1
|
Carabetta N, Siracusa C, Leo I, Panuccio G, Strangio A, Sabatino J, Torella D, De Rosa S. Cardiomyopathies: The Role of Non-Coding RNAs. Noncoding RNA 2024; 10:53. [PMID: 39449507 PMCID: PMC11503404 DOI: 10.3390/ncrna10060053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024] Open
Abstract
Cardiomyopathies are the structural and functional disorders of the myocardium. Etiopathogenesis is complex and involves an interplay of genetic, environmental, and lifestyle factors eventually leading to myocardial abnormalities. It is known that non-coding (Nc) RNAs, including micro (mi)-RNAs and long non-coding (lnc) RNAs, play a crucial role in regulating gene expression. Several studies have explored the role of miRNAs in the development of various pathologies, including heart diseases. In this review, we analyzed various patterns of ncRNAs expressed in the most common cardiomyopathies: dilated cardiomyopathy, hypertrophic cardiomyopathy and arrhythmogenic cardiomyopathy. Understanding the role of different ncRNAs implicated in cardiomyopathic processes may contribute to the identification of potential therapeutic targets and novel risk stratification models based on gene expression. The analysis of ncRNAs may also be helpful to unveil the molecular mechanisms subtended to these diseases.
Collapse
Affiliation(s)
- Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.C.); (C.S.)
| | - Chiara Siracusa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.C.); (C.S.)
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Giuseppe Panuccio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200 Berlin, Germany
| | - Antonio Strangio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.C.); (C.S.)
| |
Collapse
|
2
|
Subramanian S, Ponce-Rivera MS, Affolter J, Shmorhun D, Owens R, Fraser CD, Glass L. Accelerated Wean: A Novel 3 Day Berlin Heart Protocol in a Pediatric Patient. ASAIO J 2024:00002480-990000000-00564. [PMID: 39321094 DOI: 10.1097/mat.0000000000002318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
We present the case of a 4 year old child who developed cardiogenic shock due to permanent junctional reciprocating tachycardia, requiring left ventricular assist device (LVAD) support. Despite the initial critical clinical presentation, successful myocardial recovery was achieved over 5 months, allowing for successful LVAD explantation. The patient's young age and behavioral issues were constraining factors for a prolonged LVAD wean trial. A modified wean protocol over a 3 day period with parameters for assessment of cardiac recovery before LVAD explantation is described.
Collapse
Affiliation(s)
- Sujata Subramanian
- From the Dell Children's Medical Center, Austin, Texas
- University of Texas at Austin Dell Medical School
| | - Monica S Ponce-Rivera
- University of Texas at Austin Dell Medical School
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrery, Nuevo Leon, Mexico
| | - Jeremy Affolter
- From the Dell Children's Medical Center, Austin, Texas
- University of Texas at Austin Dell Medical School
| | - Daniel Shmorhun
- From the Dell Children's Medical Center, Austin, Texas
- University of Texas at Austin Dell Medical School
| | - Richard Owens
- From the Dell Children's Medical Center, Austin, Texas
- University of Texas at Austin Dell Medical School
| | - Charles D Fraser
- From the Dell Children's Medical Center, Austin, Texas
- University of Texas at Austin Dell Medical School
| | - Lauren Glass
- From the Dell Children's Medical Center, Austin, Texas
- University of Texas at Austin Dell Medical School
| |
Collapse
|
3
|
Pensa AV, Zheng V, Davis L, Harap RW, Wilcox JE. Clinical Perspective of Myocardial Recovery and Improvement: Definitions, Prevalence, and Relevance. Methodist Debakey Cardiovasc J 2024; 20:6-15. [PMID: 39184164 PMCID: PMC11342833 DOI: 10.14797/mdcvj.1441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 08/27/2024] Open
Abstract
Partial or complete imaging resolution of left ventricular (LV) systolic dysfunction in patients with heart failure with reduced ejection fraction (HFrEF) has gone by many names in the past few decades, including LV recovery, remission, reverse remodeling, and, most recently, improvement. This phenomenon has been described in a variety of clinical scenarios, including removal of an acute myocardial insult, unloading with durable LV assist devices, and treatment with various devices as well as pharmacotherapies, termed guideline-directed medical therapy (GDMT). Irrespective of definition, systolic improvement is associated with improved clinical outcomes compared to persistent systolic dysfunction. In the past few years, systolic improvement has been distinguished from HFrEF as a new clinical entity referred to as HF with improved EF (HFimpEF). Given the relative novelty of this condition, there is a paucity of data with regard to the clinical trajectory and management of this population. In this review, we describe the history of myocardial improvement terminology and explore notable findings that have led to the delineation of HFimpEF. Additionally, we highlight the importance of understanding LV trajectory and the potential opportunity for new GDMT management for clinicians when treating patients with HFimpEF.
Collapse
Affiliation(s)
- Anthony V. Pensa
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, US
| | - Veronica Zheng
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, US
| | - Lucia Davis
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, US
| | - Rebecca W. Harap
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, US
| | - Jane E. Wilcox
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, US
| |
Collapse
|
4
|
Trimarchi G, Teresi L, Licordari R, Pingitore A, Pizzino F, Grimaldi P, Calabrò D, Liotta P, Micari A, de Gregorio C, Di Bella G. Transient Left Ventricular Dysfunction from Cardiomyopathies to Myocardial Viability: When and Why Cardiac Function Recovers. Biomedicines 2024; 12:1051. [PMID: 38791012 PMCID: PMC11117605 DOI: 10.3390/biomedicines12051051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Transient left ventricular dysfunction (TLVD), a temporary condition marked by reversible impairment of ventricular function, remains an underdiagnosed yet significant contributor to morbidity and mortality in clinical practice. Unlike the well-explored atherosclerotic disease of the epicardial coronary arteries, the diverse etiologies of TLVD require greater attention for proper diagnosis and management. The spectrum of disorders associated with TLVD includes stress-induced cardiomyopathy, central nervous system injuries, histaminergic syndromes, various inflammatory diseases, pregnancy-related conditions, and genetically determined syndromes. Furthermore, myocardial infarction with non-obstructive coronary arteries (MINOCA) origins such as coronary artery spasm, coronary thromboembolism, and spontaneous coronary artery dissection (SCAD) may also manifest as TLVD, eventually showing recovery. This review highlights the range of ischemic and non-ischemic clinical situations that lead to TLVD, gathering conditions like Tako-Tsubo Syndrome (TTS), Kounis syndrome (KS), Myocarditis, Peripartum Cardiomyopathy (PPCM), and Tachycardia-induced cardiomyopathy (TIC). Differentiation amongst these causes is crucial, as they involve distinct clinical, instrumental, and genetic predictors that bode different outcomes and recovery potential for left ventricular function. The purpose of this review is to improve everyday clinical approaches to treating these diseases by providing an extensive survey of conditions linked with TLVD and the elements impacting prognosis and outcomes.
Collapse
Affiliation(s)
- Giancarlo Trimarchi
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| | - Lucio Teresi
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| | - Roberto Licordari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98100 Messina, Italy; (R.L.); (A.M.)
| | - Alessandro Pingitore
- Istituto di Fisiologia Clinica, Clinical Physiology Institute, CNR, 56124 Pisa, Italy;
| | - Fausto Pizzino
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy;
| | - Patrizia Grimaldi
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| | - Danila Calabrò
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| | - Paolo Liotta
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| | - Antonio Micari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98100 Messina, Italy; (R.L.); (A.M.)
| | - Cesare de Gregorio
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, 98100 Messina, Italy; (L.T.); (P.G.); (D.C.); (P.L.); (C.d.G.); (G.D.B.)
| |
Collapse
|
5
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 182] [Impact Index Per Article: 182.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
6
|
Romero E, Baltodano AF, Rocha P, Sellers-Porter C, Patel DJ, Soroya S, Bidwell J, Ebong I, Gibson M, Liem DA, Jimenez S, Bang H, Sirish P, Chiamvimonvat N, Lopez JE, Cadeiras M. Clinical, Echocardiographic, and Longitudinal Characteristics Associated With Heart Failure With Improved Ejection Fraction. Am J Cardiol 2024; 211:143-152. [PMID: 37923155 PMCID: PMC10869234 DOI: 10.1016/j.amjcard.2023.10.086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023]
Abstract
Heart failure with improved ejection fraction (HFimpEF) has better outcomes than HF with reduced EF (HFrEF). However, factors contributing to HFimpEF remain unclear. This study aimed to evaluate clinical and longitudinal characteristics associated with subsequent HFimpEF. This was a single-center retrospective HFrEF cohort study. Data were collected from 2014 to 2022. Patients with HFrEF were identified using International Classification of Diseases codes, echocardiographic data, and natriuretic peptide levels. The main end points were HFimpEF (defined as EF >40% at ≥3 months with ≥10% increase) and mortality. Cox proportional hazards and mixed effects models were used for analyses. The study included 1,307 patients with HFrEF with a median follow-up of 16.3 months (interquartile range 8.0 to 30.6). The median age was 65 years; 68% were male whereas 57% were White. On follow-up, 38.7% (n = 506) developed HFimpEF, whereas 61.3% (n = 801) had persistent HFrEF. A multivariate Cox regression model identified gender, race, co-morbidities, echocardiographic, and natriuretic peptide as significant covariates of HFimpEF (p <0.05). The HFimpEF group had better survival compared with the persistent HFrEF group (p <0.001). Echocardiographic and laboratory trajectories differed between groups. In this HFrEF cohort, 38.7% transitioned to HFimpEF and approximately 50% met the definition within the first 12 months. In a HFimpEF model, gender, co-morbidities, echocardiographic parameters, and natriuretic peptide were associated with subsequent HFimpEF. The model has the potential to identify patients at risk of subsequent persistent or improved HFrEF, thus informing the design and implementation of targeted quality-of-care improvement interventions.
Collapse
Affiliation(s)
- Erick Romero
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California.
| | | | - Paulo Rocha
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Camryn Sellers-Porter
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Dev Jaydeep Patel
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Saad Soroya
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Julie Bidwell
- Betty Irene Moore School of Nursing, Family Caregiving Institute, University of California Davis, Sacramento, California
| | - Imo Ebong
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Michael Gibson
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - David A Liem
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Shirin Jimenez
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Heejung Bang
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, California
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California
| | - Javier E Lopez
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California
| | - Martin Cadeiras
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, California.
| |
Collapse
|
7
|
Pensa AV, Khan SS, Shah RV, Wilcox JE. Heart failure with improved ejection fraction: Beyond diagnosis to trajectory analysis. Prog Cardiovasc Dis 2024; 82:102-112. [PMID: 38244827 DOI: 10.1016/j.pcad.2024.01.014] [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: 01/14/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
Left ventricular (LV) systolic dysfunction represents a highly treatable cause of heart failure (HF). A substantial proportion of patients with HF with reduced ejection fraction (EF;HFrEF) demonstrate improvement in LV systolic function (termed HF with improved EF [HFimpEF]), either spontaneously or when treated with guideline-directed medical therapy (GDMT). Although it is a relatively new HF classification, HFimpEF has emerged in recent years as an important and distinct clinical entity. Improvement in LVEF leads to decreased rates of mortality and adverse HF-related outcomes compared to patients with sustained LV systolic dysfunction (HFrEF). While numerous clinical and imaging factors have been associated with HFimpEF, identification of which patients do and do not improve requires further investigation. In addition, patients improve at different rates, and what determines the trajectory of HFimpEF patients after improvement is incompletely characterized. A proportion of patients maintain improvement in LV systolic function, while others experience a recrudescence of systolic dysfunction, especially with GDMT discontinuation. In this review we discuss the contemporary guideline-recommended classification definition of HFimpEF, the epidemiology of improvement in LV systolic function, and the clinical course of this unique patient population. We also offer evidence-based recommendations for the clinical management of HFimpEF and provide a roadmap for future directions in understanding and improving outcomes in the care of patients with HFimpEF.
