics. This result supports the hypothesis that inefficient ATP utilization by myofilaments plays a central role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Nicoletta Piroddi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - E Rosalie Witjas-Paalberends
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Claudia Ferrara
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Cecilia Ferrantini
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy.,LENS, Sesto Fiorentino (Firenze), Florence, Italy
| | - Giulia Vitale
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Beatrice Scellini
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Paul J M Wijnker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Vasco Sequiera
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Dennis Dooijes
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Clinical Genetics, University Medical Center, Utrecht, Netherlands
| | - Cristobal Dos Remedios
- Department of Anatomy and Histology, Bosch Institute, The University of Sydney, Sydney, Australia
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Man Ching Leung
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Andrew Messer
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Douglas G Ward
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Chiara Tesi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | | | - Steven B Marston
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Clinical Genetics, University Medical Center, Utrecht, Netherlands.,ICIN-Netherlands, Heart Institute, Utrecht, Netherlands
| | - Corrado Poggesi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy .,LENS, Sesto Fiorentino (Firenze), Florence, Italy
| |
Collapse
|
26
|
Cohn R, Thakar K, Lowe A, Ladha FA, Pettinato AM, Romano R, Meredith E, Chen YS, Atamanuk K, Huey BD, Hinson JT. A Contraction Stress Model of Hypertrophic Cardiomyopathy due to Sarcomere Mutations. Stem Cell Reports 2018; 12:71-83. [PMID: 30554920 PMCID: PMC6335568 DOI: 10.1016/j.stemcr.2018.11.015] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022] Open
Abstract
Thick-filament sarcomere mutations are a common cause of hypertrophic cardiomyopathy (HCM), a disorder of heart muscle thickening associated with sudden cardiac death and heart failure, with unclear mechanisms. We engineered four isogenic induced pluripotent stem cell (iPSC) models of β-myosin heavy chain and myosin-binding protein C3 mutations, and studied iPSC-derived cardiomyocytes in cardiac microtissue assays that resemble cardiac architecture and biomechanics. All HCM mutations resulted in hypercontractility with prolonged relaxation kinetics in proportion to mutation pathogenicity, but not changes in calcium handling. RNA sequencing and expression studies of HCM models identified p53 activation, oxidative stress, and cytotoxicity induced by metabolic stress that can be reversed by p53 genetic ablation. Our findings implicate hypercontractility as a direct consequence of thick-filament mutations, irrespective of mutation localization, and the p53 pathway as a molecular marker of contraction stress and candidate therapeutic target for HCM patients.
Collapse
Affiliation(s)
- Rachel Cohn
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Ketan Thakar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Andre Lowe
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Feria A Ladha
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Anthony M Pettinato
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Robert Romano
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Emily Meredith
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Yu-Sheng Chen
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Katherine Atamanuk
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Bryan D Huey
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA.
| |
Collapse
|
27
|
Ren J, Shang T, Sherry AD, Malloy CR. Unveiling a hidden 31 P signal coresonating with extracellular inorganic phosphate by outer-volume-suppression and localized 31 P MRS in the human brain at 7T. Magn Reson Med 2018; 80:1289-1297. [PMID: 29427295 PMCID: PMC6085175 DOI: 10.1002/mrm.27121] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/22/2017] [Accepted: 01/15/2018] [Indexed: 01/19/2023]
Abstract
PURPOSE The study was undertaken to demonstrate that there is more than 1 component in the extracellular Pi31 P signal ( Piex) acquired from human head using nonlocalized 31 P MRS. METHODS Outer-volume-suppression (OVS) saturation and 1D/2D 31 P CSI were utilized to reveal the presence of an additional component in the Piex signal. RESULTS 67% of the head extracellular Pi signal was attenuated upon OVS saturation of the peripheral meningeal tissues, likely reflecting elimination of the Pi signal in the meningeal fluids (the blood and CSF). Localized 1D/2D CSI data provided further support for this assignment. Upon correction for the meningeal contribution, the extracellular Pi concentration was 0.51 ± 0.07 mM, whereas the intracellular Pi was 0.85 ± 0.10 mM. The extracellular pH was measured as 7.32 ± 0.04 when using OVS, as compared to 7.39 ± 0.03 when measured without OVS (N = 7 subjects). CONCLUSION The extracellular Pi signal acquired from the human head using nonlocalized 31 P MRS contains a significant component likely contributed by peripheral blood and CSF in meninges that must be removed in order to use this signal as an endogenous probe for measuring extracellular pH and other properties in the brain.
