101
|
Ceriello A, Catrinoiu D, Chandramouli C, Cosentino F, Dombrowsky AC, Itzhak B, Lalic NM, Prattichizzo F, Schnell O, Seferović PM, Valensi P, Standl E. Heart failure in type 2 diabetes: current perspectives on screening, diagnosis and management. Cardiovasc Diabetol 2021; 20:218. [PMID: 34740359 PMCID: PMC8571004 DOI: 10.1186/s12933-021-01408-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes is one of the most relevant risk factors for heart failure, the prevalence of which is increasing worldwide. The aim of the review is to highlight the current perspectives of the pathophysiology of heart failure as it pertains to type 2 diabetes. This review summarizes the proposed mechanistic bases, explaining the myocardial damage induced by diabetes-related stressors and other risk factors, i.e., cardiomyopathy in type 2 diabetes. We highlight the complex pathology of individuals with type 2 diabetes, including the relationship with chronic kidney disease, metabolic alterations, and heart failure. We also discuss the current criteria used for heart failure diagnosis and the gold standard screening tools for individuals with type 2 diabetes. Currently approved pharmacological therapies with primary use in type 2 diabetes and heart failure, and the treatment-guiding role of NT-proBNP are also presented. Finally, the influence of the presence of type 2 diabetes as well as heart failure on COVID-19 severity is briefly discussed.
Collapse
Affiliation(s)
- Antonio Ceriello
- IRCCS MultiMedica, Via Gaudenzio Fantoli, 16/15, 20138 Milan, Italy
| | - Doina Catrinoiu
- Faculty of Medicine, Clinical Center of Diabetes, Nutrition and Metabolic Diseases, Ovidius University of Constanta, Constanta, Romania
| | - Chanchal Chandramouli
- Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute, National Heart Centre, Singapore, Singapore
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | | | - Baruch Itzhak
- Clalit Health Services and Technion Faculty of Medicine, Haifa, Israel
| | - Nebojsa Malić Lalic
- School of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University of Belgrade, Belgrade, Serbia
| | | | - Oliver Schnell
- Forschergruppe Diabetes e. V. at Helmholtz Centre Munich GmbH, Munich, Germany
| | - Petar M. Seferović
- School of Medicine, University of Belgrade, Belgrade University Medical Center, Belgrade, Serbia
| | - Paul Valensi
- Unit of Endocrinology, Diabetology, Nutrition, Jean Verdier Hospital, AP-HP, CRNH-IdF, CINFO, Paris 13 University, Bondy, France
| | - Eberhard Standl
- Forschergruppe Diabetes e. V. at Helmholtz Centre Munich GmbH, Munich, Germany
| | - the D&CVD EASD Study Group
- IRCCS MultiMedica, Via Gaudenzio Fantoli, 16/15, 20138 Milan, Italy
- Faculty of Medicine, Clinical Center of Diabetes, Nutrition and Metabolic Diseases, Ovidius University of Constanta, Constanta, Romania
- Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute, National Heart Centre, Singapore, Singapore
- Unit of Cardiology, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
- Sciarc GmbH, Baierbrunn, Germany
- Clalit Health Services and Technion Faculty of Medicine, Haifa, Israel
- School of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University of Belgrade, Belgrade, Serbia
- Forschergruppe Diabetes e. V. at Helmholtz Centre Munich GmbH, Munich, Germany
- School of Medicine, University of Belgrade, Belgrade University Medical Center, Belgrade, Serbia
- Unit of Endocrinology, Diabetology, Nutrition, Jean Verdier Hospital, AP-HP, CRNH-IdF, CINFO, Paris 13 University, Bondy, France
| |
Collapse
|
102
|
Cai H, Men H, Cao P, Zheng Y. Mechanism and prevention strategy of a bidirectional relationship between heart failure and cancer (Review). Exp Ther Med 2021; 22:1463. [PMID: 34737803 PMCID: PMC8561773 DOI: 10.3892/etm.2021.10898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
The relationship between cancer and heart failure has been extensively studied in the last decade. These studies have focused on describing heart injury caused by certain cancer treatments, including radiotherapy, chemotherapy and targeted therapy. Previous studies have demonstrated a higher incidence of cancer in patients with heart failure. Heart failure enhances an over-activation of the sympathetic nervous system and the renin-angiotensin-aldosterone system, and subsequently promotes cancer development. Other studies have found that heart failure and cancer both have a common pathological origin, flanked by chronic inflammation in certain organs. The present review aims to summarize and describe the recent discoveries, suggested mechanisms and relationships between heart failure and cancer. The current review provides more ideas on clinical prevention strategies according to the pathological mechanism involved.
Collapse
Affiliation(s)
- He Cai
- Cardiovascular Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongbo Men
- Cardiovascular Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Pengyu Cao
- Cardiovascular Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Zheng
- Cardiovascular Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
103
|
El Hajj EC, El Hajj MC, Sykes B, Lamicq M, Zile MR, Malcolm R, O'Neil PM, Litwin SE. Pragmatic Weight Management Program for Patients With Obesity and Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2021; 10:e022930. [PMID: 34713711 PMCID: PMC8751835 DOI: 10.1161/jaha.121.022930] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Obesity is associated with heart failure with preserved ejection fraction (HFpEF). Weight loss can improve exercise capacity in HFpEF. However, previously reported methods of weight loss are impractical for widespread clinical implementation. We tested the hypothesis that an intensive lifestyle modification program would lead to relevant weight loss and improvement in functional status in patients with HFpEF and obesity. Methods and Results Patients with ejection fraction >45%, at least 1 objective criteria for HFpEF, and body mass index ≥30 kg/m2 were offered enrollment in an established 15-week weight management program that included weekly visits for counseling, weight checks, and provision of meal replacements. At baseline, 15 weeks, and 26 weeks, Minnesota Living With Heart Failure score, 6-minute walk distance, echocardiography, and laboratory variables were assessed. A total of 41 patients completed the study (mean body mass index, 40.8 kg/m2), 74% of whom lost >5% of their baseline body weight following the 15-week program. At 15 weeks, mean 6-minute walk distance increased from 223 to 281 m (P=0.001) and then decreased to 267 m at 26 weeks. Minnesota Living With Heart Failure score improved from 59.9 to 37.3 at 15 weeks (P<0.001) and 37.06 at 26 weeks. Changes in weight correlated with change in Minnesota Living With Heart Failure score (r=0.452; P=0.000) and 6-minute walk distance (r=-0.388; P<0.001). Conclusions In a diverse population of patients with obesity and HFpEF, clinically relevant weight loss can be achieved with a pragmatic 15-week program. This is associated with significant improvements in quality of life and exercise capacity. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT02911337.
Collapse
Affiliation(s)
- Elia C El Hajj
- Department of Physiology Louisiana State University New Orleans LA
| | | | - Brandon Sykes
- Division of Cardiology Medical University of South Carolina Charleston SC
| | - Melissa Lamicq
- Division of Cardiology Medical University of South Carolina Charleston SC
| | - Michael R Zile
- Division of Cardiology Medical University of South Carolina Charleston SC.,Ralph J. Johnson Veterans Affairs Medical Center Charleston SC
| | - Robert Malcolm
- Department of Psychiatry and Behavioral Sciences Weight Management Center Medical University of South Carolina Charleston SC
| | - Patrick M O'Neil
- Department of Psychiatry and Behavioral Sciences Weight Management Center Medical University of South Carolina Charleston SC
| | - Sheldon E Litwin
- Division of Cardiology Medical University of South Carolina Charleston SC.,Ralph J. Johnson Veterans Affairs Medical Center Charleston SC
| |
Collapse
|
104
|
Salah HM, Pandey A, Soloveva A, Abdelmalek MF, Diehl AM, Moylan CA, Wegermann K, Rao VN, Hernandez AF, Tedford RJ, Parikh KS, Mentz RJ, McGarrah RW, Fudim M. Relationship of Nonalcoholic Fatty Liver Disease and Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2021; 6:918-932. [PMID: 34869957 PMCID: PMC8617573 DOI: 10.1016/j.jacbts.2021.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022]
Abstract
Although there is an established bidirectional relationship between heart failure with reduced ejection fraction and liver disease, the association between heart failure with preserved ejection fraction (HFpEF) and liver diseases, such as nonalcoholic fatty liver disease (NAFLD), has not been well explored. In this paper, the authors provide an in-depth review of the relationship between HFpEF and NAFLD and propose 3 NAFLD-related HFpEF phenotypes (obstructive HFpEF, metabolic HFpEF, and advanced liver fibrosis HFpEF). The authors also discuss diagnostic challenges related to the concurrent presence of NAFLD and HFpEF and offer several treatment options for NAFLD-related HFpEF phenotypes. The authors propose that NAFLD-related HFpEF should be recognized as a distinct HFpEF phenotype.