Collapse
Affiliation(s)
- Anthony V Pensa
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Sadiya S Khan
- Department of Medicine, Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Ravi V Shah
- Department of Medicine, Division of Cardiology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Jane E Wilcox
- Department of Medicine, Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America.
| |
Collapse
|
8
|
Romero E, Baltodano AF, Rocha P, Sellers-Porter C, Patel DJ, Soroya S, Bidwell J, Ebong I, Gibson M, Liem DA, Jimenez S, Bang H, Sirish P, Chiamvimonvat N, Lopez JE, Cadeiras M. Clinical, Echocardiographic, and Longitudinal Characteristics Associated with Heart Failure with Improved Ejection Fraction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.25.23294644. [PMID: 37693424 PMCID: PMC10491272 DOI: 10.1101/2023.08.25.23294644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Background Heart failure (HF) with improved ejection fraction (HFimpEF) has better outcomes than HF with reduced ejection fraction (HFrEF). However, factors contributing to HFimpEF remain unclear. This study aimed to evaluate clinical and longitudinal characteristics associated with subsequent HFimpEF. Methods This was a single-center retrospective HFrEF cohort study. Data were collected from 2014 to 2022. Patients with HFrEF were identified using ICD codes, echocardiographic data, and natriuretic peptide levels. The main endpoints were HFimpEF (defined as ejection fraction >40% at ≥3 months with ≥10% increase) and mortality. Cox proportional hazards and mixed effects models were used for analyses. Results The study included 1307 HFrEF patients with a median follow-up of 16.3 months (IQR 8.0-30.6). The median age was 65 years; 68% were male while 57% were white. On follow-up, 39% (n=506) developed HFimpEF, while 61% (n=801) had persistent HFrEF. A multivariate Cox regression model identified sex, race comorbidities, echocardiographic, and natriuretic peptide as significant covariates of HFimpEF ( p <0.05). The HFimpEF group had better survival compared to the persistent HFrEF group ( p <0.001). Echocardiographic and laboratory trajectories differed between groups. Conclusion In this HFrEF cohort, 39% transitioned to HFimpEF and approximately 50% met the definition within the first 12 months. In a HFimpEF model, sex, comorbidities, echocardiographic parameters, and natriuretic peptide were associated with subsequent HFimpEF. The model has the potential to identify patients at risk of subsequent persistent or improved HFrEF, thus informing the design and implementation of targeted quality-of-care improvement interventions.
Collapse
|
9
|
Liu D, Hu K, Schregelmann L, Hammel C, Lengenfelder BD, Ertl G, Frantz S, Nordbeck P. Determinants of ejection fraction improvement in heart failure patients with reduced ejection fraction. ESC Heart Fail 2023; 10:1358-1371. [PMID: 36732921 PMCID: PMC10053299 DOI: 10.1002/ehf2.14303] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/09/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
AIMS This study aimed to investigate the prognostic value of dynamic changes in left ventricular ejection fraction (EF) for cardiovascular (CV) outcomes in an all-comer heart failure (HF) population with reduced EF (HFrEF, EF < 40%). We sought to identify independent factors related to improvement in EF and to identify risk factors for increased risk of CV events in the subgroups of improved EF (iEF) and non-improved EF (niEF), respecively. METHODS AND RESULTS This is a retrospective sub-analysis from the REDEAL HF trial, which included consecutive patients with chronic HF who were hospitalized from July 2009 to December 2017. Baseline and follow-up echocardiography data (interval ≥12 months) of 573 consecutive patients with HFrEF were analysed. iEF was defined as absolute improvement in EF ≥ 10% and follow-up EF over 40%. The primary endpoint was defined as a composite endpoint of cardiovascular (CV) death, CV hospitalization, or appropriate implantable cardioverter-defibrillator (ICD) therapy for ventricular arrhythmia. EF improved in 37.2% of patients with HFrEF during follow-up (median period of 17 months). iEF was independently associated with shorter HF duration (>4 vs. ≤4 years, odd ratio [OR] = 0.477, 95% CI 0.305-0.745), no coronary artery disease (CAD vs. no CAD, OR = 0.583, 95% CI 0.396-0.858), and no ICD implantation (ICD vs. no ICD, OR = 0.341, 95% CI 0.228-0.511). Compared with niEF, iEF was significantly and independently associated with lower all-cause mortality (22.1% vs. 31.1%, P = 0.019; hazard ratio [HR] = 0.674, 95% CI 0.469-0.968), lower CV mortality (8.9% vs. 16.1%, P = 0.015; HR = 0.539, 95% CI 0.317-0.916), and lower CV events risk (27.2% vs. 49.2%, P < 0.001; HR 0.519, 95% CI 0.381-0.708), after adjustment for age, sex, duration of HF, and other clinical risk factors. Hypertension (HR = 2.452, P = 0.032) and elevated N-terminal prohormone of brain natriuretic peptide (NT-proBNP >1153 pg/mL, HR = 4.372, P < 0.001) were identified as independent risk factors for CV events in the iEF subgroup. ICD implantation (HR = 1.533, P = 0.011), elevated NT-proBNP (HR = 1.626, P = 0.018), increased left atrial volume index (HR = 1.461, P = 0.021), reduced lateral mitral annular plane systolic excursion (HR = 1.478, P = 0.025), and reduced tricuspid plane systolic excursion (HR = 1.491, P = 0.039) were identified as risk factors for CV events in the niEF subgroup. CONCLUSIONS Improvement in EF is independently related to the longer survival and lower CV related mortality and hospitalization rate of HFrEF. Elevated baseline NT-proBNP is identified as the strongest prognostic factor associated with increased CV events risk in HFrEF patients both with and without improved EF, regardless of age, sex, duration of HF, and other clinical risk factors.
Collapse
Affiliation(s)
- Dan Liu
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Kai Hu
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Lena Schregelmann
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Clara Hammel
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Björn Daniel Lengenfelder
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Georg Ertl
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Stefan Frantz
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| | - Peter Nordbeck
- Department of Internal Medicine IUniversity Hospital WürzburgWürzburgGermany
- Comprehensive Heart Failure CenterWürzburgGermany
| |
Collapse
|
10
|
Amrute JM, Lai L, Ma P, Koenig AL, Kamimoto K, Bredemeyer A, Shankar TS, Kuppe C, Kadyrov FF, Schulte LJ, Stoutenburg D, Kopecky BJ, Navankasattusas S, Visker J, Morris SA, Kramann R, Leuschner F, Mann DL, Drakos SG, Lavine KJ. Defining cardiac functional recovery in end-stage heart failure at single-cell resolution. NATURE CARDIOVASCULAR RESEARCH 2023; 2:399-416. [PMID: 37583573 PMCID: PMC10426763 DOI: 10.1038/s44161-023-00260-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/01/2023] [Indexed: 08/17/2023]
Abstract
Recovery of cardiac function is the holy grail of heart failure therapy yet is infrequently observed and remains poorly understood. In this study, we performed single-nucleus RNA sequencing from patients with heart failure who recovered left ventricular systolic function after left ventricular assist device implantation, patients who did not recover and non-diseased donors. We identified cell-specific transcriptional signatures of recovery, most prominently in macrophages and fibroblasts. Within these cell types, inflammatory signatures were negative predictors of recovery, and downregulation of RUNX1 was associated with recovery. In silico perturbation of RUNX1 in macrophages and fibroblasts recapitulated the transcriptional state of recovery. Cardiac recovery mediated by BET inhibition in mice led to decreased macrophage and fibroblast Runx1 expression and diminished chromatin accessibility within a Runx1 intronic peak and acquisition of human recovery signatures. These findings suggest that cardiac recovery is a unique biological state and identify RUNX1 as a possible therapeutic target to facilitate cardiac recovery.
Collapse
Affiliation(s)
- Junedh M. Amrute
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- These authors contributed equally: Junedh M. Amrute, Lulu Lai
| | - Lulu Lai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- These authors contributed equally: Junedh M. Amrute, Lulu Lai
| | - Pan Ma
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew L. Koenig
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kenji Kamimoto
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Thirupura S. Shankar
- Division of Cardiovascular Medicine & Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health & School of Medicine, Salt Lake City, UT, USA
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology and Division of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Farid F. Kadyrov
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Linda J. Schulte
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Dylan Stoutenburg
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Benjamin J. Kopecky
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sutip Navankasattusas
- Division of Cardiovascular Medicine & Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health & School of Medicine, Salt Lake City, UT, USA
| | - Joseph Visker
- Division of Cardiovascular Medicine & Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health & School of Medicine, Salt Lake City, UT, USA
| | - Samantha A. Morris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology and Division of Nephrology, RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Florian Leuschner
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Douglas L. Mann
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Stavros G. Drakos
- Division of Cardiovascular Medicine & Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health & School of Medicine, Salt Lake City, UT, USA
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
11
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation 2023; 147:e93-e621. [PMID: 36695182 DOI: 10.1161/cir.0000000000001123] [Citation(s) in RCA: 1550] [Impact Index Per Article: 1550.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2023 Statistical Update is the product of a full year's worth of effort in 2022 by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. The American Heart Association strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional COVID-19 (coronavirus disease 2019) publications, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
12
|
Amrute JM, Luo X, Penna V, Bredemeyer A, Yamawaki T, Yang S, Kadyrov F, Heo GS, Shi SY, Lee P, Koenig AL, Kuppe C, Jones C, Kopecky B, Hayat S, Ma P, Terada Y, Fu A, Furtado M, Kreisel D, Stitziel NO, Li CM, Kramann R, Liu Y, Ason B, Lavine KJ. Targeting Immune-Fibroblast Crosstalk in Myocardial Infarction and Cardiac Fibrosis. RESEARCH SQUARE 2023:rs.3.rs-2402606. [PMID: 36747878 PMCID: PMC9900986 DOI: 10.21203/rs.3.rs-2402606/v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Inflammation and tissue fibrosis co-exist and are causally linked to organ dysfunction. However, the molecular mechanisms driving immune-fibroblast crosstalk in human cardiac disease remains unexplored and there are currently no therapeutics to target fibrosis. Here, we performed multi-omic single-cell gene expression, epitope mapping, and chromatin accessibility profiling in 38 donors, acutely infarcted, and chronically failing human hearts. We identified a disease-associated fibroblast trajectory marked by cell surface expression of fibroblast activator protein (FAP), which diverged into distinct myofibroblasts and pro-fibrotic fibroblast populations, the latter resembling matrifibrocytes. Pro-fibrotic fibroblasts were transcriptionally similar to cancer associated fibroblasts and expressed high levels of collagens and periostin (POSTN), thymocyte differentiation antigen 1 (THY-1), and endothelin receptor A (EDNRA) predicted to be driven by a RUNX1 gene regulatory network. We assessed the applicability of experimental systems to model tissue fibrosis and demonstrated that 3 different in vivo mouse models of cardiac injury were superior compared to cultured human heart and dermal fibroblasts in recapitulating the human disease phenotype. Ligand-receptor analysis and spatial transcriptomics predicted that interactions between C-C chemokine receptor type 2 (CCR2) macrophages and fibroblasts mediated by interleukin 1 beta (IL-1β) signaling drove the emergence of pro-fibrotic fibroblasts within spatially defined niches. This concept was validated through in silico transcription factor perturbation and in vivo inhibition of IL-1β signaling in fibroblasts where we observed reduced pro-fibrotic fibroblasts, preferential differentiation of fibroblasts towards myofibroblasts, and reduced cardiac fibrosis. Herein, we show a subset of macrophages signal to fibroblasts via IL-1β and rewire their gene regulatory network and differentiation trajectory towards a pro-fibrotic fibroblast phenotype. These findings highlight the broader therapeutic potential of targeting inflammation to treat tissue fibrosis and restore organ function.