Collapse
Affiliation(s)
- Jimin Ren
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ty Shang
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - A. Dean Sherry
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Chemistry, University of Texas at Dallas, Richardson, TX 75080
| | - Craig R. Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- VA North Texas Health Care System, Dallas, TX 75216
| |
Collapse
|
28
|
|
29
|
Lu DY, Yalçin H, Yalçin F, Zhao M, Sivalokanathan S, Valenta I, Tahari A, Pomper MG, Abraham TP, Schindler TH, Abraham MR. Stress Myocardial Blood Flow Heterogeneity Is a Positron Emission Tomography Biomarker of Ventricular Arrhythmias in Patients With Hypertrophic Cardiomyopathy. Am J Cardiol 2018; 121:1081-1089. [PMID: 29678336 DOI: 10.1016/j.amjcard.2018.01.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/03/2018] [Accepted: 01/09/2018] [Indexed: 02/01/2023]
Abstract
Patients with hypertrophic cardiomyopathy (HC) are at increased risk of sudden cardiac death. Abnormalities in myocardial blood flow (MBF) detected by positron emission tomography (PET) are common in HC, but a PET marker that identifies patients at risk of sudden cardiac death is lacking. We hypothesized that disparities in regional myocardial perfusion detected by PET would identify patients with HC at risk of ventricular arrhythmias. To test this hypothesis, we quantified global and regional MBFs by 13NH3-PET at rest and at stress, and developed a heterogeneity index to assess MBF heterogeneity in 133 symptomatic patients with HC. The MBF heterogeneity index was computed by dividing the highest by the lowest regional MBF value, at rest and after vasodilator stress, in each patient. High stress MBF heterogeneity was defined as an index of ≧1.85. Patients with HC were stratified by the presence or the absence of ventricular arrhythmias, defined as sustained ventricular tachycardia (VT) and/or nonsustained VT, during follow-up. We found that global and regional MBFs at rest and stress were similar in patients with HC with or without ventricular arrhythmias. Variability in regional stress MBF was observed in both groups, but the stress MBF heterogeneity index was significantly higher in patients with HC who developed ventricular arrhythmias (1.82 ± 0.77 vs 1.49 ± 0.25, p <0.001). A stress MBF heterogeneity index of ≧1.85 was an independent predictor of both sustained VT (hazard ratio 16.1, 95% confidence interval 3.2 to 80.3) and all-VT (sustained-VT + nonsustained VT: hazard ratio 3.7, 95% confidence interval 1.4 to 9.7). High heterogeneity of stress MBF, reflected by an MBF heterogeneity index of ≥1.85, is a PET biomarker for ventricular arrhythmias in symptomatic patients with HC.
Collapse
Affiliation(s)
- Dai-Yin Lu
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Hulya Yalçin
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland
| | - Fatih Yalçin
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland
| | - Min Zhao
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Sanjay Sivalokanathan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland
| | - Ines Valenta
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Abdel Tahari
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Theodore P Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland; Hypertrophic Cardiomyopathy Center, Division of Cardiology, University of California San Francisco, San Francisco, California
| | - Thomas H Schindler
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - M Roselle Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland; Hypertrophic Cardiomyopathy Center, Division of Cardiology, University of California San Francisco, San Francisco, California.
| |
Collapse
|
30
|
Coronary Microcirculatory Dysfunction in Human Cardiomyopathies: A Pathologic and Pathophysiologic Review. Cardiol Rev 2018; 25:165-178. [PMID: 28574936 DOI: 10.1097/crd.0000000000000140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cardiomyopathies are a heterogeneous group of diseases of the myocardium. The term cardiomyopathy involves a wide range of pathogenic mechanisms that affect the structural and functional states of cardiomyocytes, extravascular tissues, and coronary vasculature, including both epicardial coronary arteries and the microcirculation. In the developed phase, cardiomyopathies present with various clinical symptoms: dyspnea, chest pain, palpitations, swelling of the extremities, arrhythmias, and sudden cardiac death. Due to the heterogeneity of cardiomyopathic patterns and symptoms, their diagnosis and therapies are great challenges. Despite extensive research, the relation between the structural and functional abnormalities of the myocardium and the coronary circulation are still not well understood in the various forms of cardiomyopathy. The main pathological characteristics of cardiomyopathies and the coronary microcirculation develop in a progressive manner due to (1) genetic-immunologic-systemic factors; (2) comorbidities with endothelial, myogenic, metabolic, and inflammatory changes; (3) aging-induced arteriosclerosis; and (4) myocardial fibrosis. The aim of this review is to summarize the most important common pathological features and/or adaptations of the coronary microcirculation in various types of cardiomyopathies and to integrate the present understanding of the underlying pathophysiological mechanisms responsible for the development of various types of cardiomyopathies. Although microvascular dysfunction is present and contributes to cardiac dysfunction and the potential outcome of disease, the current therapeutic approaches are not specific for the given types of cardiomyopathy.
Collapse
|
31
|
Mazurkiewicz Ł, Ziółkowska L, Petryka J, Śpiewak M, Małek Ł, Kubik A, Marczak M, Misko J, Brzezińska-Rajszys G. Left-ventricular mechanics in children with hypertrophic cardiomyopathy. CMR study. Magn Reson Imaging 2017; 43:56-65. [PMID: 28688952 DOI: 10.1016/j.mri.2017.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 01/16/2023]
Abstract
OBJECTIVES To assess the magnitude of myocardial displacement abnormalities and their alterations with the fibrosis, left-ventricular (LV) outflow tract obstruction (LVOTO) and hypertrophy in juveniles with hypertrophic cardiomyopathy (HCM). STUDY DESIGN Fifty-five children [age 12,5±4.6years, 38 (69,1%) males, 19 (34,5%) with LVOTO] with HCM and 20 controls underwent cardiovascular magnetic resonance. The LV feature tracking (FT) derived strain and strain rates were quantified. Results of FT analysis were compared between HCM subjects and controls and between children with and without LVOTO. RESULTS Children with HCM exhibited decreased strain in both hypertrophied and nonhypertrophied segments versus controls. LV global longitudinal strain (LVGLS) rate (-0.69±0.04 vs -0.91±0.05, p=0,04), LV circumferential strain (LVCR) rate (-0.98±0.09 vs -1.27±0.06, p=0,02), LV radial strain (LVR) (18,5±1.9 vs 27,4±1.4, p<0,01) and LVR rate (0,98±0.1 vs 1,53±0.08, p<0,01) were substantially compromised in subjects with LVOTO vs without. In multivariable regression all LV myocardial dynamics markers, except for LVCR, exhibited a significant association with the degree of LVOTO. LVCR rate (β=0,31, p=0,02) and LVR (β=-0.24, p=0,04) were related to LV mass and only LVCR rate (β=0,15, p=0,03) was associated with the amount of LV fibrosis. CONCLUSIONS The reduction of all indices of LV myocardial mechanics in juvenile HCM patients was global but particularly pronounced in hypertrophied segments of the LV. The majority of the LV strains and strain rates were substantially compromised in subjects with LVOTO compared to patients without the obstruction. Myocardial mechanics indices seemed to be related to the degree of LVOTO rather than either to mass or the amount of fibrosis.