Collapse
Key Words
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- AV, arteriovenous
- BCAA, branched-chain amino acid
- GLP, glucagon-like peptide
- HF, heart failure
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IL, interleukin
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- NAFLD
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NT-proBNP, N terminal pro–B-type natriuretic peptide
- RAAS, renin-angiotensin aldosterone system
- SGLT2, sodium-glucose cotransporter 2
- SPSS, spontaneous portosystemic shunt(s)
- TNF, tumor necrosis factor
- cardiomyopathy
- heart failure
- liver
Collapse
Affiliation(s)
- Husam M. Salah
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Medicine, University of Texas Southwestern, and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Anzhela Soloveva
- Department of Cardiology, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Manal F. Abdelmalek
- Division of Gastroenterology and Hepatology, Duke University, Durham, North Carolina, USA
| | - Anna Mae Diehl
- Division of Gastroenterology and Hepatology, Duke University, Durham, North Carolina, USA
| | - Cynthia A. Moylan
- Division of Gastroenterology and Hepatology, Duke University, Durham, North Carolina, USA
| | - Kara Wegermann
- Division of Gastroenterology and Hepatology, Duke University, Durham, North Carolina, USA
| | - Vishal N. Rao
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Adrian F. Hernandez
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Ryan J. Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kishan S. Parikh
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Robert J. Mentz
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Robert W. McGarrah
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Marat Fudim
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
105
|
Choi RH, Tatum SM, Symons JD, Summers SA, Holland WL. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat Rev Cardiol 2021; 18:701-711. [PMID: 33772258 PMCID: PMC8978615 DOI: 10.1038/s41569-021-00536-1] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 02/03/2023]
Abstract
Increases in calorie consumption and sedentary lifestyles are fuelling a global pandemic of cardiometabolic diseases, including coronary artery disease, diabetes mellitus, cardiomyopathy and heart failure. These lifestyle factors, when combined with genetic predispositions, increase the levels of circulating lipids, which can accumulate in non-adipose tissues, including blood vessel walls and the heart. The metabolism of these lipids produces bioactive intermediates that disrupt cellular function and survival. A compelling body of evidence suggests that sphingolipids, such as ceramides, account for much of the tissue damage in these cardiometabolic diseases. In humans, serum ceramide levels are proving to be accurate biomarkers of adverse cardiovascular disease outcomes. In mice and rats, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of diabetes, atherosclerosis, hypertension and heart failure. In cultured cells and isolated tissues, ceramides perturb mitochondrial function, block fuel usage, disrupt vasodilatation and promote apoptosis. In this Review, we discuss the body of literature suggesting that ceramides are drivers - and not merely passengers - on the road to cardiovascular disease. Moreover, we explore the feasibility of therapeutic strategies to lower ceramide levels to improve cardiovascular health.
Collapse
Affiliation(s)
- Ran Hee Choi
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
106
|
Dumeny L, Vardeny O, Edelmann F, Pieske B, Duarte JD, Cavallari LH. NR3C2 genotype is associated with response to spironolactone in diastolic heart failure patients from the Aldo-DHF trial. Pharmacotherapy 2021; 41:978-987. [PMID: 34569641 DOI: 10.1002/phar.2626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/21/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023]
Abstract
STUDY OBJECTIVE This study aimed to determine if variants in NR3C2, which codes the target protein of spironolactone, or CYP11B2, which is involved in aldosterone synthesis, were associated with spironolactone response, focused on the primary end point of diastolic function (E/e'), in Aldosterone Receptor Blockade in Diastolic Heart Failure (Aldo-DHF) participants. DESIGN Post-hoc genetic analysis. DATA SOURCE Data and samples were derived from the multi-center, randomized, double-blind, placebo-controlled Aldo-DHF trial. PATIENTS Aldo-DHF participants treated with spironolactone (n = 184) or placebo (n = 178) were included. INTERVENTION Participants were genotyped for NR3C2 rs5522, NR3C2 rs2070951 and CYP11B2 rs1799998 via pyrosequencing. MEASUREMENTS In the placebo and spironolactone arms, separate multivariable linear regression analyses were performed for change in E/e' with each single nucleotide polymorphism (SNP), adjusted for age, sex, and baseline E/e'. To discern potential mechanisms of a genotype effect, associated SNPs were further examined for their association with change in blood pressure, circulating procollagen type III N-terminal peptide (PIIINP), and left atrial area. MAIN RESULTS Carriers of the rs5522 G allele in the placebo arm had a greater increase in E/e' over the 12-month course of the trial compared to noncarriers (β = 1.10; 95% confidence interval [CI]: 0.05-2.16; p = 0.04). No corresponding E/e' worsening by rs5522 genotype was observed in the spironolactone arm. None of the other genotypes were associated with change in E/e'. Compared to noncarriers, rs5522 G carriers also had a greater increase in left atrial area with placebo (β = 0.83; 95% CI: 0.17-1.48; p = 0.01) and a greater reduction in diastolic blood pressure with spironolactone (β = -3.56; 95% CI: -6.73 to -0.39; p = 0.03). Serum PIIINP levels were similar across rs5522 genotypes. CONCLUSIONS Our results suggest that spironolactone attenuates progression of diastolic dysfunction associated with the NR3C2 rs5522 G allele. Validation of our findings is needed.
Collapse
Affiliation(s)
- Leanne Dumeny
- Center for Pharmacogenomics and Precision Medicine and Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics, Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Orly Vardeny
- Center for Care Delivery and Outcomes Research, Minneapolis Veteran Affairs Health Care System, Minneapolis, Minnesota, USA
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Julio D Duarte
- Center for Pharmacogenomics and Precision Medicine and Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics, Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Larisa H Cavallari
- Center for Pharmacogenomics and Precision Medicine and Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics, Genetics Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
107
|
Forsyth F, Brimicombe J, Cheriyan J, Edwards D, Hobbs FR, Jalaludeen N, Mant J, Pilling M, Schiff R, Taylor CJ, Zaman MJ, Deaton C, Chakravorty M, Maclachlan S, Kane E, Odone J, Thorley N, Borja‐Boluda S, Wellwood I, Sowden E, Blakeman T, Chew‐Graham C, Hossain M, Sharpley J, Gordon B, Taffe J, Long A, Aziz A, Swayze H, Rutter H, Schramm C, MacDonald S, Papworth H, Smith J, Needs C, Cronk D, Newark C, Blake D, Brown A, Basuita A, Gayton E, Glover V, Fox R, Crawshaw J, Ashdown H, A'Court C, Ayerst R, Hernandez‐Diaz B, Knox K, Wooding N, Wanninayake S, Keast C, Jones A, Brown K, Gaw M, Thomas N, Dixon S, Angeleri‐Rand E. Diagnosis of patients with heart failure with preserved ejection fraction in primary care: cohort study. ESC Heart Fail 2021. [PMCID: PMC8712851 DOI: 10.1002/ehf2.13612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aims Heart failure with preserved ejection fraction (HFpEF) accounts for half of all heart failure (HF), but low awareness and diagnostic challenges hinder identification in primary care. Our aims were to evaluate the recruitment and diagnostic strategy in the Optimise HFpEF cohort and compare with recent recommendations for diagnosing HFpEF. Methods and results Patients were recruited from 30 primary care practices in two regions in England using an electronic screening algorithm and two secondary care sites. Baseline assessment collected clinical and patient‐reported data and diagnosis by history, assessment, and trans‐thoracic echocardiogram (TTE). A retrospective evaluation compared study diagnosis with H2FPEF score and HFA‐PEFF diagnostic algorithm. A total of 152 patients (86% primary care, mean age 78.5, 40% female) were enrolled; 93 (61%) had HFpEF confirmed. Most participants had clinical features of HFpEF, but those with confirmed HFpEF were more likely female, obese, functionally impaired, and symptomatic. Some echocardiographic findings were diagnostic for HFpEF, but no difference in natriuretic peptide levels were observed. The H2FPEF and HFA‐PEFF scores were not significantly different by group, although confirmed HFpEF cases were more likely to have scores indicating high probability of HFpEF. Conclusions Patients with HFpEF in primary care are difficult to identify, and greater awareness of the condition, with clear diagnostic pathways and specialist support, are needed. Use of diagnostic algorithms and scores can provide systematic approaches to diagnosis but may be challenging to apply in older multi‐morbid patients. Where diagnostic uncertainty remains, pragmatic decisions are needed regarding the value of additional testing versus management of presumptive HFpEF.
Collapse
Affiliation(s)
- Faye Forsyth
- Primary Care Unit, Department of Public Health and Primary Care University of Cambridge School of Clinical Medicine East Forvie, Cambridge Biomedical Campus Cambridge CB2 0SR UK
| | - James Brimicombe
- Primary Care Unit, Department of Public Health and Primary Care University of Cambridge School of Clinical Medicine East Forvie, Cambridge Biomedical Campus Cambridge CB2 0SR UK
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine University of Cambridge School of Clinical Medicine Cambridge UK
| | - Duncan Edwards
- Primary Care Unit, Department of Public Health and Primary Care University of Cambridge School of Clinical Medicine East Forvie, Cambridge Biomedical Campus Cambridge CB2 0SR UK
| | - F.D. Richard Hobbs
- Nuffield Department of Primary Care Health Sciences University of Oxford Oxford UK
| | - Navazh Jalaludeen
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine University of Cambridge School of Clinical Medicine Cambridge UK
| | - Jonathan Mant
- Primary Care Unit, Department of Public Health and Primary Care University of Cambridge School of Clinical Medicine East Forvie, Cambridge Biomedical Campus Cambridge CB2 0SR UK
| | - Mark Pilling
- Primary Care Unit, Department of Public Health and Primary Care University of Cambridge School of Clinical Medicine East Forvie, Cambridge Biomedical Campus Cambridge CB2 0SR UK
| | - Rebekah Schiff
- Department of Ageing and Health Guy's and St. Thomas' NHS Foundation Trust London UK
| | - Clare J. Taylor
- Nuffield Department of Primary Care Health Sciences University of Oxford Oxford UK
| | - M. Justin Zaman
- Department of Cardiology West Suffolk Hospital Bury St Edmonds UK
| | - Christi Deaton
- Primary Care Unit, Department of Public Health and Primary Care University of Cambridge School of Clinical Medicine East Forvie, Cambridge Biomedical Campus Cambridge CB2 0SR UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Rosalia L, Ozturk C, Shoar S, Fan Y, Malone G, Cheema FH, Conway C, Byrne RA, Duffy GP, Malone A, Roche ET, Hameed A. Device-Based Solutions to Improve Cardiac Physiology and Hemodynamics in Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2021; 6:772-795. [PMID: 34754993 PMCID: PMC8559325 DOI: 10.1016/j.jacbts.2021.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022]
Abstract
Characterized by a rapidly increasing prevalence, elevated mortality and rehospitalization rates, and inadequacy of pharmaceutical therapies, heart failure with preserved ejection fraction (HFpEF) has motivated the widespread development of device-based solutions. HFpEF is a multifactorial disease of various etiologies and phenotypes, distinguished by diminished ventricular compliance, diastolic dysfunction, and symptoms of heart failure despite a normal ejection performance; these symptoms include pulmonary hypertension, limited cardiac reserve, autonomic imbalance, and exercise intolerance. Several types of atrial shunts, left ventricular expanders, stimulation-based therapies, and mechanical circulatory support devices are currently under development aiming to target one or more of these symptoms by addressing the associated mechanical or hemodynamic hallmarks. Although the majority of these solutions have shown promising results in clinical or preclinical studies, no device-based therapy has yet been approved for the treatment of patients with HFpEF. The purpose of this review is to discuss the rationale behind each of these devices and the findings from the initial testing phases, as well as the limitations and challenges associated with their clinical translation.