Collapse
Affiliation(s)
- Junedh M. Amrute
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Xin Luo
- Genome Analysis Unit, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Andrea Bredemeyer
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Tracy Yamawaki
- Genome Analysis Unit, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Farid Kadyrov
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Gyu-Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Sally Yu Shi
- Department of Cardiometabolic Disorders, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Paul Lee
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Andrew L. Koenig
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Department of Nephrology, RWTH Aachen, Medical Faculty, Aachen, Germany
| | - Cameran Jones
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Benjamin Kopecky
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Sikander Hayat
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Pan Ma
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Yuriko Terada
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Angela Fu
- Department of Cardiometabolic Disorders, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Milena Furtado
- Genome Analysis Unit, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nathan O. Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Chi-Ming Li
- Genome Analysis Unit, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Department of Nephrology, RWTH Aachen, Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation Erasmus Medical Center, Rotterdam, The Netherlands
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Brandon Ason
- Department of Cardiometabolic Disorders, Amgen Discovery Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| |
Collapse
|
13
|
Chen X, Wu M. Heart failure with recovered ejection fraction: Current understanding and future prospects. Am J Med Sci 2023; 365:1-8. [PMID: 36084706 DOI: 10.1016/j.amjms.2022.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/18/2022] [Accepted: 07/12/2022] [Indexed: 01/04/2023]
Abstract
Heart failure with reduced ejection fraction (HFrEF) is a prevalent kind of heart failure in which a significant amount of the ejection fraction can be repaired, and left ventricular remodeling and dysfunction can be reversed or even restored completely. However, a considerable number of patients still present clinical signs and biochemical features of incomplete recovery from the pathophysiology of heart failure and are at risk for adverse outcomes such as re-deterioration of systolic function and recurrence of HFrEF. Furthermore, it is revealed from a microscopic perspective that even if partial or complete reverse remodeling occurs, the morphological changes of cardiomyocytes, extracellular matrix deposition, and abnormal transcription and expression of pathological genes still exist. Patients with "recovered ejection fraction" have milder clinical symptoms and better outcomes than those with continued reduction of ejection fraction. Based on the unique characteristics of this subgroup and the existence of many unknowns, the academic community defines it as a new category-heart failure with recovered ejection fraction (HFrecEF). Because there is a shortage of natural history data for this population as well as high-quality clinical and basic research data, it is difficult to accurately evaluate clinical risk and manage this population. This review will present the current understanding of HFrecEF from the limited literature.
Collapse
Affiliation(s)
- Xi Chen
- Department of Cardiology, Affiliated Hospital of Putian University, Fujian, China
| | - Meifang Wu
- Department of Cardiology, Affiliated Hospital of Putian University, Fujian, China.
| |
Collapse
|
14
|
Quttainah M, Raveendran VV, Saleh S, Parhar R, Aljoufan M, Moorjani N, Al-Halees ZY, AlShahid M, Collison KS, Westaby S, Al-Mohanna F. Transcriptomal Insights of Heart Failure from Normality to Recovery. Biomolecules 2022; 12:biom12050731. [PMID: 35625658 PMCID: PMC9138767 DOI: 10.3390/biom12050731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Current management of heart failure (HF) is centred on modulating the progression of symptoms and severity of left ventricular dysfunction. However, specific understandings of genetic and molecular targets are needed for more precise treatments. To attain a clearer picture of this, we studied transcriptome changes in a chronic progressive HF model. Fifteen sheep (Ovis aries) underwent supracoronary aortic banding using an inflatable cuff. Controlled and progressive induction of pressure overload in the LV was monitored by echocardiography. Endomyocardial biopsies were collected throughout the development of LV failure (LVF) and during the stage of recovery. RNA-seq data were analysed using the PANTHER database, Metascape, and DisGeNET to annotate the gene expression for functional ontologies. Echocardiography revealed distinct clinical differences between the progressive stages of hypertrophy, dilatation, and failure. A unique set of transcript expressions in each stage was identified, despite an overlap of gene expression. The removal of pressure overload allowed the LV to recover functionally. Compared to the control stage, there were a total of 256 genes significantly changed in their expression in failure, 210 genes in hypertrophy, and 73 genes in dilatation. Gene expression in the recovery stage was comparable with the control stage with a well-noted improvement in LV function. RNA-seq revealed the expression of genes in each stage that are not reported in cardiovascular pathology. We identified genes that may be potentially involved in the aetiology of progressive stages of HF, and that may provide future targets for its management.
Collapse
Affiliation(s)
- Mohammed Quttainah
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.Q.); (V.V.R.); (S.S.); (R.P.); (K.S.C.)
| | - Vineesh Vimala Raveendran
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.Q.); (V.V.R.); (S.S.); (R.P.); (K.S.C.)
| | - Soad Saleh
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.Q.); (V.V.R.); (S.S.); (R.P.); (K.S.C.)
| | - Ranjit Parhar
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.Q.); (V.V.R.); (S.S.); (R.P.); (K.S.C.)
| | - Mansour Aljoufan
- Heart Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.A.); (Z.Y.A.-H.); (M.A.)
| | - Narain Moorjani
- Department of Cardiothoracic Surgery, Papworth Hospital, University of Cambridge, Cambridge CB23 3RE, UK;
| | - Zohair Y. Al-Halees
- Heart Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.A.); (Z.Y.A.-H.); (M.A.)
| | - Maie AlShahid
- Heart Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.A.); (Z.Y.A.-H.); (M.A.)
| | - Kate S. Collison
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.Q.); (V.V.R.); (S.S.); (R.P.); (K.S.C.)
| | - Stephen Westaby
- Oxford Heart Centre, John Radcliffe Hospital, Oxford OX9 3DU, UK;
| | - Futwan Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (M.Q.); (V.V.R.); (S.S.); (R.P.); (K.S.C.)
- Correspondence:
| |
Collapse
|
15
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 2700] [Impact Index Per Article: 1350.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
16
|
Terzić D, Mikić A. Significance of right ventricular function for the outcome of treatment and remodeling of the heart after left ventricular assist device implantation. MEDICINSKI PODMLADAK 2022. [DOI: 10.5937/mp73-38192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
The efficiency of the device for permanent circulatory support of the left ventricle has been proven through clinical practice with the trend of constant improvement of treatment results along with biotechnological progress and improvement of surgical implantation techniques. The published reports of most reference cardiac surgery centers present a one-year survival rate of over 85%, a two-year survival rate of 70% and a five-year survival rate of 45-50%. In addition to clear benefits for the patient, implantation of LVAD also carries significant specific risks, so infections, post-implantation bleeding, strokes, and right ventricular postimplantation weakness are the most common complications. Given that the progress of the LVAD program is ensured primarily by reducing the incidence of complications not related to the functioning of individual segments of the cardiovascular system, and as left ventricular function is completely replaced by LVAD device, the most recent challenge is the decision to install LVAD device in the heart with right ventricular, given that the postimplantation weakness of right ventricular is associated with proven increased mortality and morbidity. Since the 1990s, studies on hearts with implanted LVAD as a bridge to heart transplantation have shown regression of cell hypertrophy, normalization of cell size, muscle fiber architecture, and heart chamber geometry. The described changes are characterized by the notion of reverse remodeling, which is synonymous with function recovery. It is this process at the level of the right ventricle that is recognized as extremely important for the success of LVAD programs, especially in the group of patients who have a certain degree of right ventricular weakness preoperatively. The basic requirements of the cardiac surgery team are adequate preoperative assessment of right ventricular weakness, then application of measures to prevent damage and load on the right ventricle during and after LVAD implantation, as well as providing adequate therapeutic measures for right ventricular recovery in the postimplantation period.
Collapse
|
17
|
He Y, Ling Y, Guo W, Li Q, Yu S, Huang H, Zhang R, Gong Z, Liu J, Mo L, Yi S, Lai D, Yao Y, Liu J, Chen J, Liu Y, Chen S. Prevalence and Prognosis of HFimpEF Developed From Patients With Heart Failure With Reduced Ejection Fraction: Systematic Review and Meta-Analysis. Front Cardiovasc Med 2021; 8:757596. [PMID: 34901217 PMCID: PMC8655693 DOI: 10.3389/fcvm.2021.757596] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/15/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Heart failure with improved ejection fraction (HFimpEF) is classified as a new type of heart failure, and its prevalence and prognosis are not consistent in previous studies. There is no systematic review and meta-analysis regarding the prevalence and prognosis of the HFimpEF. Method: A systematic search was performed in MEDLINE, EMBASE, and Cochrane Library from inception to May 22, 2021 (PROSPERO registration: CRD42021260422). Studies were included for analysis if the prognosis of mortality or hospitalization were reported in HFimpEF or in patients with heart failure with recovered ejection fraction (HFrecEF). The primary outcome was all-cause mortality. Cardiac hospitalization, all-cause hospitalization, and composite events of mortality and hospitalization were considered as secondary outcomes. Result: Nine studies consisting of 9,491 heart failure patients were eventually included. During an average follow-up of 3.8 years, the pooled prevalence of HFimpEF was 22.64%. HFimpEF had a lower risk of mortality compared with heart failure patients with reduced ejection fraction (HFrEF) (adjusted HR: 0.44, 95% CI: 0.33-0.60). HFimpEF was also associated with a lower risk of cardiac hospitalization (HR: 0.40, 95% CI: 0.20-0.82) and the composite endpoint of mortality and hospitalization (HR: 0.56, 95% CI: 0.44-0.73). Compared with patients with preserved ejection fraction (HFpEF), HFimpEF was associated with a moderately lower risk of mortality (HR: 0.42, 95% CI: 0.32-0.55) and hospitalization (HR: 0.73, 95% CI: 0.58-0.92). Conclusion: Around 22.64% of patients with HFrEF would be treated to become HFimpEF, who would then obtain a 56% decrease in mortality risk. Meanwhile, HFimpEF is associated with lower heart failure hospitalization. Further studies are required to explore how to promote left ventricular ejection fraction improvement and improve the prognosis of persistent HFrEF in patients. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021260422, identifier: CRD42021260422.
Collapse
Affiliation(s)
- Yibo He
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yihang Ling
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wei Guo
- Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiang Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sijia Yu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haozhang Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Rongting Zhang
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Zhiwen Gong
- Department of Cardiology, First People's Hospital of Kashgar Prefecture, Kashgar, China
- Department of Cardiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jiaxuan Liu
- Department of Cardiology, First People's Hospital of Kashgar Prefecture, Kashgar, China
- Department of Cardiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Liyi Mo
- Department of Cardiology, First People's Hospital of Kashgar Prefecture, Kashgar, China
- Department of Cardiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shixin Yi
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Disheng Lai
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Younan Yao
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yong Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shiqun Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
18
|
Zhang R, Zhong Y, Long SY, Yang QN, Zhou B, Rao L. Association between CDK8 gene polymorphisms and dilated cardiomyopathy in a Chinese Han population. Cardiovasc Diagn Ther 2021; 11:1036-1046. [PMID: 34815954 DOI: 10.21037/cdt-21-323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/30/2021] [Indexed: 02/05/2023]
Abstract
Background Dilated cardiomyopathy (DCM) is one of the most common types of cardiomyopathies. Various genes have been verified to be related to DCM, but the pathogenesis remains unclear. Cyclin-dependent-kinase 8 (CDK8), encoded by the CDK8 gene, is a transcriptional factor that regulates the phosphorylation of RNA polymerase II. It plays an important role in the transcription process and different signaling pathways. This study aimed to investigate the potential role of CDK8 gene polymorphisms in DCM susceptibility and prognosis in a Chinese Han population. Methods Two single nucleotide polymorphisms (SNPs) of CDK8, rs17083838 (A/G) and rs7992670 (A/G), were genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) in 341 DCM patients and 381 healthy controls. Survival analysis was performed using Kaplan-Meier curves and Cox regression analysis. Results The frequencies of allele A of both SNPs rs17083838 and rs7992670 were increased in DCM patients compared to healthy controls (P<0.05). Genotypic frequencies of rs17083838 and rs7992670 were associated with the susceptibility to DCM in the codominant, and recessive models (P<0.05), and AA/AG genotypes of rs17083838 were also related to DCM susceptibility in the dominant model. AA/AG genotypes of rs17083838 and the AA genotype of rs7992670 in the dominant and recessive genetic models presented a correlation with the poor prognosis of DCM patients in both univariate (P<0.05) and multivariate analyses (P<0.05) after adjusting for age, gender, left ventricular end-diastolic diameter (LVEDD), and left ventricular ejection fraction (LVEF). Conclusions This research is the first to reveal that CDK8 gene polymorphisms might be related to DCM susceptibility and prognosis in the Chinese Han population.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yue Zhong
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Si-Yu Long
- Department of Immunology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China
| | - Qin-Ni Yang
- Department of Immunology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China
| | - Bin Zhou
- Laboratory of Molecular Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Center of Translational Medicine, Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Li Rao
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Moura B, Aimo A, Al-Mohammad A, Flammer A, Barberis V, Bayes-Genis A, Brunner-La Rocca HP, Fontes-Carvalho R, Grapsa J, Hülsmann M, Ibrahim N, Knackstedt C, Januzzi JL, Lapinskas T, Sarrias A, Matskeplishvili S, Meijers WC, Messroghli D, Mueller C, Pavo N, Simonavičius J, Teske AJ, van Kimmenade R, Seferovic P, Coats AJS, Emdin M, Richards AM. Integration of imaging and circulating biomarkers in heart failure: a consensus document by the Biomarkers and Imaging Study Groups of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2021; 23:1577-1596. [PMID: 34482622 DOI: 10.1002/ejhf.2339] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/28/2021] [Accepted: 08/29/2021] [Indexed: 12/28/2022] Open
Abstract
Circulating biomarkers and imaging techniques provide independent and complementary information to guide management of heart failure (HF). This consensus document by the Heart Failure Association (HFA) of the European Society of Cardiology (ESC) presents current evidence-based indications relevant to integration of imaging techniques and biomarkers in HF. The document first focuses on application of circulating biomarkers together with imaging findings, in the broad domains of screening, diagnosis, risk stratification, guidance of treatment and monitoring, and then discusses specific challenging settings. In each section we crystallize clinically relevant recommendations and identify directions for future research. The target readership of this document includes cardiologists, internal medicine specialists and other clinicians dealing with HF patients.