Collapse
Affiliation(s)
- Łukasz Mazurkiewicz
- Department of Cardiomyopathies, CMR Unit, Institute of Cardiology, Warsaw, Poland.
| | - Lidia Ziółkowska
- Department of Pediatric Cardiology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Joanna Petryka
- Department of Coronary and Structural Heart Diseases, CMR Unit, Institute of Cardiology, Warsaw, Poland
| | | | | | - Agata Kubik
- CMR Unit, Institute of Cardiology, Warsaw, Poland
| | | | | | | |
Collapse
|
32
|
Alamo L, Ware JS, Pinto A, Gillilan RE, Seidman JG, Seidman CE, Padrón R. Effects of myosin variants on interacting-heads motif explain distinct hypertrophic and dilated cardiomyopathy phenotypes. eLife 2017; 6:e24634. [PMID: 28606303 PMCID: PMC5469618 DOI: 10.7554/elife.24634] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 05/05/2017] [Indexed: 12/12/2022] Open
Abstract
Cardiac β-myosin variants cause hypertrophic (HCM) or dilated (DCM) cardiomyopathy by disrupting sarcomere contraction and relaxation. The locations of variants on isolated myosin head structures predict contractility effects but not the prominent relaxation and energetic deficits that characterize HCM. During relaxation, pairs of myosins form interacting-heads motif (IHM) structures that with other sarcomere proteins establish an energy-saving, super-relaxed (SRX) state. Using a human β-cardiac myosin IHM quasi-atomic model, we defined interactions sites between adjacent myosin heads and associated protein partners, and then analyzed rare variants from 6112 HCM and 1315 DCM patients and 33,370 ExAC controls. HCM variants, 72% that changed electrostatic charges, disproportionately altered IHM interaction residues (expected 23%; HCM 54%, p=2.6×10-19; DCM 26%, p=0.66; controls 20%, p=0.23). HCM variant locations predict impaired IHM formation and stability, and attenuation of the SRX state - accounting for altered contractility, reduced diastolic relaxation, and increased energy consumption, that fully characterizes HCM pathogenesis.
Collapse
Affiliation(s)
- Lorenzo Alamo
- Centro de Biología Estructural, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - James S Ware
- National Heart and Lung Institute and MRC London Institute for Medical Sciences, Imperial College London, London, United Kingdom
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Antonio Pinto
- Centro de Biología Estructural, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - Richard E Gillilan
- Macromolecular Diffraction Facility, Cornell High Energy Synchrotron Source, Ithaca, United States
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, United States
- Cardiovascular Division, Brigham and Women’s Hospital and Howard Hughes Medical Institute, Boston, United States
| | - Raúl Padrón
- Centro de Biología Estructural, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| |
Collapse
|
33
|
Principals and clinical applications of magnetic resonance cardiac spectroscopy in heart failure. Heart Fail Rev 2017; 22:491-499. [DOI: 10.1007/s10741-017-9611-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Secchi F, Di Leo G, Petrini M, Spairani R, Alì M, Guazzi M, Sardanelli F. 1H- and 31P-myocardial magnetic resonance spectroscopy in non-obstructive hypertrophic cardiomyopathy patients and competitive athletes. Radiol Med 2017; 122:265-272. [PMID: 28070839 DOI: 10.1007/s11547-016-0718-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
PURPOSE The clinical differentiation between athlete's heart and mild forms of non-obstructive hypertrophic cardiomyopathy (HCM) is crucial. We hypothesized that differences do exist between the myocardial metabolism of patients with non-obstructive HCM and competitive athletes (CAs). Our aim was to evaluate myocardial metabolism with 31P-MRS and 1H-MRS in HCM patients and CAs. MATERIALS AND METHODS After Ethics Committee approval, 15 CAs and 7 HCM patients were prospectively enrolled. They underwent a 1.5-T cardiac MR including electrocardiographically triggered cine images, single-voxel 1H-MRS and multivoxel 31P-MRS. 1H-MRS was performed after imaging using standard coil with the patient in the supine position; thereafter, 31P-MRS was performed using a dedicated coil, in the prone position. Data were reported as median and interquartile range. Mann-Whitney U test was used. RESULTS In CAs, left ventricular mass index was 72 (66-83) g/m2, septal thickness 10 (10-11) mm, end diastolic volume index 95 (85-102) ml/m2, end systolic volume index 30 (28-32) ml/m2 and ejection fraction 68% (65-69%); in HCM patients, 81 (76-111) g/m2 (P = 0.052), 18 (15-21) mm (P = 0.003), 73 (58-76) ml/m2 (P = 0.029), 20 (16-34) ml/m2 (P = 0.274) and 68% (55-73%) (P = 1.000), respectively. At 1H-MRS, total lipids were 35 (0-183) arbitrary units (au) for CA and 763 (155-1994) au for HCM patients (P = 0.046). At 31P-MRS, PCr/γATP was 5 (4-6) au for CA and 4 (2-5) au for HCM patients (P = 0.230). Examination time was 20 min for imaging only, 5 min for 1H-MRS and 15 min for 31P-MRS. CONCLUSIONS We observed a significant increase of myocardial lipids, but a preserved PCr/γATP ratio in the metabolism of HCM patients compared with competitive CAs.