Collapse
Key Words
- BAT, baroreceptor activation therapy
- CCM, cardiac contractility modulation
- CRT, cardiac resynchronization therapy
- HF, heart failure
- HFmEF, heart failure with mid-range ejection fraction
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IASD, Interatrial Shunt Device
- LAAD, left atrial assist device
- LAP, left atrial pressure
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MCS, mechanical circulatory support
- NYHA, New York Heart Association
- PCWP, pulmonary capillary wedge pressure
- QoL, quality of life
- TAA, transapical approach
- atrial shunt devices
- electrostimulation
- heart failure devices
- heart failure with preserved ejection fraction
- left ventricular expanders
- mechanical circulatory support
- neuromodulation
Collapse
Affiliation(s)
- Luca Rosalia
- Health Sciences and Technology Program, Harvard–Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Caglar Ozturk
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Grainne Malone
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Faisal H. Cheema
- HCA Healthcare, Houston, Texas, USA
- University of Houston, College of Medicine, Houston, Texas, USA
| | - Claire Conway
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Robert A. Byrne
- Department of Cardiology, Mater Private Hospital, Dublin, Ireland
- Cardiovascular Research, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Garry P. Duffy
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, Galway, Ireland
- Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
- Advanced Materials for Biomedical Engineering and Regenerative Medicine, Trinity College Dublin, and National University of Ireland Galway, Galway, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Andrew Malone
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ellen T. Roche
- Health Sciences and Technology Program, Harvard–Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
109
|
Natterson-Horowitz B, Baccouche BM, Mary J, Shivkumar T, Bertelsen MF, Aalkjær C, Smerup MH, Ajijola OA, Hadaya J, Wang T. Did giraffe cardiovascular evolution solve the problem of heart failure with preserved ejection fraction? EVOLUTION MEDICINE AND PUBLIC HEALTH 2021; 9:248-255. [PMID: 34447575 PMCID: PMC8385250 DOI: 10.1093/emph/eoab016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 06/04/2021] [Indexed: 11/18/2022]
Abstract
The evolved adaptations of other species can be a source of insight for novel biomedical innovation. Limitations of traditional animal models for the study of some pathologies are fueling efforts to find new approaches to biomedical investigation. One emerging approach recognizes the evolved adaptations in other species as possible solutions to human pathology. The giraffe heart, for example, appears resistant to pathology related to heart failure with preserved ejection fraction (HFpEF)—a leading form of hypertension-associated cardiovascular disease in humans. Here, we postulate that the physiological pressure-induced left ventricular thickening in giraffes does not result in the pathological cardiovascular changes observed in humans with hypertension. The mechanisms underlying this cardiovascular adaptation to high blood pressure in the giraffe may be a bioinspired roadmap for preventive and therapeutic strategies for human HFpEF.
Collapse
Affiliation(s)
- Barbara Natterson-Horowitz
- Department of Medicine, Harvard Medical School, Boston, MA, USA.,Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.,Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Basil M Baccouche
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.,Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jennifer Mary
- Zoobiquity Research Initiative at UCLA, Los Angeles, CA 90024, USA
| | | | | | | | - Morten H Smerup
- Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joseph Hadaya
- UCLA Cardiac Arrhythmia Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Molecular, Cellular and Integrative Physiology Program, UCLA, Los Angeles, CA, USA
| | - Tobias Wang
- Zoophysiology, Department of Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
110
|
Withaar C, Lam CSP, Schiattarella GG, de Boer RA, Meems LMG. Heart failure with preserved ejection fraction in humans and mice: embracing clinical complexity in mouse models. Eur Heart J 2021; 42:4420-4430. [PMID: 34414416 PMCID: PMC8599003 DOI: 10.1093/eurheartj/ehab389] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/15/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a multifactorial disease accounting for a large and increasing proportion of all clinical HF presentations. As a clinical syndrome, HFpEF is characterized by typical signs and symptoms of HF, a distinct cardiac phenotype and raised natriuretic peptides. Non-cardiac comorbidities frequently co-exist and contribute to the pathophysiology of HFpEF. To date, no therapy has proven to improve outcomes in HFpEF, with drug development hampered, at least partly, by lack of consensus on appropriate standards for pre-clinical HFpEF models. Recently, two clinical algorithms (HFA-PEFF and H2FPEF scores) have been developed to improve and standardize the diagnosis of HFpEF. In this review, we evaluate the translational utility of HFpEF mouse models in the context of these HFpEF scores. We systematically recorded evidence of symptoms and signs of HF or clinical HFpEF features and included several cardiac and extra-cardiac parameters as well as age and sex for each HFpEF mouse model. We found that most of the pre-clinical HFpEF models do not meet the HFpEF clinical criteria, although some multifactorial models resemble human HFpEF to a reasonable extent. We therefore conclude that to optimize the translational value of mouse models to human HFpEF, a novel approach for the development of pre-clinical HFpEF models is needed, taking into account the complex HFpEF pathophysiology in humans.
Collapse
Affiliation(s)
- Coenraad Withaar
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Carolyn S P Lam
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.,National University Heart Centre, Singapore and Duke-National University of Singapore
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Department of Cardiology, Center for Cardiovascular Research (CCR), Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Laura M G Meems
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
111
|
Fusco-Allison G, Li DK, Hunter B, Jackson D, Bannon PG, Lal S, O'Sullivan JF. Optimizing the discovery and assessment of therapeutic targets in heart failure with preserved ejection fraction. ESC Heart Fail 2021; 8:3643-3655. [PMID: 34342166 PMCID: PMC8497375 DOI: 10.1002/ehf2.13504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 01/09/2023] Open
Abstract
There is an urgent need for models that faithfully replicate heart failure with preserved ejection fraction (HFpEF), now recognized as the most common form of heart failure in the world. In vitro approaches have several shortcomings, most notably the immature nature of stem cell‐derived human cardiomyocytes [induced pluripotent stem cells (iPSC)] and the relatively short lifespan of primary cardiomyocytes. Three‐dimensional ‘organoids’ incorporating mature iPSCs with other cell types such as endothelial cells and fibroblasts are a significant advance, but lack the complexity of true myocardium. Animal models can replicate many features of human HFpEF, and rodent models are the most common, and recent attempts to incorporate haemodynamic, metabolic, and ageing contributions are encouraging. Differences relating to species, physiology, heart rate, and heart size are major limitations for rodent models. Porcine models mitigate many of these shortcomings and approximate human physiology more closely, but cost and time considerations limit their potential for widespread use. Ex vivo analysis of failing hearts from animal models offer intriguing possibilities regarding cardiac substrate utilisation, but are ultimately subject to the same constrains as the animal models from which the hearts are obtained. Ex vivo approaches using human myocardial biopsies can uncover new insights into pathobiology leveraging myocardial energetics, substrate turnover, molecular changes, and systolic/diastolic function. In collaboration with a skilled cardiothoracic surgeon, left ventricular endomyocardial biopsies can be obtained at the time of valvular surgery in HFpEF patients. Critically, these tissues maintain their disease phenotype, preserving inter‐relationship of myocardial cells and extracellular matrix. This review highlights a novel approach, where ultra‐thin myocardial tissue slices from human HFpEF hearts can be used to assess changes in myocardial structure and function. We discuss current approaches to modelling HFpEF, describe in detail the novel tissue slice model, expand on exciting opportunities this model provides, and outline ways to improve this model further.