Collapse
Affiliation(s)
- Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiology Department, Porto Armed Forces Hospital, Porto, Portugal
| | - Alberto Aimo
- Scuola Superiore Sant'Anna, and Fondazione G. Monasterio, Pisa, Italy
| | - Abdallah Al-Mohammad
- Medical School, University of Sheffield and Sheffield Teaching Hospitals, Sheffield, UK
| | | | | | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Hans-Peter Brunner-La Rocca
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ricardo Fontes-Carvalho
- Cardiovascular Research and Development Unit (UnIC), Faculty of Medicine University of Porto, Porto, Portugal.,Cardiology Department, Centro Hospitalar de Vila Nova Gaia/Espinho, Espinho, Portugal
| | - Julia Grapsa
- Department of Cardiology, Guys and St Thomas NHS Hospitals Trust, London, UK
| | - Martin Hülsmann
- Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nasrien Ibrahim
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christian Knackstedt
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomas Lapinskas
- Department of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Axel Sarrias
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | | | - Daniel Messroghli
- Department of Internal Medicine-Cardiology, Deutsches Herzzentrum Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Mueller
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Noemi Pavo
- Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Justas Simonavičius
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania
| | - Arco J Teske
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roland van Kimmenade
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | | | - Michele Emdin
- Scuola Superiore Sant'Anna, and Fondazione G. Monasterio, Pisa, Italy
| | - A Mark Richards
- Christchurch Heart Institute, University of Otago, Dunedin, New Zealand.,Cardiovascular Research Institute, National University of Singapore, Singapore
| |
Collapse
|
20
|
Abstract
This review provides a comprehensive overview of the past 25+ years of research into the development of left ventricular assist device (LVAD) to improve clinical outcomes in patients with severe end-stage heart failure and basic insights gained into the biology of heart failure gleaned from studies of hearts and myocardium of patients undergoing LVAD support. Clinical aspects of contemporary LVAD therapy, including evolving device technology, overall mortality, and complications, are reviewed. We explain the hemodynamic effects of LVAD support and how these lead to ventricular unloading. This includes a detailed review of the structural, cellular, and molecular aspects of LVAD-associated reverse remodeling. Synergisms between LVAD support and medical therapies for heart failure related to reverse remodeling, remission, and recovery are discussed within the context of both clinical outcomes and fundamental effects on myocardial biology. The incidence, clinical implications and factors most likely to be associated with improved ventricular function and remission of the heart failure are reviewed. Finally, we discuss recognized impediments to achieving myocardial recovery in the vast majority of LVAD-supported hearts and their implications for future research aimed at improving the overall rates of recovery.
Collapse
Affiliation(s)
| | | | - Gabriel Sayer
- Cardiovascular Research Foundation, New York, NY (D.B.)
| | - Nir Uriel
- Cardiovascular Research Foundation, New York, NY (D.B.)
| |
Collapse
|
21
|
Dasari TW, Csipo T, Amil F, Lipecz A, Fulop GA, Jiang Y, Samannan R, Johnston S, Zhao YD, Silva-Palacios F, Stavrakis S, Yabluchanskiy A, Po SS. Effects of Low-Level Tragus Stimulation on Endothelial Function in Heart Failure With Reduced Ejection Fraction. J Card Fail 2021; 27:568-576. [PMID: 33387632 PMCID: PMC9473302 DOI: 10.1016/j.cardfail.2020.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Autonomic dysregulation in heart failure with reduced ejection fraction plays a major role in endothelial dysfunction. Low-level tragus stimulation (LLTS) is a novel, noninvasive method of autonomic modulation. METHODS AND RESULTS We enrolled 50 patients with heart failure with reduced ejection fraction (left ventricular ejection fraction of ≤40%) in a randomized, double-blinded, crossover study. On day 1, patients underwent 60 minutes of LLTS with a transcutaneous stimulator (20 Hz, 200 μs pulse width) or sham (ear lobule) stimulation. Macrovascular function was assessed using flow-mediated dilatation in the brachial artery and cutaneous microcirculation with laser speckle contrast imaging in the hand and nail bed. On day 2, patients were crossed over to the other study arm and underwent sham or LLTS; vascular tests were repeated before and after stimulation. Compared with the sham, LLTS improved flow-mediated dilatation by increasing the percent change in the brachial artery diameter (from 5.0 to 7.5, LLTS on day 1, P = .02; and from 4.9 to 7.1, LLTS on day 2, P = .003), compared with no significant change in the sham group (from 4.6 to 4.7, P = .84 on day 1; and from 5.6 to 5.9 on day 2, P = .65). Cutaneous microcirculation in the hand showed no improvement and perfusion of the nail bed showed a trend toward improvement. CONCLUSIONS Our study demonstrated the beneficial effects of acute neuromodulation on macrovascular function. Larger studies to validate these findings and understand mechanistic links are warranted.
Collapse
Affiliation(s)
- Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine; Heart Rhythm Institute.
| | - Tamas Csipo
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | - Faris Amil
- Cardiovascular Section, Department of Internal Medicine
| | - Agnes Lipecz
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | - Gabor A Fulop
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | | | | | - Sarah Johnston
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yan D Zhao
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | | | - Stavros Stavrakis
- Cardiovascular Section, Department of Internal Medicine; Heart Rhythm Institute
| | - Andriy Yabluchanskiy
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine; Heart Rhythm Institute
| |
Collapse
|
22
|
Perry A, Loh F, Adamo L, Zhang KW, Deych E, Foraker R, Mann DL. Unsupervised cluster analysis of patients with recovered left ventricular ejection fraction identifies unique clinical phenotypes. PLoS One 2021; 16:e0248317. [PMID: 33735249 PMCID: PMC7971566 DOI: 10.1371/journal.pone.0248317] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/24/2021] [Indexed: 01/08/2023] Open
Abstract
Background Patients with heart failure (HF) with recovered ejection fraction (HFrecEF) are a recently identified cohort that are phenotypically and biologically different from HFrEF and HFpEF patients. Whether there are unique phenotypes among HFrecEF patients is not known. Methods We studied all patients at a large medical center, who had an improvement in LVEF from ≤ 35% to ≥ 50% (LVrecEF) between January 1, 2005 and December 31, 2013. We identified a set of 11 clinical variables and then performed unsupervised clustering analyses to identify unique clinical phenotypes among patients with LVrecEF, followed by a Kaplan-Meier analysis to identify differences in survival and the proportion of LVrecEF patients who maintained an LVEF ≥ 50% during the study period. Results We identified 889 patients with LVrecEF who clustered into 7 unique phenotypes ranging in size from 37 to 420 patients. Kaplan-Meier analysis demonstrated significant differences in mortality across clusters (logrank p<0.0001), with survival ranging from 14% to 87% at 1000 days, as well as significant differences in the proportion of LVrecEF patients who maintained an LVEF ≥ 50%. Conclusion There is significant clinical heterogeneity among patients with LVrecEF. Clinical outcomes are distinct across phenotype clusters as defined by clinical cardiac characteristics and co-morbidities. Clustering algorithms may identify patients who are at high risk for recurrent HF, and thus be useful for guiding treatment strategies for patients with LVrecEF.
Collapse
Affiliation(s)
- Andrew Perry
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Francis Loh
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Luigi Adamo
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kathleen W. Zhang
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elena Deych
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Randi Foraker
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Douglas L. Mann
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
23
|
Schiffer WB, Perry A, Deych E, Brown DL, Adamo L. Association of early versus delayed normalisation of left ventricular ejection fraction with mortality in ischemic cardiomyopathy. Open Heart 2021; 8:e001528. [PMID: 33723015 PMCID: PMC7970261 DOI: 10.1136/openhrt-2020-001528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE In patients with non-ischaemic cardiomyopathy and reduced left ventricular ejection fraction (LVEF), normalisation of LVEF is associated with improved outcomes. However, data on patients with ischaemic cardiomyopathy and recovered LVEF are lacking. The goal of this study was to assess the prognostic significance of normalisation of the LVEF in patients with ischaemic cardiomyopathy. METHODS/RESULTS We performed a non-prespecified post hoc analysis of the Surgical Treatment for Ischaemic Heart Failure (STICH) trial to determine the association between normalisation of LVEF (>50%) and mortality during follow-up. Of the 1212 patients with LVEF <35% enroled in the STICH trial, 932 underwent assessment of LVEF at 4 months and/or 2 years after enrolment. Among them, 18 patients experienced normalisation in LVEF at 4-month follow-up and 35 patients experienced recovery in LVEF at 2 years. Recovery of LVEF at 4 months and recovery of LVEF at 2 years were not correlated. Recovery of LVEF at 4 months was not associated with reduced all-cause mortality in unadjusted analysis (log-rank test p=0.54) or in Cox proportional hazards analysis (HR: 0.93; 95% CI: 0.48 to 1.80; p=0.82). Ejection fraction recovery at 2 years was associated with a reduction in all-cause mortality, both in unadjusted analysis (log-rank test p=0.004) and in the Cox proportional hazard model (HR: 0.41; 95% CI: 0.21 to 0.80; p=0.009). CONCLUSIONS In patients with ischaemic cardiomyopathy, delayed normalisation of LVEF is associated with reduced mortality, whereas early recovery of LVEF is not. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Walter B Schiffer
- Internal Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Andrew Perry
- Cardiovascular Division, Department of Internal Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elena Deych
- Cardiovascular Division, Department of Internal Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - David L Brown
- Cardiovascular Division, Department of Internal Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Luigi Adamo
- Cardiovascular Division, Department of Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Cheng S, Delling FN, Elkind MSV, Evenson KR, Ferguson JF, Gupta DK, Khan SS, Kissela BM, Knutson KL, Lee CD, Lewis TT, Liu J, Loop MS, Lutsey PL, Ma J, Mackey J, Martin SS, Matchar DB, Mussolino ME, Navaneethan SD, Perak AM, Roth GA, Samad Z, Satou GM, Schroeder EB, Shah SH, Shay CM, Stokes A, VanWagner LB, Wang NY, Tsao CW. Heart Disease and Stroke Statistics-2021 Update: A Report From the American Heart Association. Circulation 2021; 143:e254-e743. [PMID: 33501848 DOI: 10.1161/cir.0000000000000950] [Citation(s) in RCA: 3220] [Impact Index Per Article: 1073.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2021 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population, an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, the global burden of cardiovascular disease, and further evidence-based approaches to changing behaviors related to cardiovascular disease. RESULTS Each of the 27 chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policy makers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
25
|
Wilcox JE, Fang JC, Margulies KB, Mann DL. Heart Failure With Recovered Left Ventricular Ejection Fraction: JACC Scientific Expert Panel. J Am Coll Cardiol 2021; 76:719-734. [PMID: 32762907 DOI: 10.1016/j.jacc.2020.05.075] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/07/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022]
Abstract
Reverse left ventricular (LV) remodeling and recovery of LV function are associated with improved clinical outcomes in patients with heart failure with reduced ejection fraction. A growing body of evidence suggests that even among patients who experience a complete normalization of LV ejection fraction, a significant proportion will develop recurrent LV dysfunction accompanied by recurrent heart failure events. This has led to intense interest in understanding how to manage patients with heart failure with recovered ejection fraction (HFrecEF). Because of the lack of a standard definition for HFrecEF, and the paucity of clinical data with respect to the natural history of HFrecEF patients, there are no current guidelines on how these patients should be followed up and managed. Accordingly, this JACC Scientific Expert Panel reviews the biology of reverse LV remodeling and the clinical course of patients with HFrecEF, as well as provides guidelines for defining, diagnosing, and managing patients with HFrecEF.