Collapse
Affiliation(s)
- Francesco Secchi
- Unit of Radiology, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097, Milan, Italy
| | - Giovanni Di Leo
- Unit of Radiology, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097, Milan, Italy
| | - Marcello Petrini
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono 7, 20100, Milan, Italy
| | - Riccardo Spairani
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono 7, 20100, Milan, Italy
| | - Marco Alì
- Ph.D. Course in Integrated Biomedical Research, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
| | - Marco Guazzi
- Cardiology University Department, IRCCS Policlinico San Donato, Università degli Studi di Milano, Via Morandi 30, San Donato Milanese, 20097, Milan, Italy
| | - Francesco Sardanelli
- Unit of Radiology, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097, Milan, Italy.,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Morandi 30, 20133, Milan, Italy
| |
Collapse
|
35
|
Brown DA, Perry JB, Allen ME, Sabbah HN, Stauffer BL, Shaikh SR, Cleland JGF, Colucci WS, Butler J, Voors AA, Anker SD, Pitt B, Pieske B, Filippatos G, Greene SJ, Gheorghiade M. Expert consensus document: Mitochondrial function as a therapeutic target in heart failure. Nat Rev Cardiol 2016; 14:238-250. [PMID: 28004807 PMCID: PMC5350035 DOI: 10.1038/nrcardio.2016.203] [Citation(s) in RCA: 542] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Heart failure is a pressing worldwide public-health problem with millions of patients having worsening heart failure. Despite all the available therapies, the condition carries a very poor prognosis. Existing therapies provide symptomatic and clinical benefit, but do not fully address molecular abnormalities that occur in cardiomyocytes. This shortcoming is particularly important given that most patients with heart failure have viable dysfunctional myocardium, in which an improvement or normalization of function might be possible. Although the pathophysiology of heart failure is complex, mitochondrial dysfunction seems to be an important target for therapy to improve cardiac function directly. Mitochondrial abnormalities include impaired mitochondrial electron transport chain activity, increased formation of reactive oxygen species, shifted metabolic substrate utilization, aberrant mitochondrial dynamics, and altered ion homeostasis. In this Consensus Statement, insights into the mechanisms of mitochondrial dysfunction in heart failure are presented, along with an overview of emerging treatments with the potential to improve the function of the failing heart by targeting mitochondria.
Collapse
Affiliation(s)
- David A Brown
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, 1035 Integrated Life Sciences Building, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Justin B Perry
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, 1035 Integrated Life Sciences Building, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Mitchell E Allen
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, 1035 Integrated Life Sciences Building, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Hani N Sabbah
- Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, Michigan 48202, USA
| | - Brian L Stauffer
- Division of Cardiology, Department of Medicine, University of Colorado Denver, 12700 East 19th Avenue, B139, Aurora, Colorado 80045, USA
| | - Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA
| | - John G F Cleland
- National Heart &Lung Institute, National Institute of Health Research Cardiovascular Biomedical Research Unit, Royal Brompton &Harefield Hospitals, Imperial College, London, UK
| | - Wilson S Colucci
- Cardiovascular Medicine Section, Boston University School of Medicine and Boston Medical Center, 88 East Newton Street, C-8, Boston, Massachusetts 02118, USA
| | - Javed Butler
- Division of Cardiology, Health Sciences Center, T-16 Room 080, SUNY at Stony Brook, New York 11794, USA
| | - Adriaan A Voors
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen 9713 GZ, Netherlands
| | - Stefan D Anker
- Department of Innovative Clinical Trials, University Medical Centre Göttingen (UMG), Robert-Koch-Straße, D-37075, Göttingen, Germany
| | - Bertram Pitt
- University of Michigan School of Medicine, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| | - Burkert Pieske
- Department of Cardiology, Charité University Medicine, Campus Virchow Klinikum, and German Heart Center Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Gerasimos Filippatos
- National and Kopodistrian University of Athens, School of Medicine, Heart Failure Unit, Department of Cardiology, Athens University Hospital Attikon, Rimini 1, Athens 12462, Greece
| | - Stephen J Greene
- Division of Cardiology, Duke University Medical Center, 2301 Erwin Road Suite 7400, Durham, North Carolina 27705, USA
| | - Mihai Gheorghiade
- Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, 201 East Huron, Galter 3-150, Chicago, Illinois 60611, USA
| |
Collapse
|
36
|
Abstract
Cardiorenal syndrome type 5 (CRS-5) includes conditions where there is a simultaneous involvement of the heart and kidney from a systemic disorder. This is a bilateral organ cross talk. Fabry's disease (FD) is a devastating progressive inborn error of metabolism with lysosomal glycosphingolipid deposition in variety of cell types, capillary endothelial cells, renal, cardiac and nerve cells. Basic effect is absent or deficient activity of lysosomal exoglycohydrolase a-galactosidase A. Renal involvement consists of proteinuria, isosthenuria, altered tubular function, presenting in second or third decade leading to azotemia and end-stage renal disease in third to fifth decade mainly due to irreversible changes to glomerular, tubular and vascular structures, especially highlighted by podocytes foot process effacement. Cardiac involvement consists of left ventricular hypertrophy, right ventricular hypertrophy, arrhythmias (sinus node and conduction system impairment), diastolic dysfunction, myocardial ischemia, infarction, transmural replacement fibrosis, congestive heart failure and cardiac death. Management of FD is based on enzymatic replacement therapy and control of renal (with anti-proteinuric agents such as angiotensin-converting enzyme inhibitors-and/or angiotensin II receptor blockers), brain (coated aspirin, clopidogrel and statin to prevent strokes) and heart complications (calcium channel blockers for ischemic cardiomyopathy, warfarin and amiodarone or cardioverter device for arrhythmias).