Collapse
Affiliation(s)
- Gabrielle Fusco-Allison
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond K Li
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Dan Jackson
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul G Bannon
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Faculty of Medicine, TU Dresden, Dresden, Germany
| |
Collapse
|
112
|
Sousa LS, Nascimento FDA, Rocha J, Rocha-Parise M. Cardioprotective Effects of Sodium-glucose Cotransporter 2 Inhibitors Regardless of Type 2 Diabetes Mellitus: A Meta-analysis. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2021. [DOI: 10.36660/ijcs.20200339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
113
|
Difference in Prognosis between Continuation and Discontinuation of A 5-Month Cardiac Rehabilitation Program in Outpatients with Heart Failure with Preserved Ejection Fraction. J Clin Med 2021; 10:jcm10153306. [PMID: 34362090 PMCID: PMC8348181 DOI: 10.3390/jcm10153306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Cardiac rehabilitation (CR) is a requisite component of care for patients with heart failure (HF). We aimed to evaluate the clinical outcomes in outpatients with HF with preserved ejection fraction (HFpEF) compared to those in patients with non-HFpEF who did and did not continue a 5-month CR program. Methods: 173 outpatients with HF who participated in a 5-month CR program were registered. We divided them into two groups: HFpEF (n = 84, EF 63 ± 7%) and non-HFpEF (n = 89, EF 31 ± 11%). We further divided the patients into those who continued the CR program (continued group) and those who did not (discontinued group) in the HFpEF and non-HFpEF groups. The clinical outcomes at 5 months were compared among the groups. Results: There were no significant differences in patient characteristics at baseline between the continued and discontinued groups in the HFpEF and non-HFpEF groups except for % diabetes mellitus in the non-HFpEF group. The rates of all-cause death and hospital admissions in the continued group in both the HFpEF and non-HFpEF groups were significantly lower than those in the discontinued group. The all-cause death and hospital admissions in each group were independently associated with the continuation of the CR program. Conclusions: The continuation of a 5-month CR program was associated with the prevention of all-cause death and hospital admissions in both the HFpEF and non-HFpEF groups.
Collapse
|
114
|
Loureiro TN, Valete COS, Castier MB, Leite MDFM, Sztajnbok FR. Analysis of Diastolic Left Ventricular Function in Adolescents with Juvenile Systemic Lupus Erythematosus. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2021. [DOI: 10.36660/ijcs.20200210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
115
|
Effects of exercise training in heart failure with preserved ejection fraction: an updated systematic literature review. Heart Fail Rev 2021; 25:703-711. [PMID: 31399956 DOI: 10.1007/s10741-019-09841-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Physical activity is associated with a lower risk of adverse cardiovascular outcomes, including heart failure (HF). Exercise training is a class IA level recommendation in patients with stable HF, but its impact is less clear in heart failure with preserved ejection fraction (HFpEF). The aim of this study was to analyze the effects of the exercise training on cardiovascular outcomes in patients with HFpEF. A systematic literature search was conducted on the main electronic databases, proceedings of major meetings, and reference lists of the identified studies, using specific terms for only English language studies published between 2000 and 2018. We followed the PRISMA to perform our review. Quality of studies was also assessed. The systematic review identified 9 studies on 348 patients, of moderate (n = 2) to good (n = 7) quality. The training consisted of a combination of supervised in-hospital and home-based outpatient programs, including aerobic exercise, endurance and resistance training, walking, and treadmill and bicycle ergometer. Most of the protocols ranged 12-16 weeks, with a frequency of 2-3 sessions weekly, lasting 20-60 min per session. There were significant improvements in peak oxygen uptake, 6-min walking test distance, and ventilatory threshold, whereas quality of life and echocardiographic parameters improved only in some studies. Endothelial function/arterial stiffness remained unchanged. No adverse events were reported. Appropriate exercise programs are able to get a favorable cardiovascular outcome in patients with HFpEF. This could also benefit in terms of quality of life, even if more controversial. Further researches are necessary.
Collapse
|
116
|
Berdy AE, Upadhya B, Ponce S, Swett K, Stacey RB, Kaplan R, Vasquez PM, Qi Q, Schneiderman N, Hurwitz BE, Daviglus ML, Kansal M, Evenson KR, Rodriguez CJ. Associations between physical activity, sedentary behaviour and left ventricular structure and function from the Echocardiographic Study of Latinos (ECHO-SOL). Open Heart 2021; 8:e001647. [PMID: 34261776 PMCID: PMC8311330 DOI: 10.1136/openhrt-2021-001647] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/15/2021] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE The cross-sectional association between accelerometer-measured physical activity (PA), sedentary behaviour (SB) and cardiac structure and function is less well described. This study's primary aim was to compare echocardiographic measures of cardiac structure and function with accelerometer measured PA and SB. METHODS Participants included 1206 self-identified Hispanic/Latino men and women, age 45-74 years, from the Echocardiographic Study of Latinos. Standard echocardiographic measures included M-mode, two-dimensional, spectral, tissue Doppler and myocardial strain. Participants wore an Actical accelerometer at the hip for 1 week. RESULTS The mean±SE age for the cohort was 56±0.4 years, 57% were women. Average moderate to vigorous PA (MVPA) was 21±1.1 min/day, light PA was 217±4.2 min/day and SB was 737±8.1 min/day. Both higher levels of light PA and MVPA (min/day) were associated with lower left ventricular (LV) mass index (LVMI)/end-diastolic volume and a lower E/e' ratio. Higher levels of MVPA (min/day) were associated with better right ventricular systolic function. Higher levels of SB were associated with increased LVMI. In a multivariable linear regression model adjusted for demographics and cardiovascular disease modifiable factors, every 10 additional min/day of light PA was associated with a 0.03 mL/m2 increase in left atrial volume index (LAVI) (p<0.01) and a 0.004 cm increase in tricuspid annular plane systolic excursion (p<0.01); every 10 additional min/day of MVPA was associated with a 0.18 mL/m2 increase in LAVI (p<0.01) and a 0.24% improvement in global circumferential strain (p<0.01). CONCLUSIONS Our findings highlight the potential positive association between the MVPA and light PA on cardiac structure and function.
Collapse
Affiliation(s)
- Andrew E Berdy
- Cardiology, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Bharathi Upadhya
- Cardiology, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Sonia Ponce
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
| | - Katrina Swett
- Department of Medicine, Epidemiology & Population Health, Albert Einstein College of Medicine Department of Neurology, Bronx, New York, USA
| | - Richard B Stacey
- Cardiology, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Robert Kaplan
- Department of Medicine, Epidemiology & Population Health, Albert Einstein College of Medicine Department of Neurology, Bronx, New York, USA
| | - Priscilla M Vasquez
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
| | - Qibin Qi
- Department of Medicine, Epidemiology & Population Health, Albert Einstein College of Medicine Department of Neurology, Bronx, New York, USA
| | - Neil Schneiderman
- Department of Psychology, University of Miami, Coral Gables, Florida, USA
| | - Barry E Hurwitz
- Department of Psychology, University of Miami, Coral Gables, Florida, USA
| | - Martha L Daviglus
- Cardiovascular Medicine, University of Illinois Hospital and Health Sciences, Chicago, Illinois, USA
| | - Mayank Kansal
- Cardiovascular Medicine, University of Illinois Hospital and Health Sciences, Chicago, Illinois, USA
| | - Kelly R Evenson
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina - Chapel Hill, Chapel Hill, North Carolina, USA
| | - Carlos J Rodriguez
- Department of Medicine, Epidemiology & Population Health, Albert Einstein College of Medicine Department of Neurology, Bronx, New York, USA
| |
Collapse
|
117
|
Abstract
Exercise intolerance represents a typical feature of heart failure with preserved ejection fraction (HFpEF), and is associated with a poor quality of life, frequent hospitalizations, and increased all-cause mortality. The cardiopulmonary exercise test is the best method to quantify exercise intolerance, and allows detection of the main mechanism responsible for the exercise limitation, influencing treatment and prognosis. Exercise training programs improve exercise tolerance in HFpEF. However, studies are needed to identify appropriate type and duration. This article discusses the pathophysiology of exercise limitation in HFpEF, describes methods of determining exercise tolerance class, and evaluates prognostic implications and potential therapeutic strategies.
Collapse
|
118
|
Gentile F, Ghionzoli N, Borrelli C, Vergaro G, Pastore MC, Cameli M, Emdin M, Passino C, Giannoni A. Epidemiological and clinical boundaries of heart failure with preserved ejection fraction. Eur J Prev Cardiol 2021; 29:1233-1243. [PMID: 33963839 DOI: 10.1093/eurjpc/zwab077] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is highly prevalent and is associated with relevant morbidity and mortality. However, an evidence-based treatment is still absent. The heterogeneous definitions, differences in aetiology/pathophysiology, and diagnostic challenges of HFpEF made it difficult to define its epidemiological landmarks so far. Several large registries and observational studies have recently disclosed an increasing incidence/prevalence, as well as its prognostic significance. An accurate definition of HFpEF epidemiological boundaries and phenotypes is mandatory to develop novel effective and rational therapeutic approaches.