Collapse
Affiliation(s)
- Jane E Wilcox
- Division of Cardiovascular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - James C Fang
- Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City, Utah
| | - Kenneth B Margulies
- Translational Research Center, Department of Medicine, University of Pennsylvania Pearlman School of Medicine, Philadelphia, Pennsylvania
| | - Douglas L Mann
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
26
|
Rationale and Study Design of the Withdrawal of Spironolactone for Heart Failure with Improved Left Ventricular Ejection Fraction. INTERNATIONAL JOURNAL OF HEART FAILURE 2021; 3:51-58. [PMID: 36263115 PMCID: PMC9536720 DOI: 10.36628/ijhf.2020.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 11/18/2022]
|
27
|
Margonato D, Mazzetti S, De Maria R, Gorini M, Iacoviello M, Maggioni AP, Mortara A. Heart Failure With Mid-range or Recovered Ejection Fraction: Differential Determinants of Transition. Card Fail Rev 2020; 6:e28. [PMID: 33133642 PMCID: PMC7592465 DOI: 10.15420/cfr.2020.13] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022] Open
Abstract
The recent definition of an intermediate clinical phenotype of heart failure (HF) based on an ejection fraction (EF) of between 40% and 49%, namely HF with mid-range EF (HFmrEF), has fuelled investigations into the clinical profile and prognosis of this patient group. HFmrEF shares common clinical features with other HF phenotypes, such as a high prevalence of ischaemic aetiology, as in HF with reduced EF (HFrEF), or hypertension and diabetes, as in HF with preserved EF (HFpEF), and benefits from the cornerstone drugs indicated for HFrEF. Among the HF phenotypes, HFmrEF is characterised by the highest rate of transition to either recovery or worsening of the severe systolic dysfunction profile that is the target of disease-modifying therapies, with opposite prognostic implications. This article focuses on the epidemiology, clinical characteristics and therapeutic approaches for HFmrEF, and discusses the major determinants of transition to HFpEF or HFrEF.
Collapse
Affiliation(s)
- Davide Margonato
- Department of Clinical Cardiology, Policlinico di Monza Monza, Italy.,Department of Cardiology, University of Pavia Pavia, Italy
| | - Simone Mazzetti
- Department of Clinical Cardiology, Policlinico di Monza Monza, Italy
| | - Renata De Maria
- National Research Council, Institute of Clinical Physiology, ASST Great Metropolitan Hospital Niguarda Milan, Italy
| | | | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia Foggia, Italy
| | | | - Andrea Mortara
- Department of Clinical Cardiology, Policlinico di Monza Monza, Italy
| |
Collapse
|
28
|
Ovics P, Regev D, Baskin P, Davidor M, Shemer Y, Neeman S, Ben-Haim Y, Binah O. Drug Development and the Use of Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Disease Modeling and Drug Toxicity Screening. Int J Mol Sci 2020; 21:E7320. [PMID: 33023024 PMCID: PMC7582587 DOI: 10.3390/ijms21197320] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 12/19/2022] Open
Abstract
: Over the years, numerous groups have employed human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) as a superb human-compatible model for investigating the function and dysfunction of cardiomyocytes, drug screening and toxicity, disease modeling and for the development of novel drugs for heart diseases. In this review, we discuss the broad use of iPSC-CMs for drug development and disease modeling, in two related themes. In the first theme-drug development, adverse drug reactions, mechanisms of cardiotoxicity and the need for efficient drug screening protocols-we discuss the critical need to screen old and new drugs, the process of drug development, marketing and Adverse Drug reactions (ADRs), drug-induced cardiotoxicity, safety screening during drug development, drug development and patient-specific effect and different mechanisms of ADRs. In the second theme-using iPSC-CMs for disease modeling and developing novel drugs for heart diseases-we discuss the rationale for using iPSC-CMs and modeling acquired and inherited heart diseases with iPSC-CMs.
Collapse
Affiliation(s)
- Paz Ovics
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Danielle Regev
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Polina Baskin
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Mor Davidor
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yuval Shemer
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Shunit Neeman
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yael Ben-Haim
- Institute of Molecular and Clinical Sciences, St. George’s University of London, London SW17 0RE, UK;
- Cardiology Clinical Academic Group, St. George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| |
Collapse
|
29
|
Park CS, Park JJ, Mebazaa A, Oh IY, Park HA, Cho HJ, Lee HY, Kim KH, Yoo BS, Kang SM, Baek SH, Jeon ES, Kim JJ, Cho MC, Chae SC, Oh BH, Choi DJ. Characteristics, Outcomes, and Treatment of Heart Failure With Improved Ejection Fraction. J Am Heart Assoc 2020; 8:e011077. [PMID: 30845873 PMCID: PMC6475046 DOI: 10.1161/jaha.118.011077] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Many patients with heart failure (HF) with reduced ejection fraction (HFrEF) experience improvement or recovery of left ventricular ejection fraction (LVEF). Data on clinical characteristics, outcomes, and medical therapy in patients with HF with improved ejection fraction (HFiEF) are scarce. Methods and Results Of 5625 consecutive patients hospitalized for acute HF in the KorAHF (Registry [Prospective Cohort] for Heart Failure in Korea) study, 5103 patients had baseline echocardiography and 2302 patients had follow‐up echocardiography at 12 months. HF phenotypes were defined as persistent HFrEF (LVEF ≤40% at baseline and at 1‐year follow‐up), HFiEF (LVEF ≤40% at baseline and improved up to 40% at 1‐year follow‐up), HF with midrange ejection fraction (LVEF between 40% and <50%), and HF with preserved ejection fraction (LVEF ≥50%). The primary outcome was 4‐year all‐cause mortality from the time of HFiEF diagnosis. Among 1509 HFrEF patients who had echocardiography 1 year after index hospitalization, 720 (31.3%) were diagnosed as having HFiEF. Younger age, female sex, de novo HF, hypertension, atrial fibrillation, and β‐blocker use were positive predictors and diabetes mellitus and ischemic heart disease were negative predictors of HFiEF. During 4‐year follow‐up, patients with HFiEF showed lower mortality than those with persistent HFrEF in univariate, multivariate, and propensity‐score–matched analyses. β‐Blockers, but not renin–angiotensin system inhibitors or mineralocorticoid receptor antagonists, were associated with a reduced all‐cause mortality risk (hazard ratio: 0.59; 95% CI, 0.40–0.87; P=0.007). Benefits for outcome seemed similar among patients receiving low‐ or high‐dose β‐blockers (log‐rank, P=0.304). Conclusions HFiEF is a distinct HF phenotype with better clinical outcomes than other phenotypes. The use of β‐blockers may be beneficial for these patients. Clinical Trial Registration URL: https://www.clinicaltrials.gov. Unique identifier: NCT01389843.
Collapse
Affiliation(s)
- Chan Soon Park
- 1 Graduate School of Medical Science and Engineering Korea Advanced Institute of Science and Technology Daejeon Republic of Korea
| | - Jin Joo Park
- 2 Cardiovascular Center Division of Cardiology Seoul National University Bundang Hospital Seongnam Republic of Korea
| | - Alexandre Mebazaa
- 3 Department of Anesthesiology and Intensive Care Medicine Hôpitaux Universitaires Saint Louis Lariboisière APHP Paris France.,4 Huslw Dept Anesthesie University Paris Diderot Paris France.,5 Laboratoire Marqueurs cardiovasculaires en situations de stress UMR 942 Inserm Paris France
| | - Il-Young Oh
- 2 Cardiovascular Center Division of Cardiology Seoul National University Bundang Hospital Seongnam Republic of Korea
| | - Hyun-Ah Park
- 6 Department of Family Medicine Inje University Seoul Paik Hospital Seoul Republic of Korea
| | - Hyun-Jai Cho
- 7 Department of Internal Medicine Seoul National University Hospital Seoul Republic of Korea
| | - Hae-Young Lee
- 7 Department of Internal Medicine Seoul National University Hospital Seoul Republic of Korea
| | - Kye Hun Kim
- 8 Heart Research Center Chonnam National University Gwangju Republic of Korea
| | - Byung-Su Yoo
- 9 Department of Internal Medicine Yonsei University Wonju College of Medicine Wonju Republic of Korea
| | - Seok-Min Kang
- 10 Department of Internal Medicine Yonsei University College of Medicine Seoul Republic of Korea
| | - Sang Hong Baek
- 11 Department of Internal Medicine Catholic University of Korea Seoul Republic of Korea
| | - Eun-Seok Jeon
- 12 Department of Internal Medicine Sungkyunkwan University College of Medicine Seoul Republic of Korea
| | - Jae-Joong Kim
- 13 Division of Cardiology Asan Medical Center Seoul Republic of Korea
| | - Myeong-Chan Cho
- 14 Department of Internal Medicine Chungbuk National University College of Medicine Cheongju Republic of Korea
| | - Shung Chull Chae
- 15 Department of Internal Medicine Kyungpook National University College of Medicine Daegu Republic of Korea
| | - Byung-Hee Oh
- 16 Department of Internal Medicine Mediplex Sejong Hospital Incheon Republic of Korea
| | - Dong-Ju Choi
- 2 Cardiovascular Center Division of Cardiology Seoul National University Bundang Hospital Seongnam Republic of Korea
| |
Collapse
|
30
|
Gurka J, Piherova L, Majer F, Chaloupka A, Zakova D, Pelak O, Krebsova A, Peichl P, Krejci J, Freiberger T, Melenovsky V, Kautzner J, Kalina T, Sikora J, Kubanek M. Danon disease is an underdiagnosed cause of advanced heart failure in young female patients: a LAMP2 flow cytometric study. ESC Heart Fail 2020; 7:2534-2543. [PMID: 32657043 PMCID: PMC7524080 DOI: 10.1002/ehf2.12823] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/04/2020] [Accepted: 05/20/2020] [Indexed: 01/14/2023] Open
Abstract
Aims Danon disease (DD) is a rare X‐linked disorder caused by mutations in the lysosomal‐associated membrane protein type 2 gene (LAMP2). DD is difficult to distinguish from other causes of dilated or hypertrophic cardiomyopathy (HCM) in female patients. As DD female patients regularly progress into advanced heart failure (AHF) aged 20–40 years, their early identification is critical to improve patient survival and facilitate genetic counselling. In this study, we evaluated the prevalence of DD among female patients with non‐ischemic cardiomyopathy, who reached AHF and were younger than 40 years. Methods and results The study cohort comprised 60 female patients: 47 (78%) heart transplant recipients, 2 (3%) patients treated with ventricular assist device, and 11 (18%) patients undergoing pre‐transplant assessment. Aetiology of the cardiomyopathy was known in 15 patients (including two DD patients). LAMP2 expression in peripheral white blood cells (WBC) was tested by flow cytometry (FC) in the remaining 45 female patients. Whole exome sequencing was used as an alternative independent testing method to FC. Five additional female DD patients (two with different novel LAMP2 mutations) were identified by FC. The total prevalence of DD in this cohort was 12%. HCM phenotype (57% vs. 9%, *P = 0.022) and delta waves identified by electrocardiography (43% vs. 0%, **P = 0.002) were significantly more frequent in DD female patients. Conclusions Danon disease is an underdiagnosed cause of AHF in young female patients. LAMP2 expression testing in peripheral WBCs by FC can be used as an effective screening/diagnostic tool to identify DD in this patient population.