Collapse
|
37
|
|
38
|
Nuclear Imaging for Assessment of Myocardial Perfusion, Metabolism, and Innervation in Hypertrophic Cardiomyopathy. CURRENT CARDIOVASCULAR IMAGING REPORTS 2016. [DOI: 10.1007/s12410-016-9379-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
39
|
Löring J, van der Kemp WJM, Almujayyaz S, van Oorschot JWM, Luijten PR, Klomp DWJ. Whole-body radiofrequency coil for (31) P MRSI at 7 T. NMR IN BIOMEDICINE 2016; 29:709-20. [PMID: 27037615 DOI: 10.1002/nbm.3517] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/05/2016] [Accepted: 02/19/2016] [Indexed: 05/12/2023]
Abstract
Widespread use of ultrahigh-field (31) P MRSI in clinical studies is hindered by the limited field of view and non-uniform radiofrequency (RF) field obtained from surface transceivers. The non-uniform RF field necessitates the use of high specific absorption rate (SAR)-demanding adiabatic RF pulses, limiting the signal-to-noise ratio (SNR) per unit of time. Here, we demonstrate the feasibility of using a body-sized volume RF coil at 7 T, which enables uniform excitation and ultrafast power calibration by pick-up probes. The performance of the body coil is examined by bench tests, and phantom and in vivo measurements in a 7-T MRI scanner. The accuracy of power calibration with pick-up probes is analyzed at a clinical 3-T MR system with a close to identical (1) H body coil integrated at the MR system. Finally, we demonstrate high-quality three-dimensional (31) P MRSI of the human body at 7 T within 5 min of data acquisition that includes RF power calibration. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- J Löring
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - W J M van der Kemp
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - J W M van Oorschot
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - P R Luijten
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - D W J Klomp
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
- MR Coils BV, Zaltbommel, the Netherlands
| |
Collapse
|
40
|
Ultrastructural myocardial changes in seven cats with spontaneous hypertrophic cardiomyopathy. J Vet Cardiol 2015; 17 Suppl 1:S220-32. [DOI: 10.1016/j.jvc.2015.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 10/05/2015] [Accepted: 10/15/2015] [Indexed: 11/22/2022]
|
41
|
Dass S, Cochlin LE, Suttie JJ, Holloway CJ, Rider OJ, Carden L, Tyler DJ, Karamitsos TD, Clarke K, Neubauer S, Watkins H. Exacerbation of cardiac energetic impairment during exercise in hypertrophic cardiomyopathy: a potential mechanism for diastolic dysfunction. Eur Heart J 2015; 36:1547-54. [PMID: 25990345 DOI: 10.1093/eurheartj/ehv120] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 03/24/2015] [Indexed: 11/14/2022] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is the commonest cause of sudden cardiac death in the young, with an excess of exercise-related deaths. The HCM sarcomere mutations increase the energy cost of contraction and impaired resting cardiac energetics has been documented by measurement of phosphocreatine/ATP (PCr/ATP) using (31)Phosphorus MR Spectroscopy ((31)P MRS). We hypothesized that cardiac energetics are further impaired acutely during exercise in HCM and that this would have important functional consequences. METHODS AND RESULTS (31)P MRS was performed in 35 HCM patients and 20 age- and gender-matched normal volunteers at rest and during leg exercise with 2.5 kg ankle weights. Peak left-ventricular filling rates (PFRs) and myocardial perfusion reserve (MPRI) were calculated during adenosine stress. Resting PCr/ATP was significantly reduced in HCM (HCM: 1.71 ± 0.35, normal 2.14 ± 0.35 P < 0.0001). During exercise, there was a further reduction in PCr/ATP in HCM (1.56 ± 0.29, P = 0.02 compared with rest) but not in normals (2.16 ± 0.26, P = 0.98 compared with rest). There was no correlation between PCr/ATP reduction and cardiac mass, wall thickness, MPRI, or late-gadolinium enhancement. PFR and PCr/ATP were significantly correlated at rest (r = 0.48, P = 0.02) and stress (r = 0.53, P = 0.01). CONCLUSION During exercise, the pre-existing energetic deficit in HCM is further exacerbated independent of hypertrophy, perfusion reserve, or degree of fibrosis. This is in keeping with the change at the myofilament level. We offer a potential explanation for exercise-related diastolic dysfunction in HCM.
Collapse
Affiliation(s)
- Sairia Dass
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Lowri E Cochlin
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Joseph J Suttie
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Cameron J Holloway
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Leah Carden
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Theodoros D Karamitsos
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Anatomy and Genetics, Oxford University, Oxford, UK
| |
Collapse
|
42
|
Christiansen LB, Dela F, Koch J, Hansen CN, Leifsson PS, Yokota T. Impaired cardiac mitochondrial oxidative phosphorylation and enhanced mitochondrial oxidative stress in feline hypertrophic cardiomyopathy. Am J Physiol Heart Circ Physiol 2015; 308:H1237-47. [DOI: 10.1152/ajpheart.00727.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/10/2015] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction and oxidative stress are important players in the development of various cardiovascular diseases, but their roles in hypertrophic cardiomyopathy (HCM) remain unknown. We examined whether mitochondrial oxidative phosphorylation (OXPHOS) capacity was impaired with enhanced mitochondrial oxidative stress in HCM. Cardiac and skeletal muscles were obtained from 9 domestic cats with spontaneously occurring HCM with preserved left ventricular systolic function and from 15 age-matched control cats. Mitochondrial OXPHOS capacities with nonfatty acid and fatty acid substrates in permeabilized fibers and isolated mitochondria were assessed using high-resolution respirometry. ROS release originating from isolated mitochondria was assessed by spectrofluorometry. Thiobarbituric acid-reactive substances were also measured as a marker of oxidative damage. Mitochondrial ADP-stimulated state 3 respiration with complex I-linked nonfatty acid substrates and with fatty acid substrates, respectively, was significantly lower in the hearts of HCM cats compared with control cats. Mitochondrial ROS release during state 3 with complex I-linked substrates and thiobarbituric acid-reactive substances in the heart were significantly increased in cats with HCM. In contrast, there were no significant differences in mitochondrial OXPHOS capacity, mitochondrial ROS release, and oxidative damage in skeletal muscle between groups. Mitochondrial OXPHOS capacity with both nonfatty acid substrates and fatty acid substrates was impaired with increased mitochondrial ROS release in the feline HCM heart. These findings provide new insights into the pathophysiology of HCM and support the hypothesis that restoration of the redox state in the mitochondria is beneficial in the treatment of HCM.