Collapse
Affiliation(s)
- Francesco Gentile
- Department of Cardiology and Cardiovascular Medicine, Fondazione Toscana G. Monasterio, Pisa 56124, Italy.,Cardiothoracic Department, Cardiology Division, University Hospital of Pisa, Pisa 56124, Italy
| | - Nicolò Ghionzoli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena 53100, Italy
| | - Chiara Borrelli
- Department of Cardiology and Cardiovascular Medicine, Fondazione Toscana G. Monasterio, Pisa 56124, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, Pisa 56127, Italy
| | - Giuseppe Vergaro
- Department of Cardiology and Cardiovascular Medicine, Fondazione Toscana G. Monasterio, Pisa 56124, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, Pisa 56127, Italy
| | - Maria Concetta Pastore
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena 53100, Italy
| | - Matteo Cameli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena 53100, Italy
| | - Michele Emdin
- Department of Cardiology and Cardiovascular Medicine, Fondazione Toscana G. Monasterio, Pisa 56124, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, Pisa 56127, Italy
| | - Claudio Passino
- Department of Cardiology and Cardiovascular Medicine, Fondazione Toscana G. Monasterio, Pisa 56124, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, Pisa 56127, Italy
| | - Alberto Giannoni
- Department of Cardiology and Cardiovascular Medicine, Fondazione Toscana G. Monasterio, Pisa 56124, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, Pisa 56127, Italy
| |
Collapse
|
119
|
Chen MS, Lee RT, Garbern JC. Senescence mechanisms and targets in the heart. Cardiovasc Res 2021; 118:1173-1187. [PMID: 33963378 DOI: 10.1093/cvr/cvab161] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/27/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Cellular senescence is a state of irreversible cell cycle arrest associated with ageing. Senescence of different cardiac cell types can direct the pathophysiology of cardiovascular diseases such as atherosclerosis, myocardial infarction, and cardiac fibrosis. While age-related telomere shortening represents a major cause of replicative senescence, the senescent state can also be induced by oxidative stress, metabolic dysfunction, and epigenetic regulation, among other stressors. It is critical that we understand the molecular pathways that lead to cellular senescence and the consequences of cellular senescence in order to develop new therapeutic approaches to treat cardiovascular disease. In this review, we discuss molecular mechanisms of cellular senescence, explore how cellular senescence of different cardiac cell types (including cardiomyocytes, cardiac endothelial cells, cardiac fibroblasts, vascular smooth muscle cells, valve interstitial cells) can lead to cardiovascular disease, and highlight potential therapeutic approaches that target molecular mechanisms of cellular senescence to prevent or treat cardiovascular disease.
Collapse
Affiliation(s)
- Maggie S Chen
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138.,Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115
| | - Jessica C Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138.,Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115
| |
Collapse
|
120
|
Yuan S, Schmidt HM, Wood KC, Straub AC. CoenzymeQ in cellular redox regulation and clinical heart failure. Free Radic Biol Med 2021; 167:321-334. [PMID: 33753238 DOI: 10.1016/j.freeradbiomed.2021.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Coenzyme Q (CoQ) is ubiquitously embedded in lipid bilayers of various cellular organelles. As a redox cycler, CoQ shuttles electrons between mitochondrial complexes and extramitochondrial reductases and oxidases. In this way, CoQ is crucial for maintaining the mitochondrial function, ATP synthesis, and redox homeostasis. Cardiomyocytes have a high metabolic rate and rely heavily on mitochondria to provide energy. CoQ levels, in both plasma and the heart, correlate with heart failure in patients, indicating that CoQ is critical for cardiac function. Moreover, CoQ supplementation in clinics showed promising results for treating heart failure. This review provides a comprehensive view of CoQ metabolism and its interaction with redox enzymes and reactive species. We summarize the clinical trials and applications of CoQ in heart failure and discuss the caveats and future directions to improve CoQ therapeutics.
Collapse
Affiliation(s)
- Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heidi M Schmidt
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
121
|
Tromp J, Claggett BL, Liu J, Jackson AM, Jhund PS, Køber L, Widimský J, Boytsov SA, Chopra VK, Anand IS, Ge J, Chen CH, Maggioni AP, Martinez F, Packer M, Pfeffer MA, Pieske B, Redfield MM, Rouleau JL, Van Veldhuisen DJ, Zannad F, Zile MR, Rizkala AR, Inubushi-Molessa A, Lefkowitz MP, Shi VC, McMurray JJV, Solomon SD, Lam CSP. Global Differences in Heart Failure With Preserved Ejection Fraction: The PARAGON-HF Trial. Circ Heart Fail 2021; 14:e007901. [PMID: 33866828 DOI: 10.1161/circheartfailure.120.007901] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a global public health problem with important regional differences. We investigated these differences in the PARAGON-HF trial (Prospective Comparison of Angiotensin Receptor Neprilysin Inhibitor With Angiotensin Receptor Blocker Global Outcomes in HFpEF), the largest and most inclusive global HFpEF trial. METHODS We studied differences in clinical characteristics, outcomes, and treatment effects of sacubitril/valsartan in 4796 patients with HFpEF from the PARAGON-HF trial, grouped according to geographic region. RESULTS Regional differences in patient characteristics and comorbidities were observed: patients from Western Europe were oldest (mean 75±7 years) with the highest prevalence of atrial fibrillation/flutter (36%); Central/Eastern European patients were youngest (mean 71±8 years) with the highest prevalence of coronary artery disease (50%); North American patients had the highest prevalence of obesity (65%) and diabetes (49%); Latin American patients were younger (73±9 years) and had a high prevalence of obesity (53%); and Asia-Pacific patients had a high prevalence of diabetes (44%), despite a low prevalence of obesity (26%). Rates of the primary composite end point of total hospitalizations for HF and death from cardiovascular causes were lower in patients from Central Europe (9 per 100 patient-years) and highest in patients from North America (28 per 100 patient-years), which was primarily driven by a greater number of total hospitalizations for HF. The effect of treatment with sacubitril-valsartan was not modified by region (interaction P>0.05). CONCLUSIONS Among patients with HFpEF recruited worldwide in PARAGON-HF, there were important regional differences in clinical characteristics and outcomes, which may have implications for the design of future clinical trials. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT01920711.
Collapse
Affiliation(s)
- Jasper Tromp
- National Heart Centre Singapore (J.T., C.S.P.L.).,Duke-NUS Medical School, Singapore (J.T., C.S.P.L.).,Department of Cardiology, University Medical Centre Groningen, University of Groningen, the Netherlands (J.T., D.J.V.V., C.S.P.L.).,Saw Swee Hock School of Public Health, National University of Singapore, Singapore (J.T.)
| | - Brian L Claggett
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.L.C., J.L., M.A.P., S.D.S.)
| | - Jiankang Liu
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.L.C., J.L., M.A.P., S.D.S.)
| | - Alice M Jackson
- British Heart Foundation Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (A.M.J., P.S.J., J.J.V.M.)
| | - Pardeep S Jhund
- British Heart Foundation Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (A.M.J., P.S.J., J.J.V.M.)
| | - Lars Køber
- Department of Cardiology, Heart Centre, Rigshospitalet, Copenhagen University Hospital, Denmark (L.K.)
| | - Jiří Widimský
- First Faculty of Medicine, Charles University Prague, Czech Republic (J.W.)
| | - Sergey A Boytsov
- National Research Center for Cardiology of the Ministry of Health of the Russian Federation, Moscow (S.B.)
| | - Vijay K Chopra
- Heart Failure and Research Max Super Specialty Hospital Saket, New Delhi, India (V.C.)
| | - Inder S Anand
- Department of Medicine, VA Medical Center and University of Minnesota, Minneapolis (I.S.A.)
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China (J.G.)
| | - Chen-Huan Chen
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China (C.-H.C.)
| | - Aldo P Maggioni
- Associazione Nazionale Medici Cardiologi Ospedalieri, Florence, Italy (A.P.M.)
| | | | - Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX (M.P.)
| | - Marc A Pfeffer
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.L.C., J.L., M.A.P., S.D.S.)
| | - Burkert Pieske
- Department of Internal Medicine, Cardiology Charité, Universitaetsmedizin Berlin, Campus Virchow Klinikum Berlin, Germany (B.P.)
| | | | - Jean L Rouleau
- Institut de Cardiologie de Montréal, Université de Montréal, QC, Canada (J.L.R.)
| | - Dirk J Van Veldhuisen
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, the Netherlands (J.T., D.J.V.V., C.S.P.L.)
| | - Faiez Zannad
- Inserm CIC 1433 and Université de Lorraine, Centre Hospitalier Régional Universitaire, Nancy, France (F.Z.)
| | - Michael R Zile
- Medical University of South Carolina and Ralph H. Johnson Veterans Administration Medical Center, Charleston (M.R.Z.)
| | - Adel R Rizkala
- Novartis Pharmaceuticals Corporation, East Hanover, NJ (A.R.R., A.I.-M., M.P.L., V.C.S.)
| | - Akiko Inubushi-Molessa
- Novartis Pharmaceuticals Corporation, East Hanover, NJ (A.R.R., A.I.-M., M.P.L., V.C.S.)
| | - Martin P Lefkowitz
- Novartis Pharmaceuticals Corporation, East Hanover, NJ (A.R.R., A.I.-M., M.P.L., V.C.S.)
| | - Victor C Shi
- Novartis Pharmaceuticals Corporation, East Hanover, NJ (A.R.R., A.I.-M., M.P.L., V.C.S.)
| | - John J V McMurray
- British Heart Foundation Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (A.M.J., P.S.J., J.J.V.M.)
| | - Scott D Solomon
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.L.C., J.L., M.A.P., S.D.S.)
| | - Carolyn S P Lam
- National Heart Centre Singapore (J.T., C.S.P.L.).,Duke-NUS Medical School, Singapore (J.T., C.S.P.L.).,Department of Cardiology, University Medical Centre Groningen, University of Groningen, the Netherlands (J.T., D.J.V.V., C.S.P.L.)