Collapse
Affiliation(s)
- Jiri Gurka
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lenka Piherova
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Filip Majer
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Anna Chaloupka
- 1st Internal Cardioangiologic Clinic, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - Daniela Zakova
- Centre of Cardiovascular and Transplant Surgery, St. Annes University Hospital, Brno, Czech Republic
| | - Ondrej Pelak
- Department of Paediatric Haematology and Oncology, Childhood Leukaemia Investigation Prague, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Alice Krebsova
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Peichl
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Krejci
- 1st Internal Cardioangiologic Clinic, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - Tomas Freiberger
- Centre of Cardiovascular and Transplant Surgery, St. Annes University Hospital, Brno, Czech Republic
| | - Vojtech Melenovsky
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tomas Kalina
- Department of Paediatric Haematology and Oncology, Childhood Leukaemia Investigation Prague, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jakub Sikora
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.,Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Milos Kubanek
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
31
|
Parolin M, Dassie F, Carlo ED, Vettor R, Maffei P. Dome-and-dart T Waves and Hyperthyroidism - A Case Report. EUROPEAN ENDOCRINOLOGY 2020; 16:69-71. [PMID: 32595773 DOI: 10.17925/ee.2020.16.1.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 11/29/2019] [Indexed: 11/24/2022]
Abstract
We briefly describe a case of a 31-year-old man with persistent hyperthyroidism, despite medical treatment with high dose methimazole. Twelve-lead 24-hour Holter monitoring showed bifid (or dome-and-dart) T waves and echocardiography revealed mild left ventricle dilatation. Hyperthyroidism was eventually treated with total thyroidectomy, and thereafter, T waves became normal and the left ventricle returned to normal dimensions. Hyperthyroidism should be considered among the differential diagnoses when T wave abnormalities are observed on electrocardiogram and when mild left ventricle dilatation is observed on an echocardiogram. The correction of hyperthyroidism can reverse these abnormalities.
Collapse
Affiliation(s)
- Matteo Parolin
- Department of Medicine, Internal Medicine-3, University of Padova, Padova, Italy
| | - Francesca Dassie
- Department of Medicine, Internal Medicine-3, University of Padova, Padova, Italy
| | - Eugenio De Carlo
- Department of Medicine, Internal Medicine-3, University of Padova, Padova, Italy
| | - Roberto Vettor
- Department of Medicine, Internal Medicine-3, University of Padova, Padova, Italy
| | - Pietro Maffei
- Department of Medicine, Internal Medicine-3, University of Padova, Padova, Italy
| |
Collapse
|
32
|
Iwahana T, Okada S, Kanda M, Oshima M, Iwama A, Matsumiya G, Kobayashi Y. Novel myocardial markers GADD45G and NDUFS5 identified by RNA-sequencing predicts left ventricular reverse remodeling in advanced non-ischemic heart failure: a retrospective cohort study. BMC Cardiovasc Disord 2020; 20:116. [PMID: 32138671 PMCID: PMC7059273 DOI: 10.1186/s12872-020-01396-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Background Left ventricular reverse remodeling (LVRR) has been detected in non-ischemic dilated cardiomyopathy (NIDCM) patients following optimal treatment. However, its prediction with only conventional modalities is often difficult. This study sought to examine whether RNA sequencing (RNA-seq) of myocardium tissue samples could predict LVRR in NIDCM. Methods A total of 17 advanced NIDCM patients with left ventricular ejection fraction (LVEF) below 30% who underwent cardiac biopsy from Left ventricle (LV) were prospectively recruited. They received optimal treatment and followed with echocardiogram every 6 months. Based on LVRR status after 12 months of treatment, patients were divided into the reverse remodeling (RR) or non-RR group. Tissue samples were analyzed by RNA-seq, and a functional analysis of differentially expressed genes was carried out. Results There were eight and nine patients in the RR and non-RR groups, respectively. No difference was found in age, sex, disease duration, LV end-diastolic diameter, and LVEF between the two groups. There were 155 genes that were differentially expressed between the two groups. Nicotinamide adenine dinucleotide ubiquinone oxidoreductase subunit (NDUF)S5 and Growth arrest and DNA-damage-inducible protein (GADD)45G, along with several genes related to the mitochondrial respiratory chain and ribosome, were significantly downregulated in the RR as compared to the non-RR group. Conclusion GADD45G and NDUFS5 are potential biomarkers for LVRR in patients with advanced NIDCM.
Collapse
Affiliation(s)
- Togo Iwahana
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Masato Kanda
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Motohiko Oshima
- Department of Cellular and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Goro Matsumiya
- Department of Cardiovascular Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| |
Collapse
|
33
|
Tayal U, Wage R, Newsome S, Manivarmane R, Izgi C, Muthumala A, Dungu JN, Assomull R, Hatipoglu S, Halliday BP, Lota AS, Ware JS, Gregson J, Frenneaux M, Cook SA, Pennell DJ, Scott AD, Cleland JG, Prasad SK. Predictors of left ventricular remodelling in patients with dilated cardiomyopathy – a cardiovascular magnetic resonance study. Eur J Heart Fail 2020; 22:1160-1170. [DOI: 10.1002/ejhf.1734] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/05/2019] [Accepted: 11/29/2019] [Indexed: 01/28/2023] Open
Affiliation(s)
- Upasana Tayal
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Ricardo Wage
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Simon Newsome
- Department of Medical Statistics London School of Hygiene and Tropical Medicine London UK
| | | | - Cemil Izgi
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Amal Muthumala
- North Middlesex University Hospital and St Bartholomew's Hospital London UK
| | | | | | - Suzan Hatipoglu
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Brian P. Halliday
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Amrit S. Lota
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - James S. Ware
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
- MRC London Institute of Medical Sciences London UK
| | - John Gregson
- Department of Medical Statistics London School of Hygiene and Tropical Medicine London UK
| | - Michael Frenneaux
- National Heart Lung Institute Imperial College London UK
- University of East Anglia Norwich UK
| | | | - Dudley J. Pennell
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Andrew D. Scott
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - John G.F. Cleland
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Sanjay K. Prasad
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| |
Collapse
|
34
|
Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Delling FN, Djousse L, Elkind MSV, Ferguson JF, Fornage M, Khan SS, Kissela BM, Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino ME, Perak AM, Rosamond WD, Roth GA, Sampson UKA, Satou GM, Schroeder EB, Shah SH, Shay CM, Spartano NL, Stokes A, Tirschwell DL, VanWagner LB, Tsao CW. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation 2020; 141:e139-e596. [PMID: 31992061 DOI: 10.1161/cir.0000000000000757] [Citation(s) in RCA: 4995] [Impact Index Per Article: 1248.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports on the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2020 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population, metrics to assess and monitor healthy diets, an enhanced focus on social determinants of health, a focus on the global burden of cardiovascular disease, and further evidence-based approaches to changing behaviors, implementation strategies, and implications of the American Heart Association's 2020 Impact Goals. RESULTS Each of the 26 chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policy makers, media professionals, clinicians, healthcare administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
35
|
Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, Delling FN, Djousse L, Elkind MSV, Ferguson JF, Fornage M, Jordan LC, Khan SS, Kissela BM, Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino ME, O'Flaherty M, Pandey A, Perak AM, Rosamond WD, Roth GA, Sampson UKA, Satou GM, Schroeder EB, Shah SH, Spartano NL, Stokes A, Tirschwell DL, Tsao CW, Turakhia MP, VanWagner LB, Wilkins JT, Wong SS, Virani SS. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation 2019; 139:e56-e528. [PMID: 30700139 DOI: 10.1161/cir.0000000000000659] [Citation(s) in RCA: 5463] [Impact Index Per Article: 1092.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Brann A, Tran H, Greenberg B. Contemporary approach to treating heart failure. Trends Cardiovasc Med 2019; 30:507-518. [PMID: 31901378 DOI: 10.1016/j.tcm.2019.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/20/2019] [Accepted: 11/24/2019] [Indexed: 01/01/2023]
Abstract
Over the past several decades, important advances have been made in the treatment of patients with heart failure (HF). Whereas in the past, the main goal of drug therapy was to relieve congestion, there is now compelling evidence from randomized clinical trials (RCTs) showing that several classes of drugs, most of which work predominantly by blocking or modulating neurohormonal activity, can substantially reduce morbidity and mortality as well as improve quality of life in patients with HF. Most of these trials, however, separated patients according to whether their ejection fraction (EF) was reduced (HFrEF) or preserved (HFpEF) and for the most part, favorable effects on clinical outcomes were demonstrated only in patients with HFrEF. In addition to the paucity of effective agents for managing patients with HFpEF, it has become apparent that underutilization of available therapies has greatly limited the overall impact of medical therapy on outcomes. This review provides an overview of current medical management of HF across the spectrum of EF, including the underutilization of treatment modalities. The focus is to provide clinicians the rationale for the use of specific agents and to present a practical approach for patient management. The strategies discussed are based on results of RCTs, guideline recommendations and the authors' own experience in managing patients with HF over the years.
Collapse
Affiliation(s)
- Alison Brann
- Department of Cardiology and the Sulpizio Family Cardiovascular Center, University of California, San Diego Medical Center, La Jolla, CA, United States
| | - Hao Tran
- Department of Cardiology and the Sulpizio Family Cardiovascular Center, University of California, San Diego Medical Center, La Jolla, CA, United States
| | - Barry Greenberg
- Department of Cardiology and the Sulpizio Family Cardiovascular Center, University of California, San Diego Medical Center, La Jolla, CA, United States.
| |
Collapse
|
37
|
Iyngkaran P, Liew D, Neil C, Driscoll A, Marwick TH, Hare DL. Moving From Heart Failure Guidelines to Clinical Practice: Gaps Contributing to Readmissions in Patients With Multiple Comorbidities and Older Age. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2018; 12:1179546818809358. [PMID: 30618487 PMCID: PMC6299336 DOI: 10.1177/1179546818809358] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
Abstract
This feature article for the thematic series on congestive heart failure (CHF) readmissions aims to outline important gaps in guidelines for patients with multiple comorbidities and the elderly. Congestive heart failure diagnosis manifests as a 3-phase journey between the hospital and community, during acute, chronic stable, and end-of-life (palliative) phases. This journey requires in variable intensities a combination of multidisciplinary care within tertiary hospital or ambulatory care from hospital outpatients or primary health services, within the general community. Management goals are uniform, ie, to achieve the lowest New York Heart Association class possible, with improvement in ejection fraction, by delivering gold standard therapies within a CHF program. Comorbidities are an important common denominator that influences outcomes. Comorbidities include diabetes mellitus, chronic obstructive airways disease, chronic renal impairment, hypertension, obesity, sleep apnea, and advancing age. Geriatric care includes the latter as well as syndromes such as frailty, falls, incontinence, and confusion. Many systems still fail to comprehensively achieve all aspects of such programs. This review explores these factors.