Collapse
Affiliation(s)
- Liselotte B. Christiansen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; and
| | - Flemming Dela
- Department of Biomedical Sciences, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; and
| | - Jørgen Koch
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina N. Hansen
- Department of Biomedical Sciences, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; and
| | - Pall S. Leifsson
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Takashi Yokota
- Department of Biomedical Sciences, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; and
| |
Collapse
|
43
|
Cardim N, Galderisi M, Edvardsen T, Plein S, Popescu BA, D'Andrea A, Bruder O, Cosyns B, Davin L, Donal E, Freitas A, Habib G, Kitsiou A, Petersen SE, Schroeder S, Lancellotti P, Camici P, Dulgheru R, Hagendorff A, Lombardi M, Muraru D, Sicari R. Role of multimodality cardiac imaging in the management of patients with hypertrophic cardiomyopathy: an expert consensus of the European Association of Cardiovascular Imaging Endorsed by the Saudi Heart Association. Eur Heart J Cardiovasc Imaging 2015; 16:280. [PMID: 25650407 DOI: 10.1093/ehjci/jeu291] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Taking into account the complexity and limitations of clinical assessment in hypertrophic cardiomyopathy (HCM), imaging techniques play an essential role in the evaluation of patients with this disease. Thus, in HCM patients, imaging provides solutions for most clinical needs, from diagnosis to prognosis and risk stratification, from anatomical and functional assessment to ischaemia detection, from metabolic evaluation to monitoring of treatment modalities, from staging and clinical profiles to follow-up, and from family screening and preclinical diagnosis to differential diagnosis. Accordingly, a multimodality imaging (MMI) approach (including echocardiography, cardiac magnetic resonance, cardiac computed tomography, and cardiac nuclear imaging) is encouraged in the assessment of these patients. The choice of which technique to use should be based on a broad perspective and expert knowledge of what each technique has to offer, including its specific advantages and disadvantages. Experts in different imaging techniques should collaborate and the different methods should be seen as complementary, not as competitors. Each test must be selected in an integrated and rational way in order to provide clear answers to specific clinical questions and problems, trying to avoid redundant and duplicated information, taking into account its availability, benefits, risks, and cost.
Collapse
MESH Headings
- Cardiac Imaging Techniques/methods
- Cardiac Imaging Techniques/standards
- Cardiomyopathy, Hypertrophic/diagnosis
- Cardiomyopathy, Hypertrophic/therapy
- Consensus
- Echocardiography, Doppler/methods
- Echocardiography, Doppler/standards
- Europe
- Female
- Humans
- Image Interpretation, Computer-Assisted
- Magnetic Resonance Imaging, Cine/methods
- Magnetic Resonance Imaging, Cine/standards
- Male
- Multimodal Imaging/methods
- Multimodal Imaging/standards
- Positron-Emission Tomography/methods
- Positron-Emission Tomography/standards
- Practice Guidelines as Topic/standards
- Role
- Saudi Arabia
- Societies, Medical/standards
- Tomography, X-Ray Computed/methods
- Tomography, X-Ray Computed/standards
Collapse
|
44
|
Månsson A. Hypothesis and theory: mechanical instabilities and non-uniformities in hereditary sarcomere myopathies. Front Physiol 2014; 5:350. [PMID: 25309450 PMCID: PMC4163974 DOI: 10.3389/fphys.2014.00350] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/26/2014] [Indexed: 12/23/2022] Open
Abstract
Familial hypertrophic cardiomyopathy (HCM), due to point mutations in genes for sarcomere proteins such as myosin, occurs in 1/500 people and is the most common cause of sudden death in young individuals. Similar mutations in skeletal muscle, e.g., in the MYH7 gene for slow myosin found in both the cardiac ventricle and slow skeletal muscle, may also cause severe disease but the severity and the morphological changes are often different. In HCM, the modified protein function leads, over years to decades, to secondary remodeling with substantial morphological changes, such as hypertrophy, myofibrillar disarray, and extensive fibrosis associated with severe functional deterioration. Despite intense studies, it is unclear how the moderate mutation-induced changes in protein function cause the long-term effects. In hypertrophy of the heart due to pressure overload (e.g., hypertension), mechanical stress in the myocyte is believed to be major initiating stimulus for activation of relevant cell signaling cascades. Here it is considered how expression of mutated proteins, such as myosin or regulatory proteins, could have similar consequences through one or both of the following mechanisms: (1) contractile instabilities within each sarcomere (with more than one stable velocity for a given load), (2) different tension generating capacities of cells in series. These mechanisms would have the potential to cause increased tension and/or stretch of certain cells during parts of the cardiac cycle. Modeling studies are used to illustrate these ideas and experimental tests are proposed. The applicability of similar ideas to skeletal muscle is also postulated, and differences between heart and skeletal muscle are discussed.