| | | |
Collapse
|
122
|
Abstract
Purpose of review Heart failure with preserved ejection fraction (HFpEF) is a complex and heterogeneous condition of multiple causes, characterized by a clinical syndrome resulting from elevated left ventricular filling pressures, with an apparently unimpaired left ventricular systolic function. Although HFpEF has been long recognized as a distinct entity with significant morbidity for patients, its diagnosis remains challenging to this day. In recent years, few diagnostic algorithms have been postulated to aid in the identification of this condition. Invasive hemodynamic and metabolic evaluation is often warranted for the conclusive diagnosis and risk stratification of HFpEF, in patients presenting with undifferentiated DOE. Recent findings Rest and provoked hemodynamics remain the golden-standard diagnostic tool to unequivocally confirm the diagnosis of both established and incipient HFpEF, respectively. Cycle exercise hemodynamics is the paramount provocative maneuver to unveil this condition. Rapid saline loading does not offer a significant benefit over that of cycle exercise. Vasoactive agents can also uncover and confirm incipient HFpEF disease. The role of metabolic evaluation in patients presenting with idiopathic dyspnea on exertion (DOE) is of unparalleled value for those who have expertise in cardiopulmonary exercise test (CPET) interpretation; however, the average clinician who focuses solely on oxygen consumption will find it underwhelming. Invasive CPET stands alone as the ultimate diagnostic tool to discriminate between pulmonary, cardiovascular, and skeletal muscle disorders, and their respective contribution to DOE and exercise intolerance. Summary Several hemodynamic and metabolic parameters have demonstrated not only strong diagnostic value, but also predictive power in HFpEF. Additionally, these diagnostic methods have given rise to several therapeutic interventions that are now part of our clinical armamentarium. Regrettably, due to the heterogeneity and multicausality of HFpEF, none of the targeted interventions have been so far successful in decreasing the mortality burden of this prevalent condition.
Collapse
|
123
|
Paitazoglou C, Bergmann MW. The atrial flow regulator: current overview on technique and first experience. Ther Adv Cardiovasc Dis 2021; 14:1753944720919577. [PMID: 32972299 PMCID: PMC7522821 DOI: 10.1177/1753944720919577] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Martin W Bergmann
- Interventional Cardiology, Cardiologicum Hamburg, Schloßgarten 3-7, Hamburg 22401, Germany
| |
Collapse
|
124
|
Zhou L, Guo Z, Wang B, Wu Y, Li Z, Yao H, Fang R, Yang H, Cao H, Cui Y. Risk Prediction in Patients With Heart Failure With Preserved Ejection Fraction Using Gene Expression Data and Machine Learning. Front Genet 2021; 12:652315. [PMID: 33828587 PMCID: PMC8019773 DOI: 10.3389/fgene.2021.652315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) has become a major health issue because of its high mortality, high heterogeneity, and poor prognosis. Using genomic data to classify patients into different risk groups is a promising method to facilitate the identification of high-risk groups for further precision treatment. Here, we applied six machine learning models, namely kernel partial least squares with the genetic algorithm (GA-KPLS), the least absolute shrinkage and selection operator (LASSO), random forest, ridge regression, support vector machine, and the conventional logistic regression model, to predict HFpEF risk and to identify subgroups at high risk of death based on gene expression data. The model performance was evaluated using various criteria. Our analysis was focused on 149 HFpEF patients from the Framingham Heart Study cohort who were classified into good-outcome and poor-outcome groups based on their 3-year survival outcome. The results showed that the GA-KPLS model exhibited the best performance in predicting patient risk. We further identified 116 differentially expressed genes (DEGs) between the two groups, thus providing novel therapeutic targets for HFpEF. Additionally, the DEGs were enriched in Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways related to HFpEF. The GA-KPLS-based HFpEF model is a powerful method for risk stratification of 3-year mortality in HFpEF patients.
Collapse
Affiliation(s)
- Liye Zhou
- Division of Health Management, School of Management, Shanxi Medical University, Taiyuan, China
| | - Zhifei Guo
- Division of Health Management, School of Management, Shanxi Medical University, Taiyuan, China
| | - Bijue Wang
- Division of Health Management, School of Management, Shanxi Medical University, Taiyuan, China
| | - Yongqing Wu
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Zhi Li
- Department of Hematology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongmei Yao
- Department of Cardiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiling Fang
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Haitao Yang
- Division of Health Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Hongyan Cao
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Major Disease Risk Assessment, Shanxi Medical University, Taiyuan, China
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
125
|
LaPenna KB, Polhemus DJ, Doiron JE, Hidalgo HA, Li Z, Lefer DJ. Hydrogen Sulfide as a Potential Therapy for Heart Failure-Past, Present, and Future. Antioxidants (Basel) 2021; 10:485. [PMID: 33808673 PMCID: PMC8003444 DOI: 10.3390/antiox10030485] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous, gaseous signaling molecule that plays a critical role in cardiac and vascular biology. H2S regulates vascular tone and oxidant defenses and exerts cytoprotective effects in the heart and circulation. Recent studies indicate that H2S modulates various components of metabolic syndrome, including obesity and glucose metabolism. This review will discuss studies exhibiting H2S -derived cardioprotective signaling in heart failure with reduced ejection fraction (HFrEF). We will also discuss the role of H2S in metabolic syndrome and heart failure with preserved ejection fraction (HFpEF).
Collapse
Affiliation(s)
- Kyle B. LaPenna
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David J. Polhemus
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Jake E. Doiron
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Hunter A. Hidalgo
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Zhen Li
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David J. Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
126
|
Fujiwara K, Shimada K, Nishitani-Yokoyama M, Kunimoto M, Matsubara T, Matsumori R, Abulimiti A, Aikawa T, Ouchi S, Shimizu M, Fukao K, Miyazaki T, Honzawa A, Yamada M, Saitoh M, Morisawa T, Takahashi T, Daida H, Minamino T. Arterial Stiffness Index and Exercise Tolerance in Patients Undergoing Cardiac Rehabilitation. Int Heart J 2021; 62:230-237. [PMID: 33731517 DOI: 10.1536/ihj.20-418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Arterial stiffness contributes to the development of cardiovascular disease (CVD). However, the relationship between the arterial stiffness and exercise tolerance in CVD patients with preserved ejection fraction (pEF) and those with reduced EF (rEF) is unclear. We enrolled 358 patients who participated in cardiac rehabilitation and underwent cardiopulmonary exercise testing at Juntendo University Hospital. After excluding 195 patients who had undergone open heart surgery and 20 patients with mid-range EF, the patients were divided into pEF (n = 99) and rEF (n = 44) groups. Arterial stiffness was assessed using arterial velocity pulse index (AVI) and arterial pressure volume index (API) at rest. The patients in the pEF group were significantly older and had a higher prevalence of coronary artery disease than the rEF group. The pEF group had significantly lower AVI levels and higher API levels than the rEF group. In the pEF group, the peak oxygen uptake (peak VO2) and the anaerobic threshold was significantly higher than those in the rEF group. The peak VO2 was significantly and negatively correlated with AVI and API in the pEF group (All, P < 0.05), but not in the rEF group. Multivariate linear regression analyses demonstrated that AVI was independently associated with peak VO2 (β = -0.34, P < 0.05) in the pEF group. In conclusion, AVI may be a useful factor for assessing exercise tolerance, particularly in CVD patients with pEF.
Collapse
Affiliation(s)
- Kei Fujiwara
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Kazunori Shimada
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine.,Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital.,Spotology Center, Juntendo University Graduate School of Medicine
| | - Miho Nishitani-Yokoyama
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine.,Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital
| | - Mitsuhiro Kunimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Tomomi Matsubara
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Rie Matsumori
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Abidan Abulimiti
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine.,Spotology Center, Juntendo University Graduate School of Medicine
| | - Tatsuro Aikawa
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Shohei Ouchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Megumi Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Kosuke Fukao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Tetsuro Miyazaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Akio Honzawa
- Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital
| | - Miki Yamada
- Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital
| | | | | | | | - Hiroyuki Daida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine.,Spotology Center, Juntendo University Graduate School of Medicine.,Juntendo University, Faculty of Health Science
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine.,Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development
| |
Collapse
|
127
|
Khan SS, Huffman MD, Shah SJ. Could a Low-Dose Diuretic Polypill Improve Outcomes in Heart Failure With Preserved Ejection Fraction? Circ Heart Fail 2021; 14:e008090. [PMID: 33663231 DOI: 10.1161/circheartfailure.120.008090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Sadiya S Khan
- Division of Cardiology, Department of Medicine (S.S.K.,M.D.H., S.J.S.), Northwestern University Feinberg School of Medicine, Chicago, IL.,Department of Preventive Medicine (S.S.K., M.D.H.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mark D Huffman
- Division of Cardiology, Department of Medicine (S.S.K.,M.D.H., S.J.S.), Northwestern University Feinberg School of Medicine, Chicago, IL.,Department of Preventive Medicine (S.S.K., M.D.H.), Northwestern University Feinberg School of Medicine, Chicago, IL.,The George Institute for Global Health, University of New South Wales, Sydney, Australia (M.D.H.)
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine (S.S.K.,M.D.H., S.J.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
128
|
Luxán G, Dimmeler S. The vasculature: a therapeutic target in heart failure? Cardiovasc Res 2021; 118:53-64. [PMID: 33620071 PMCID: PMC8752358 DOI: 10.1093/cvr/cvab047] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
It is well established that the vasculature plays a crucial role in maintaining oxygen and nutrients supply to the heart. Increasing evidence further suggest that the microcirculation has additional roles in supporting a healthy microenvironment. Heart failure is well known to be associated with changes and functional impairment of the microvasculature. The specific ablation of protective signals in endothelial cells in experimental models is sufficient to induce heart failure. Therefore, restoring a healthy endothelium and microcirculation may be a valuable therapeutic strategy to treat heart failure. The present review article will summarize the current understanding of the vascular contribution to heart failure with reduced or preserved ejection fraction. Novel therapeutic approaches including next generation pro-angiogenic therapies and non-coding RNA therapeutics, as well as the targeting of metabolites or metabolic signaling, vascular inflammation and senescence will be discussed.