Collapse
Affiliation(s)
- Pupalan Iyngkaran
- Northern Territory Medical Program, Flinders University, Darwin, NT, Australia
- Pupalan Iyngkaran, Yellow Building 4 Cnr University Drive North & University Drive West Charles Darwin University, Casuarina, NT 0815, Australia.
| | - Danny Liew
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Christopher Neil
- Department of Medicine—Western Precinct, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrea Driscoll
- School of Nursing and Midwifery, Deakin University, Geelong, VIC, Australia
- Austin Health, Melbourne, VIC, Australia
| | | | - David L Hare
- Cardiovascular Research, The University of Melbourne, Melbourne, VIC, Australia
- Heart Failure Services, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
38
|
Chaloupka A, Piherova L, Grochova I, Binova J, Krejci J, Spinarova L, Stranecky V, Kmoch S, Kubanek M. Genetic architecture of recent-onset dilated cardiomyopathy in Moravian region assessed by whole-exome sequencing and its clinical correlates. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2018; 163:309-317. [PMID: 30275597 DOI: 10.5507/bp.2018.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/07/2018] [Indexed: 01/02/2023] Open
Abstract
AIMS Recent-onset dilated cardiomyopathy (RODCM) is a disease of heterogeneous aetiology and clinical outcome. In this pilot study, we aimed to assess its genetic architecture and correlate genotype with left ventricular reverse remodelling (LVRR). PATIENTS AND METHODS In this multi-centre prospective observational study, we enrolled 83 Moravian patients with RODCM and a history of symptoms of less than 6 months, for whole-exome sequencing (WES). All patients underwent 12-month clinical and echocardiographic follow-up. LVRR was defined as an absolute increase in left ventricular ejection fraction > 10% accompanied by a relative decrease of left ventricular end-diastolic diameter > 10% at 12 months. RESULTS WES identified at least one disease-related variant in 45 patients (54%). LVRR occurred in 28 patients (34%), most often in carriers of isolated titin truncated variants, followed by individuals with a negative, or inconclusive WES and carriers of other disease-related variants (56% vs. 42% vs. 19%, P=0.041). CONCLUSION A substantial proportion of RODCM cases have a monogenic or oligogenic genetic background. Carriers of non-titin disease-related variants are less likely to reach LVRR at 12- months than other individuals. Genetic testing could contribute to better prognosis prediction and individualized treatment of RODCM.
Collapse
Affiliation(s)
- Anna Chaloupka
- 1 st Internal Clinic of Cardio-angiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Piherova
- Research Unit for Rare Diseases, Department of Paediatrics and Adolescent Medicine, 1 st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ilga Grochova
- 1 st Internal Clinic of Cardio-angiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Binova
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Krejci
- 1 st Internal Clinic of Cardio-angiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Spinarova
- 1 st Internal Clinic of Cardio-angiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Viktor Stranecky
- Research Unit for Rare Diseases, Department of Paediatrics and Adolescent Medicine, 1 st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Paediatrics and Adolescent Medicine, 1 st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Milos Kubanek
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
39
|
Sun X, Shan A, Wei Z, Xu B. Intravenous mesenchymal stem cell-derived exosomes ameliorate myocardial inflammation in the dilated cardiomyopathy. Biochem Biophys Res Commun 2018; 503:2611-2618. [PMID: 30126637 DOI: 10.1016/j.bbrc.2018.08.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have been shown to be efficacy to attenuating cardiovascular inflammation; however, there are many limitations to stem cell treatment. Present study was to prove MSC-derived exosomes (MSC-Exos) could alleviating inflammatory cardiomyopathy by improving the inflammatory microenvironment of myocardium, especially by regulating the activity of macrophages. Mice were intraperitoneal injected of doxorubicin (DOX) to establish a dilated cardiomyopathy (DCM) model, and then received intravenous injection of either MSC-Exos or PBS as control. Mice receiving MSC-Exos showed improved cardiac function via echocardiography and attenuated cardiac dilation via HE staining, as well as reduced cardiomyocytes apoptosis. Expression levels of inflammatory factors were reduced. And there was a significant decrease of the inflammatory cells infiltration in the MSC-Exos treatment group comparing to the PBS group. Meanwhile, MSC-Exos could remarkably attenuate the pro-inflammatory macrophages amount in both blood and heart, which was proved that MSC-Exos relied on the JAK2-STAT6 pathway mediating macrophages activation. MSC-Exos improved the inflammatory microenvironment of dilated cardiomyopathy by regulating the polarization of the macrophage, which may hold promise for dilated cardiomyopathy clinical therapy.
Collapse
Affiliation(s)
- Xuan Sun
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Anqi Shan
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zilun Wei
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
40
|
Gulati G, Udelson JE. Heart Failure With Improved Ejection Fraction: Is it Possible to Escape One's Past? JACC-HEART FAILURE 2018; 6:725-733. [PMID: 30098965 DOI: 10.1016/j.jchf.2018.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022]
Abstract
Among patients with heart failure with reduced ejection fraction, investigators have repeatedly identified a subgroup whose left ventricular ejection fraction and structural remodeling can improve to normal or nearly normal levels with or without medical therapy. This subgroup of patients with "heart failure with improved ejection fraction" has distinct clinical characteristics and a more favorable prognosis compared with patients who continue to have reduced ejection fraction. However, many of these patients also manifest clinical and biochemical signs of incomplete resolution of heart failure pathophysiology and remain at some risk of adverse outcomes, thus indicating that they may not have completely recovered. Although rigorous evidence on managing these patients is sparse, there are several reasons to recommend continuation of heart failure therapies, including device therapies, to prevent clinical deterioration. Notable exceptions to this recommendation may include patients who recover from peripartum cardiomyopathy, fulminant myocarditis, or stress cardiomyopathy, whose excellent long-term prognoses may imply true myocardial recovery. More research on these patients is needed to better understand the mechanisms that lead to improvement in ejection fraction and to guide their clinical management.
Collapse
Affiliation(s)
- Gaurav Gulati
- Division of Cardiology and the CardioVascular Center, Tufts Medical Center, Boston, Massachusetts
| | - James E Udelson
- Division of Cardiology and the CardioVascular Center, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
41
|
Sustained Cardiac Recovery Hinges on Timing and Natural History of Underlying Condition. Am J Med Sci 2018; 356:47-55. [DOI: 10.1016/j.amjms.2018.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/14/2017] [Accepted: 02/21/2018] [Indexed: 01/12/2023]
|
42
|
Valika A, Paprockas K, Villines D, Costanzo MR. Importance of baseline heart rate as a predictor of cardiac functional recovery in newly diagnosed heart failure with reduced ejection fraction. Clin Cardiol 2018; 41:752-757. [PMID: 29512170 DOI: 10.1002/clc.22937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 02/24/2018] [Accepted: 02/28/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Left ventricular ejection fraction (LVEF) has shown to predict outcomes in patients with heart failure (HF). Left ventricular recovery (LVR) has shown to improve prognosis. HYPOTHESIS Guideline-directed medical therapy will predict LVR in patients with HF and reduced LVEF. METHODS We studied 244 patients with newly diagnosed HF and an LVEF ≤35%. LVR was defined as an increase in LVEF ≥40%. Patients who experienced LVR were compared with those who had persistent left ventricular dysfunction. RESULTS Population characteristics included ischemic etiology, 38.1%; baseline LVEF, 23% ±6%; and mean baseline heart rate (HR), 75 ±13 bpm. Guideline-directed medical therapy was achieved as follows: angiotensin-converting enzyme inhibitors, 74.3%; β-blockers (BB), 95.4%; target dosing of angiotensin-converting enzyme inhibitors, 33.7%; target dosing of BB, 40.2%. LVR occurred in 154/244 patients (63.1%). By multivariable analysis, baseline HR ≤70 bpm was the only independent predictor of LVR (odds ratio: 3.39, 95% confidence interval: 1.5-7.5, P = 0.003). Target dosing of BB therapy was predictive of LVR only in the univariate analysis (odds ratio: 1.9, 95% confidence interval: 1.1-3.4, P = 0.03). Furthermore, the composite endpoint of HF hospitalization or mortality occurred less frequently in those who did vs those who did not achieve target BB doses (5.4% vs 16.7%, respectively; P = 0.023). CONCLUSIONS The novel findings of our analysis reveal that the only predictor of LVR in this study was a low baseline HR. Early modulation of HR in newly diagnosed HF patients may increase the rates of LVR.
Collapse
Affiliation(s)
- Ali Valika
- Department of Cardiology, Advocate Heart Institute, Oak Brook, Illinois
| | - Kim Paprockas
- Advocate Research Institute, Advocate Health Care, Oak Brook, Illinois
| | - Dana Villines
- Advocate Research Institute, Advocate Health Care, Oak Brook, Illinois
| | | |
Collapse
|
43
|
Affiliation(s)
- Bojan Vrtovec
- From the Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, Slovenia
| |
Collapse
|
44
|
Abstract
Advances in medical and device therapies have demonstrated the capacity of the heart to reverse the failing phenotype. The development of normative changes to ventricular size and function led to the concept of reverse remodelling. Among heart failure therapies, durable mechanical circulatory support is most consistently associated with the largest degree of reverse remodelling. Accordingly, research to analyse human tissue after a period of mechanical circulatory support continues to yield a wealth of information. In this Review, we summarize the latest findings on reverse remodelling and myocardial recovery. Accumulating evidence shows that the molecular changes associated with heart failure, in particular in the transcriptome, metabalome, and extracellular matrix, persist in the reverse-remodelled myocardium despite apparent normalization of macrolevel properties. Therefore, reverse remodelling should be distinguished from true myocardial recovery, in which a failing heart regains both normal function and molecular makeup. These findings have implications for future research to develop therapies to repair fully the failing myocardium. Meanwhile, recognition by society guidelines of this new clinical phenotype, which is coming to be known as a state of heart failure remission, underscores the need to accurately define and identify reverse modelled myocardium for the establishment of appropriate therapies.
Collapse
|
45
|
Adamo L, Perry A, Novak E, Makan M, Lindman BR, Mann DL. Abnormal Global Longitudinal Strain Predicts Future Deterioration of Left Ventricular Function in Heart Failure Patients With a Recovered Left Ventricular Ejection Fraction. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003788. [PMID: 28559418 DOI: 10.1161/circheartfailure.116.003788] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/24/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Patients with recovery of left ventricular ejection fraction (LVEF) remain at risk for future deterioration of LVEF. However, there are no tools to risk stratify these patients. We hypothesized that global longitudinal strain (GLS) could predict sustained recovery within this population. METHODS AND RESULTS We retrospectively identified 96 patients with a reduced LVEF <50% (screening echocardiogram), whose LVEF had increased by at least 10% and normalized (>50%) on evidence-based medical therapies (baseline echocardiogram). We examined absolute GLS on the baseline echocardiogram in relation to changes in LVEF on a follow-up echocardiogram. Patients with recovered LVEF had a wide range of GLS. The GLS on the baseline study correlated with the LVEF at the time of follow-up (r=0.33; P<0.001). The likelihood of having an LVEF >50% on follow-up increased by 24% for each point increase in absolute GLS on the baseline study (odds ratio, 1.24; P=0.001). An abnormal GLS (≤16%) at baseline had a sensitivity of 88%, a specificity of 46%, and an accuracy of 0.67 (P<0.001) as a predictor of a decrease in LVEF >5% during follow-up. A normal GLS (>16%) on the baseline study had a sensitivity of 47%, a specificity of 83%, and an accuracy of 0.65 (P=0.002) for predicting a stable LVEF (-5% to 5%) on follow-up. CONCLUSIONS In patients with a recovered LVEF, an abnormal GLS predicts the likelihood of having a decreased LVEF during follow-up, whereas a normal GLS predicts the likelihood of stable LVEF during recovery.