Collapse
Affiliation(s)
- Alf Månsson
- Department of Chemistry and Biomedical Sciences, Linnaeus University Kalmar, Sweden
| |
Collapse
|
45
|
Vakrou S, Abraham MR. Hypertrophic cardiomyopathy: a heart in need of an energy bar? Front Physiol 2014; 5:309. [PMID: 25191275 PMCID: PMC4137386 DOI: 10.3389/fphys.2014.00309] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/30/2014] [Indexed: 01/08/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) has been recently recognized as the most common inherited cardiovascular disorder, affecting 1 in 500 adults worldwide. HCM is characterized by myocyte hypertrophy resulting in thickening of the ventricular wall, myocyte disarray, interstitial and/or replacement fibrosis, decreased ventricular cavity volume and diastolic dysfunction. HCM is also the most common cause of sudden death in the young. A large proportion of patients diagnosed with HCM have mutations in sarcomeric proteins. However, it is unclear how these mutations lead to the cardiac phenotype, which is variable even in patients carrying the same causal mutation. Abnormalities in calcium cycling, oxidative stress, mitochondrial dysfunction and energetic deficiency have been described constituting the basis of therapies in experimental models of HCM and HCM patients. This review focuses on evidence supporting the role of cellular metabolism and mitochondria in HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Division of Cardiology, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - M Roselle Abraham
- Division of Cardiology, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
46
|
Esterhammer R, Klug G, Wolf C, Mayr A, Reinstadler S, Feistritzer HJ, Metzler B, Schocke MFH. Cardiac high-energy phosphate metabolism alters with age as studied in 196 healthy males with the help of 31-phosphorus 2-dimensional chemical shift imaging. PLoS One 2014; 9:e97368. [PMID: 24940736 PMCID: PMC4062408 DOI: 10.1371/journal.pone.0097368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/18/2014] [Indexed: 11/26/2022] Open
Abstract
Recently published studies have elucidated alterations of mitochondrial oxidative metabolism during ageing. The intention of the present study was to evaluate the impact of ageing on cardiac high-energy phosphate metabolism and cardiac function in healthy humans. 31-phosphorus 2-dimensional chemical shift imaging (31P 2D CSI) and echocardiography were performed in 196 healthy male volunteers divided into groups of 20 to 40 years (I, n = 43), 40 to 60 years (II, n = 123) and >60 years (III, n = 27) of age. Left ventricular PCr/β-ATP ratio, myocardial mass (MM), ejection fraction and E/A ratio were assessed. Mean PCr/β-ATP ratios were significantly different among the three groups of volunteers (I, 2.10±0.37; II, 1.77±0.37; III, 1.45±0.28; all p<0.001). PCr/β-ATP ratios were inversely related to age (r2 = −0.25; p<0.001) with a decrease from 2.65 by 0.02 per year of ageing. PCr/β-ATP ratios further correlated with MM (r = −0.371; p<0.001) and E/A ratios (r = 0.213; p<0.02). Moreover, E/A ratios (r = −0.502, p<0.001), MM (r = 0.304, p<0.001), glucose-levels (r = 0.157, p<0.05) and systolic blood pressure (r = 0.224, p<0.005) showed significant correlations with age. The ejection fraction did not significantly differ between the groups. This study shows that cardiac PCr/β-ATP ratios decrease moderately with age indicating an impairment of mitochondrial oxidative metabolism due to age. Furthermore, MM increases, and E/A ratio decreases with age. Both correlate with left-ventricular PCr/β-ATP ratios. The findings of the present study confirm numerous experimental studies showing an impairment of cardiac mitochondrial function with age.
Collapse
Affiliation(s)
- Regina Esterhammer
- Department of Radiology, Medical University Innsbruck, Innsbruck, Austria
| | - Gert Klug
- Department of Internal Medicine III, Division of Cardiology, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Wolf
- Department of Radiology, Medical University Innsbruck, Innsbruck, Austria
- Department of Radiology, District Hospital Reutte, Ehenbichl, Austria
| | - Agnes Mayr
- Department of Radiology, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastian Reinstadler
- Department of Internal Medicine III, Division of Cardiology, Medical University Innsbruck, Innsbruck, Austria
| | - Hans-Josef Feistritzer
- Department of Internal Medicine III, Division of Cardiology, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard Metzler
- Department of Internal Medicine III, Division of Cardiology, Medical University Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
47
|
Ingwall JS. The energetic cost of contraction is higher in the myocardium of patients with hypertrophic cardiomyopathy. Cardiovasc Res 2014; 103:192-3. [PMID: 24935429 DOI: 10.1093/cvr/cvu145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Joanne S Ingwall
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Witjas-Paalberends ER, Güçlü A, Germans T, Knaapen P, Harms HJ, Vermeer AMC, Christiaans I, Wilde AAM, Dos Remedios C, Lammertsma AA, van Rossum AC, Stienen GJM, van Slegtenhorst M, Schinkel AF, Michels M, Ho CY, Poggesi C, van der Velden J. Gene-specific increase in the energetic cost of contraction in hypertrophic cardiomyopathy caused by thick filament mutations. Cardiovasc Res 2014; 103:248-57. [PMID: 24835277 DOI: 10.1093/cvr/cvu127] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Disease mechanisms regarding hypertrophic cardiomyopathy (HCM) are largely unknown and disease onset varies. Sarcomere mutations might induce energy depletion for which until now there is no direct evidence at sarcomere level in human HCM. This study investigated if mutations in genes encoding myosin-binding protein C (MYBPC3) and myosin heavy chain (MYH7) underlie changes in the energetic cost of contraction in the development of human HCM disease. METHODS AND RESULTS Energetic cost of contraction was studied in vitro by measurements of force development and ATPase activity in cardiac muscle strips from 26 manifest HCM patients (11 MYBPC3mut, 9 MYH7mut, and 6 sarcomere mutation-negative, HCMsmn). In addition, in vivo, the ratio between external work (EW) and myocardial oxygen consumption (MVO2) to obtain myocardial external efficiency (MEE) was determined in 28 pre-hypertrophic mutation carriers (14 MYBPC3mut and 14 MYH7mut) and 14 healthy controls using [(11)C]-acetate positron emission tomography and cardiovascular magnetic resonance imaging. Tension cost (TC), i.e. ATPase activity during force development, was higher in MYBPC3mut and MYH7mut compared with HCMsmn at saturating [Ca(2+)]. TC was also significantly higher in MYH7mut at submaximal, more physiological [Ca(2+)]. EW was significantly lower in both mutation carrier groups, while MVO2 did not differ. MEE was significantly lower in both mutation carrier groups compared with controls, showing the lowest efficiency in MYH7 mutation carriers. CONCLUSION We provide direct evidence that sarcomere mutations perturb the energetic cost of cardiac contraction. Gene-specific severity of cardiac abnormalities may underlie differences in disease onset and suggests that early initiation of metabolic treatment may be beneficial, in particular, in MYH7 mutation carriers.