Collapse
Affiliation(s)
- Guillermo Luxán
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany, German Center for Cardiovascular Research DZHK, Berlin, Germany, partner site Frankfurt Rhine-Main, Germany, Cardiopulmonary Institute, Goethe University Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany, German Center for Cardiovascular Research DZHK, Berlin, Germany, partner site Frankfurt Rhine-Main, Germany, Cardiopulmonary Institute, Goethe University Frankfurt, Germany
| |
Collapse
|
129
|
Berezin AE, Berezin AA. Shift of conventional paradigm of heart failure treatment: from angiotensin receptor neprilysin inhibitor to sodium-glucose co-transporter 2 inhibitors? Future Cardiol 2021; 17:497-506. [PMID: 33615880 DOI: 10.2217/fca-2020-0178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current clinical guidelines for heart failure (HF) contain a brand new therapeutic strategy for HF with reduced ejection fraction (HFrEF), which is based on neurohumoral modulation through the use of angiotensin receptor neprilysin inhibitors. There is a large body of evidence for the fact that sodium-glucose co-transporter 2 inhibitors may significantly improve all-cause mortality, cardiovascular mortality and hospitalization for HF in patients with HFrEF who received renin-angiotensin system blockers including angiotensin receptor neprilysin inhibitors, β-blockers and mineralocorticoid receptor antagonists. The review discusses that sodium-glucose co-transporter 2 inhibitors have a wide spectrum of favorable molecular effects and contribute to tissue protection, improving survival in HFrEF patients.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, State Medical University of Zaporozhye, 26, Mayakovsky av., Zaporozhye, UA-69035, Ukraine
| | - Alexander A Berezin
- Internal Medicine Department, Medical Academy of Post-Graduate Education, Ministry of Health of Ukraine, Zaporozhye, 69096, Ukraine
| |
Collapse
|
130
|
Alem MM. Clinical, Echocardiographic, and Therapeutic Characteristics of Heart Failure in Patients with Preserved, Mid-Range, and Reduced Ejection Fraction: Future Directions. Int J Gen Med 2021; 14:459-467. [PMID: 33623418 PMCID: PMC7896794 DOI: 10.2147/ijgm.s288733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023] Open
Abstract
Background Heart failure (HF) is recognized as a worldwide epidemic. Definitions and risk stratification are usually based upon measurements of left ventricular ejection fraction (LVEF) but such classifications reflect an underlying spectrum of different pathologic, phenotypic, and therapeutic patterns. Methods This was a retrospective cohort study of HF patients in Saudi Arabia. Patients were divided into three categories based on LVEF: those with preserved ejection fraction (EF) (EF≥50%, HFpEF); those with mid-range EF (EF 40–49%, HFmrEF); and those with reduced EF (EF <40%, HFrEF). Their demographics, co-morbid conditions, echocardiographic findings, pharmacological treatments and all-cause mortality (ACS) after a follow-up period of 24 months were compared. Results A total of 293 HF patients were identified (mean age: 63 years). In total, 65% were males, 79% were Saudi nationals, and 70% had type 2 diabetes mellitus (DM). Classification based on EF was established in 288 patients: HFpEF (105 patients, 36.5%), HFmrEF (49, 17.0%), and HFrEF (134, 46.5%). The 3 groups differed in sex distribution: 51% females in the HFpEF group and 78% males in the HFrEF group (P<0.001). Body mass index (BMI) was highest in the HFpEF group and lowest in the HFrEF group (31.5 vs 26.6; P<0.001). Although systolic blood pressure (SBP in mmHg) was highest in patients with HFpEF, left ventricular mass index (LVMI in g/cm2) was highest in patients with HFrEF 121.00 (94.50, 151.50), and eccentric hypertrophy was the dominant LV geometrical characteristic (54.6%). HFrEF patients had the highest use of ACE inhibitors (60.5%), loop diuretics (79.9%), and aldosterone receptor antagonists (56.7%) (P values; 0.009, 0.007, and <0.001, respectively). A total of 42 deaths occurred during follow-up: HFpEF (17 events), HFmrEF (3 events) and HFrEF (22 events) (Logrank test P=0.189). Conclusion This Saudi HF population shows similarities to other populations: EF category distribution, sex distribution, therapeutic trends, and survival outcomes. However, findings related to the underlying risk factors, namely type 2 DM and obesity, have identified HFpEF as an emerging threat in this (relatively) young population.
Collapse
Affiliation(s)
- Manal M Alem
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
131
|
Panhwar MS, Chatterjee S, Kalra A. Training the Critical Care Cardiologists of the Future: An Interventional Cardiology Critical Care Pathway. J Am Coll Cardiol 2021; 75:2984-2988. [PMID: 32527405 DOI: 10.1016/j.jacc.2020.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Muhammad Siyab Panhwar
- Tulane Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, Louisiana
| | - Saurav Chatterjee
- Division of Cardiovascular Medicine, North Shore-Long Island Jewish Medical Centers, Northwell Health, Zucker School of Medicine New York, New York
| | - Ankur Kalra
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio; Section of Cardiovascular Research, Heart, Vascular and Thoracic Department, Cleveland Clinic Akron General, Akron, Ohio.
| |
Collapse
|
132
|
Garg P, Assadi H, Jones R, Chan WB, Metherall P, Thomas R, van der Geest R, Swift AJ, Al-Mohammad A. Left ventricular fibrosis and hypertrophy are associated with mortality in heart failure with preserved ejection fraction. Sci Rep 2021; 11:617. [PMID: 33436786 PMCID: PMC7804435 DOI: 10.1038/s41598-020-79729-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/07/2020] [Indexed: 01/05/2023] Open
Abstract
Cardiac magnetic resonance (CMR) is emerging as an important tool in the assessment of heart failure with preserved ejection fraction (HFpEF). This study sought to investigate the prognostic value of multiparametric CMR, including left and right heart volumetric assessment, native T1-mapping and LGE in HFpEF. In this retrospective study, we identified patients with HFpEF who have undergone CMR. CMR protocol included: cines, native T1-mapping and late gadolinium enhancement (LGE). The mean follow-up period was 3.2 ± 2.4 years. We identified 86 patients with HFpEF who had CMR. Of the 86 patients (85% hypertensive; 61% males; 14% cardiac amyloidosis), 27 (31%) patients died during the follow up period. From all the CMR metrics, LV mass (area under curve [AUC] 0.66, SE 0.07, 95% CI 0.54-0.76, p = 0.02), LGE fibrosis (AUC 0.59, SE 0.15, 95% CI 0.41-0.75, p = 0.03) and native T1-values (AUC 0.76, SE 0.09, 95% CI 0.58-0.88, p < 0.01) were the strongest predictors of all-cause mortality. The optimum thresholds for these were: LV mass > 133.24 g (hazard ratio [HR] 1.58, 95% CI 1.1-2.2, p < 0.01); LGE-fibrosis > 34.86% (HR 1.77, 95% CI 1.1-2.8, p = 0.01) and native T1 > 1056.42 ms (HR 2.36, 95% CI 0.9-6.4, p = 0.07). In multivariate cox regression, CMR score model comprising these three variables independently predicted mortality in HFpEF when compared to NTproBNP (HR 4 vs HR 1.65). In non-amyloid HFpEF cases, only native T1 > 1056.42 ms demonstrated higher mortality (AUC 0.833, p < 0.01). In patients with HFpEF, multiparametric CMR aids prognostication. Our results show that left ventricular fibrosis and hypertrophy quantified by CMR are associated with all-cause mortality in patients with HFpEF.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, UK.
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK.
- Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Hosamadin Assadi
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, UK
| | - Rachel Jones
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, UK
| | - Wei Bin Chan
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Peter Metherall
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Richard Thomas
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | | | - Andrew J Swift
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, UK
| | - Abdallah Al-Mohammad
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, UK
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
133
|
Chang SN, Sung KT, Huang WH, Lin JW, Chien SC, Hung TC, Su CH, Hung CL, Tsai CT, Wu YW, Chiang FT, Yeh HI, Hwang JJ. Sex, racial differences and healthy aging in normative reference ranges on diastolic function in Ethnic Asians: 2016 ASE guideline revisited. J Formos Med Assoc 2021; 120:2160-2175. [PMID: 33423900 DOI: 10.1016/j.jfma.2020.12.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/08/2020] [Accepted: 12/24/2020] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Diastolic dysfunction (DD) has shown to be a hallmark pathological intermediate in the development of heart failure with preserved ejection fraction (HFpEF). We aim to establish age- and sex-stratified normal reference values of diastolic indices and to explore racial-differences. METHODS We explored age- and sex-related structural/functional alterations from 6023 healthy ethnic Asians (47.1 ± 10.9 years, 61.3% men) according to 2016 American Society of Echocardiography (ASE) diastolic dysfunction (DD) criteria. Racial comparisons were made using data from London Life Sciences Prospective Population (LOLIPOP) study. RESULTS Age- and sex-based normative ranges (including mean, median, 10% and 90% lower and upper reference values) were extracted from our large healthy population. In fully adjusted models, advanced age was independently associated with cardiac structural remodeling and worsened diastolic parameters including larger indexed LA volume (LAVi), lower e', higher E/e', and higher TR velocity; all p < 0.001), which were more prominent in women (P interaction: <0.05). Broadly, markedly lower e', higher E/e' and smaller LAVi were observed in ethnic Asians compared to Whites. DD defined by 2016 ASE criteria, despite at low prevalence (0.42%) in current healthy population, increased drastically with advanced age and performed perfectly in excluding abnormal NT-proBNP (≥125 pg/mL) (Specificity: 99.8%, NPV: 97.6%). CONCLUSION This is to date the largest cohort exploring the normative reference values using guideline-centered diastolic parameters from healthy Asians, with aging played as central role in diastolic dysfunction. Our observed sex and ethnic differences in defining healthy diastolic cut-offs likely impact future clinical definition for DD in Asians.