Collapse
Affiliation(s)
- Luigi Adamo
- From the Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Andrew Perry
- From the Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Eric Novak
- From the Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Majesh Makan
- From the Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Brian R Lindman
- From the Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Douglas L Mann
- From the Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
46
|
Lindley KJ, Conner SN, Cahill AG, Novak E, Mann DL. Impact of Preeclampsia on Clinical and Functional Outcomes in Women With Peripartum Cardiomyopathy. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003797. [PMID: 28572214 DOI: 10.1161/circheartfailure.116.003797] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/24/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Preeclampsia is a risk factor for the development of peripartum cardiomyopathy (PPCM), but it is unknown whether preeclampsia impacts clinical or left ventricular (LV) functional outcomes. This study sought to assess clinical and functional outcomes in women with PPCM complicated by preeclampsia. METHODS AND RESULTS This retrospective cohort study included women diagnosed with PPCM delivering at Barnes-Jewish Hospital between 2004 to 2014. The primary outcome was one-year event-free survival rate for the combined end point of death and hospital readmission. The secondary outcome was recovery of LV ejection fraction. Seventeen of 39 women (44%) with PPCM had preeclampsia. The groups had similar mean LV ejection fraction at diagnosis (29.6 with versus 27.3 without preeclampsia; P=0.5). Women with preeclampsia had smaller mean LV end-diastolic diameters (5.2 versus 6.0 cm; P=0.001), greater relative wall thickness (0.41 versus 0.35 mm Hg; P=0.009), and lower incidence of eccentric remodeling (12% versus 48%; P=0.03). Clinical follow-up was available for 32 women; 5 died of cardiovascular complications within 1 year of diagnosis (4/15 with versus 1/17 without preeclampsia; P=0.16). In time to event analysis, patients with preeclampsia had worse event-free survival during 1-year follow-up (P=0.047). Echocardiographic follow-up was available in 10 survivors with and 16 without preeclampsia. LV ejection fraction recovered in 80% of survivors with versus 25% without preeclampsia (P=0.014). CONCLUSIONS PPCM with concomitant preeclampsia is associated with increased morbidity and mortality and different patterns of LV remodeling and recovery of LV function when compared with patients with PPCM that is not complicated by preeclampsia.
Collapse
Affiliation(s)
- Kathryn J Lindley
- From the Cardiovascular Division, Department of Medicine (K.J.L., E.N., D.L.M.) and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (S.N.C., A.G.C.),Washington University School of Medicine, St Louis, MO.
| | - Shayna N Conner
- From the Cardiovascular Division, Department of Medicine (K.J.L., E.N., D.L.M.) and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (S.N.C., A.G.C.),Washington University School of Medicine, St Louis, MO
| | - Alison G Cahill
- From the Cardiovascular Division, Department of Medicine (K.J.L., E.N., D.L.M.) and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (S.N.C., A.G.C.),Washington University School of Medicine, St Louis, MO
| | - Eric Novak
- From the Cardiovascular Division, Department of Medicine (K.J.L., E.N., D.L.M.) and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (S.N.C., A.G.C.),Washington University School of Medicine, St Louis, MO
| | - Douglas L Mann
- From the Cardiovascular Division, Department of Medicine (K.J.L., E.N., D.L.M.) and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (S.N.C., A.G.C.),Washington University School of Medicine, St Louis, MO
| |
Collapse
|
47
|
Heart Failure with Myocardial Recovery - The Patient Whose Heart Failure Has Improved: What Next? Prog Cardiovasc Dis 2017; 60:226-236. [PMID: 28551473 DOI: 10.1016/j.pcad.2017.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 02/06/2023]
Abstract
In an important number of heart failure (HF) patients substantial or complete myocardial recovery occurs. In the strictest sense, myocardial recovery is a return to both normal structure and function of the heart. HF patients with myocardial recovery or recovered ejection fraction (EF; HFrecEF) are a distinct population of HF patients with different underlying etiologies, demographics, comorbidities, response to therapies and outcomes compared to HF patients with persistent reduced (HFrEF) or preserved ejection fraction (HFpEF). Improvement of left ventricular EF has been systematically linked to improved quality of life, lower rehospitalization rates and mortality. However, mortality and morbidity in HFrecEF patients remain higher than in the normal population. Also, persistent abnormalities in biomarker and gene expression profiles in these patients lends weight to the hypothesis that pathological processes are ongoing. Currently, there remains a lack of data to guide the management of HFrecEF patients. This review will discuss specific characteristics, pathophysiology, clinical implications and future needs for HFrecEF.
Collapse
|
48
|
Cho JY, Kim KH, Song JE, Kim JE, Park H, Yoon HJ, Yoon NS, Hong YJ, Park HW, Kim JH, Ahn Y, Jeong MH, Cho JG, Park JC. Predictors of Left Ventricular Functional Recovery and Their Impact on Clinical Outcomes in Patients With Newly Diagnosed Dilated Cardiomyopathy and Heart Failure. Heart Lung Circ 2017; 27:41-49. [PMID: 28420549 DOI: 10.1016/j.hlc.2017.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/31/2017] [Accepted: 02/09/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND To identify the predictors of left ventricular functional recovery (LVFR) and its impacts on clinical outcomes in acute heart failure (AHF) patients with newly diagnosed dilated cardiomyopathy (DCM). METHODS A total of 175 consecutive patients with newly diagnosed DCM and AHF were divided into two groups according to LVFR on FU echocardiography; the recovered group (n=54, 54.3±18.5years, 31 males) vs. the non-recovered group (n=121, 60.5±15.1years, 79 males). Clinical, laboratory, and echocardiographic findings were compared, and major adverse cardiac and cerebrovascular events (MACCE) including death, rehospitalisation, and stroke were analysed. RESULTS Left ventricular function (LV) was normalised in 54 patients (30.8%) on follow-up echocardiography. The change in the level of N-terminal pro-B-type natriuretic peptide (ΔNT-proBNP) between initial presentation and discharge >1633.5pg/mL was an independent predictor of LVFR, whereas diabetes and LV end-systolic diameter >50mm were negative predictors of LVFR on multivariate analysis. During five years of clinical follow-up, MACCE developed in 91 patients: 58 deaths, 29 rehospitalisations, and 4 strokes. On multivariate analysis, baseline LVEF <30% and no LVFR were independent predictors of MACCE. CONCLUSION Left ventricular functional recovery was not uncommon in newly diagnosed DCM with AHF. The changes in NT-proBNP level during hospitalisation, diabetes, and larger initial LV size were independent predictors of LVFR, and LVFR was an independent predictor of future MACCE. Serial monitoring of NT-proBNP and LV function would be useful in the risk stratification of newly diagnosed DCM with AHF.
Collapse
Affiliation(s)
- Jae Yeong Cho
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Kye Hun Kim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea.
| | - Ji Eun Song
- Presbyterian Medical Center, Jeonju, Republic of Korea
| | - Ji Eun Kim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyukjin Park
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyun Ju Yoon
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Nam Sik Yoon
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Young Joon Hong
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyung Wook Park
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Ju Han Kim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Jeong Gwan Cho
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Jong Chun Park
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| |
Collapse
|
49
|
Narayan HK, Finkelman B, French B, Plappert T, Hyman D, Smith AM, Margulies KB, Ky B. Detailed Echocardiographic Phenotyping in Breast Cancer Patients: Associations With Ejection Fraction Decline, Recovery, and Heart Failure Symptoms Over 3 Years of Follow-Up. Circulation 2017; 135:1397-1412. [PMID: 28104715 PMCID: PMC5388560 DOI: 10.1161/circulationaha.116.023463] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 01/13/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cardiovascular disease in patients with breast cancer is of growing concern. The longitudinal effects of commonly used therapies, including doxorubicin and trastuzumab, on cardiac remodeling and function remain unknown in this population. We aimed to define the changes in echocardiographic parameters of structure, function, and ventricular-arterial coupling, and their associations with left ventricular ejection fraction (LVEF) and heart failure symptoms. METHODS In a longitudinal prospective cohort study of 277 breast cancer participants receiving doxorubicin (Dox), trastuzumab (Tras), or both (Dox+Tras), we obtained 1249 echocardiograms over a median follow-up of 2.0 (interquartile range, 1.0-3.0) years. Left ventricular structure, diastolic and contractile function, and ventricular-arterial coupling measures were quantified in a core laboratory blinded to participant characteristics. We evaluated changes in echocardiographic parameters over time, and used repeated-measures regression models to define their association with LVEF decline and recovery. Linear regression models defined the association between early changes in these parameters and subsequent changes in LVEF and heart failure symptoms. RESULTS Overall, 177 (64%) received Dox, 51 (18%) received Tras, and 49 (18%) received Dox+Tras. With Dox, there was a sustained, modest decrease in LVEF over the follow-up duration (1-year change in LVEF -3.6%; 95% confidence interval [CI], -4.4% to -2.8%; 3-year change -3.8%; 95% CI, -5.1% to -2.5%). With Tras, a similar LVEF decline was observed at 1 year (-4.5%; 95% CI, -6.0% to -2.9%) and 3 years (-2.8%; 95%CI, -5.3 to -0.4%). Participants receiving Dox+Tras demonstrated the greatest declines at 1 year (-6.6%; 95% CI, -8.2 to -5.0%), with partial recovery at 3 years (-2.8%; 95% CI, -4.8 to -0.8%). LVEF declines and recovery were associated primarily with changes in systolic volumes, longitudinal and circumferential strain, and ventricular-arterial coupling indices, effective arterial elastance (Ea) and the coupling ratio Ea/Eessb, without evidence for effect modification across therapies. Early changes in volumes, strain, and Ea/Eessb at 4 to 6 months were associated with 1- and 2-year LVEF changes. Similarly, early changes in strain and Ea were associated with worsening heart failure symptoms at 1 year. CONCLUSIONS Doxorubicin and trastuzumab resulted in modest, persistent declines in LVEF at 3 years. Changes in volumes, strain, and ventricular-arterial coupling were consistently associated with concurrent and subsequent LVEF declines and recovery across therapies.
Collapse
Affiliation(s)
- Hari K Narayan
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - Brian Finkelman
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - Benjamin French
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - Theodore Plappert
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - David Hyman
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - Amanda M Smith
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - Kenneth B Margulies
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.)
| | - Bonnie Ky
- From Department of Pediatrics, Division of Cardiology, The Children's Hospital of Philadelphia, PA (H.K.N.); Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, B. French, B.K.); and Department of Medicine, Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (B. Finkelman, T.P., D.H., A.M.S., K.B.M., B.K.).
| |
Collapse
|
50
|
Marinescu KK, Uriel N, Mann DL, Burkhoff D. Left ventricular assist device-induced reverse remodeling: it's not just about myocardial recovery. Expert Rev Med Devices 2016; 14:15-26. [PMID: 27871197 DOI: 10.1080/17434440.2017.1262762] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The abnormal structure, function and molecular makeup of dilated cardiomyopathic hearts can be partially normalized in patients supported by a left ventricular assist device (LVAD), a process called reverse remodeling. This leads to recovery of function in many patients, though the rate of full recovery is low and in many cases is temporary, leading to the concept of heart failure remission, rather than recovery. Areas covered: We summarize data indicative of ventricular reverse remodeling, recovery and remission during LVAD support. These terms were used in searches performed in Pubmed. Duplication of topics covered in depth in prior review articles were avoided. Expert commentary: Although most patients undergoing mechanical circulatory support (MCS) show a significant degree of reverse remodeling, very few exhibit sufficiently improved function to justify device explantation, and many from whom LVADs have been explanted have relapsed back to the original heart failure phenotype. Future research has the potential to clarify the ideal combination of pharmacological, cell, gene, and mechanical therapies that would maximize recovery of function which has the potential to improve exercise tolerance of patients while on support, and to achieve a higher degree of myocardial recovery that is more likely to persist after device removal.
Collapse
Affiliation(s)
- Karolina K Marinescu
- a Department of Medicine, Division of Cardiology, Advanced Heart Failure , Rush University Medical Center , Chicago , IL , USA
| | - Nir Uriel
- b Department of Medicine, Division of Cardiology , University of Chicago , Chicago , IL , USA
| | - Douglas L Mann
- c Department of Medicine, Division of Cardiology , Washington University School of Medicine/Barnes Jewish Hospital , St. Louis , MO , USA
| | - Daniel Burkhoff
- d Department of Medicine, Division of Cardiology , Columbia University Medical Center/New York-Presbyterian Hospital , New York , NY , USA
| |
Collapse
|