Collapse
Affiliation(s)
- E Rosalie Witjas-Paalberends
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Ahmet Güçlü
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Paul Knaapen
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Hendrik J Harms
- Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Alexa M C Vermeer
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Imke Christiaans
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Arthur A M Wilde
- Department of Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Cris Dos Remedios
- Institute for Biomedical Research, Muscle Research Unit, University of Sydney, Sydney, Australia
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Albert C van Rossum
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ger J M Stienen
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands Department of Physics and Astronomy, VU University, Amsterdam, The Netherlands
| | | | - Arend F Schinkel
- Thorax Center, Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michelle Michels
- Thorax Center, Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carolyn Y Ho
- Brigham and Women's Hospital, Cardiology, Boston, MA, USA
| | - Corrado Poggesi
- Department of Physiology, University of Florence, Florence, Italy
| | - Jolanda van der Velden
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
49
|
Poggesi C, Ho CY. Muscle dysfunction in hypertrophic cardiomyopathy: what is needed to move to translation? J Muscle Res Cell Motil 2014; 35:37-45. [PMID: 24493262 DOI: 10.1007/s10974-014-9374-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/02/2014] [Indexed: 02/04/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere genes. As such, HCM provides remarkable opportunities to study how changes to the heart's molecular motor apparatus may influence cardiac structure and function. Although the genetic basis of HCM is well-described, there is much more limited understanding of the precise consequences of sarcomere mutations--how they remodel the heart, and how these changes lead to the dramatic clinical consequences associated with HCM. More precise characterization of the mechanisms leading from sarcomere mutation to altered cardiac muscle function is critical to gain insight into fundamental disease biology and phenotypic evolution. Such knowledge will help foster development of novel treatment strategies aimed at correcting and preventing disease development in HCM.
Collapse
Affiliation(s)
- Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 63, 50134, Florence, Italy,
| | | |
Collapse
|
50
|
Abraham MR, Bottomley PA, Dimaano VL, Pinheiro A, Steinberg A, Traill TA, Abraham TP, Weiss RG. Creatine kinase adenosine triphosphate and phosphocreatine energy supply in a single kindred of patients with hypertrophic cardiomyopathy. Am J Cardiol 2013; 112:861-6. [PMID: 23751935 DOI: 10.1016/j.amjcard.2013.05.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/02/2013] [Accepted: 05/02/2013] [Indexed: 01/20/2023]
Abstract
A lethal and extensively characterized familial form of hypertrophic cardiomyopathy (HC) is due to a point mutation (Arg403Gln) in the cardiac β-myosin heavy chain gene. Although this is associated with abnormal energy metabolism and progression to heart failure in an animal model, in vivo cardiac energetics have not been characterized in patients with this mutation. Noninvasive phosphorus saturation transfer magnetic resonance spectroscopy was used to measure the adenosine triphosphate supplied by the creatine kinase (CK) reaction and phosphocreatine, the heart's primary energy reserve, in 9 of 10 patients from a single kindred with HC caused by the Arg403GIn mutation and 17 age-matched healthy controls. Systolic and diastolic function was assessed by echocardiography in all 10 patients with HC. The patients with HC had impairment of diastolic function and mild systolic dysfunction, when assessed using global systolic longitudinal strain. Myocardial phosphocreatine was significantly decreased by 24% in patients (7.1 ± 2.3 μmol/g) compared with the controls (9.4 ± 1.2 μmol/g; p = 0.003). The pseudo-first-order CK rate-constant was 26% lower (0.28 ± 0.15 vs 0.38 ± 0.07 s⁻¹, p = 0.035) and the forward CK flux was 44% lower (2.0 ± 1.4 vs 3.6 ± 0.9 μmol/g/s, p = 0.001) than in the controls. The contractile abnormalities did not correlate with the metabolic indexes. In conclusion, myocardial phosphocreatine and CK-ATP delivery are significantly reduced in patients with HC caused by the Arg403Gln mutation, akin to previous results from mice with the same mutation. A lack of a relation between energetic and contractile abnormalities suggests the former result from the sarcomeric mutation and not a late consequence of mechanical dysfunction.
Collapse
|