Collapse
Affiliation(s)
- Sheng-Nan Chang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Dou-Liu City, Taiwan
| | - Kuo-Tzu Sung
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Wen-Hung Huang
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Jou-Wei Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Dou-Liu City, Taiwan
| | - Shih-Chieh Chien
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan; Department of Critical Care Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ta-Chuan Hung
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Cheng-Huang Su
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Lieh Hung
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan; Institute of Biomedical Sciences, Mackay Medical College, New Taipei, Taiwan.
| | - Chia-Ti Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Wen Wu
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, Taipei, Taiwan
| | - Fu-Tien Chiang
- Division of Cardiology, Department of Internal Medicine, Fu-Jen Catholic University Hospital and Fu-Jen Catholic University, Taipei, Taiwan
| | - Hung-I Yeh
- Department of Medicine, Mackay Medical College, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Juey-Jen Hwang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Dou-Liu City, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
134
|
Wu YL. Cardiac MRI Assessment of Mouse Myocardial Infarction and Regeneration. Methods Mol Biol 2021; 2158:81-106. [PMID: 32857368 DOI: 10.1007/978-1-0716-0668-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Small animal models are indispensable for cardiac regeneration research. Studies in mouse and rat models have provided important insights into the etiology and mechanisms of cardiovascular diseases and accelerated the development of therapeutic strategies. It is vitally important to be able to evaluate the therapeutic efficacy and have reliable surrogate markers for therapeutic development for cardiac regeneration research. Magnetic resonance imaging (MRI), a versatile and noninvasive imaging modality with excellent penetration depth, tissue coverage, and soft-tissue contrast, is becoming a more important tool in both clinical settings and research arenas. Cardiac MRI (CMR) is versatile, noninvasive, and capable of measuring many different aspects of cardiac functions, and, thus, is ideally suited to evaluate therapeutic efficacy for cardiac regeneration. CMR applications include assessment of cardiac anatomy, regional wall motion, myocardial perfusion, myocardial viability, cardiac function assessment, assessment of myocardial infarction, and myocardial injury. Myocardial infarction models in mice are commonly used model systems for cardiac regeneration research. In this chapter, we discuss various CMR applications to evaluate cardiac functions and inflammation after myocardial infarction.
Collapse
Affiliation(s)
- Yijen L Wu
- Department of Developmental Biology, Rangos Research Center Animal Imaging Core, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
135
|
Badrov MB, Mak S, Floras JS. Cardiovascular Autonomic Disturbances in Heart Failure With Preserved Ejection Fraction. Can J Cardiol 2020; 37:609-620. [PMID: 33310140 DOI: 10.1016/j.cjca.2020.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/09/2023] Open
Abstract
In heart failure with reduced ejection fraction (HFrEF), diminished tonic and reflex vagal heart rate modulation and exaggerated sympathetic outflow and neural norepinephrine release are evident from disease inception. Each of these disturbances of autonomic regulation has been independently associated with shortened survival, and β-adrenoceptor antagonism and therapeutic autonomic modulation by other means have been demonstrated, in clinical trials, to lessen symptoms and prolong survival. In contrast, data concerning the autonomic status of patients with heart failure with preserved ejection fraction (HFpEF) are comparatively sparse. Little is known concerning the prognostic consequences of autonomic dysregulation in such individuals, and therapies applied with success in HFrEF have in most trials failed to improve symptoms or survival of those with HFpEF. A recent HFpEF Expert Scientific Panel report emphasised that without a deeper understanding of the pathophysiology of HFpEF, establishing effective treatment will be challenging. One aspect of such pathology may be cardiovascular autonomic disequilibrium, often worsened by acute exercise or routine daily activity. This review aims to summarise existing knowledge concerning parasympathetic and sympathetic function of patients with HFpEF, consider potential mechanisms and specific consequences of autonomic disturbances that have been identified, and propose hypotheses for future investigation.
Collapse
Affiliation(s)
- Mark B Badrov
- Division of Cardiology, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Susanna Mak
- Division of Cardiology, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - John S Floras
- Division of Cardiology, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
136
|
Multiscale classification of heart failure phenotypes by unsupervised clustering of unstructured electronic medical record data. Sci Rep 2020; 10:21340. [PMID: 33288774 PMCID: PMC7721729 DOI: 10.1038/s41598-020-77286-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
As a leading cause of death and morbidity, heart failure (HF) is responsible for a large portion of healthcare and disability costs worldwide. Current approaches to define specific HF subpopulations may fail to account for the diversity of etiologies, comorbidities, and factors driving disease progression, and therefore have limited value for clinical decision making and development of novel therapies. Here we present a novel and data-driven approach to understand and characterize the real-world manifestation of HF by clustering disease and symptom-related clinical concepts (complaints) captured from unstructured electronic health record clinical notes. We used natural language processing to construct vectorized representations of patient complaints followed by clustering to group HF patients by similarity of complaint vectors. We then identified complaints that were significantly enriched within each cluster using statistical testing. Breaking the HF population into groups of similar patients revealed a clinically interpretable hierarchy of subgroups characterized by similar HF manifestation. Importantly, our methodology revealed well-known etiologies, risk factors, and comorbid conditions of HF (including ischemic heart disease, aortic valve disease, atrial fibrillation, congenital heart disease, various cardiomyopathies, obesity, hypertension, diabetes, and chronic kidney disease) and yielded additional insights into the details of each HF subgroup's clinical manifestation of HF. Our approach is entirely hypothesis free and can therefore be readily applied for discovery of novel insights in alternative diseases or patient populations.
Collapse
|
137
|
Toth PP, Gauthier D. Heart failure with preserved ejection fraction: disease burden for patients, caregivers, and the health-care system. Postgrad Med 2020; 133:140-145. [PMID: 33131371 DOI: 10.1080/00325481.2020.1842621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) will soon become the most prevalent form of HF because of an aging population and an accompanying increase in the number of risk factors for this disease. The high frequency of comorbidities typical of this population contributes to an increased risk for hospitalization and death. It is also partially responsible for the symptomatic deterioration that results in hospitalization and impaired quality of life and functional capacity in patients. The effects of HFpEF are felt by patients and their caregivers, who might experience detriment to their own health and their social and working lives. Financial burden is associated with HFpEF, stemming from hospitalization and long-term care costs, as well as absenteeism from work in the case of caregivers. Early identification of patients at risk and aggressive management are key to preventing this disease and its progression.
Collapse
Affiliation(s)
- Peter P Toth
- Preventive Cardiology, CGH Medical Center, Rock Falls, IL, USA.,Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Diane Gauthier
- Section of Cardiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
138
|
Jamaly S, Carlsson L, Peltonen M, Andersson-Assarsson JC, Karason K. Heart failure development in obesity: underlying risk factors and mechanistic pathways. ESC Heart Fail 2020; 8:356-367. [PMID: 33231382 PMCID: PMC7835624 DOI: 10.1002/ehf2.13081] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 01/04/2023] Open
Abstract
Aims People with obesity are at risk for developing heart failure (HF), but little is known about the mechanistic pathways that link obesity with cardiac dysfunction. Methods and results We included 2030 participants from the Swedish Obese Subjects study who received conventional obesity treatment. First‐time detection of HF was obtained by cross‐checking the study population with the Swedish National Patient Register and the Swedish Cause of Death Register. We also examined if atrial fibrillation and myocardial infarction as time‐dependent variables could predict incident HF The mean age of the study cohort was 48.7 years, and 28% were men. The mean body mass index at baseline was 40.1 kg/m2 and remained stable during a median follow‐up of 20.1 years. First‐time diagnosis of HF occurred in 266 of patients and was related to male sex, increasing age, greater waist–hip ratio, hypertension, higher cholesterol, diabetes mellitus, and elevated free thyroxine in univariable analysis. Estimated glomerular filtration rate was negatively related to HF risk. In multivariable analysis, atrial fibrillation, which is related to HF with preserved ejection fraction (HFpEF), and myocardial infarction, which is linked to HF with reduced ejection fraction (HFrEF), were strongly associated with incident HF with sub‐hazard ratios 3.75 (95% confidence interval: 2.72–5.18, P < 0.001) and 3.68 (95% confidence interval: 2.55–5.30, P < 0.001), respectively. Conclusions Both atrial fibrillation and myocardial infarction as time‐dependent variables were independently and strongly related to incident HF in people with excess body fat, suggesting two main obesity‐related mechanistic pathways leading to either HFpEF or HFrEF.
Collapse
Affiliation(s)
- Shabbar Jamaly
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, 41345, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lena Carlsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Markku Peltonen
- National Institute for Health and Welfare, Helsinki, Finland
| | - Johanna C Andersson-Assarsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Kristjan Karason
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, 41345, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Tranplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
139
|
Spiesshoefer J, Henke C, Kabitz HJ, Bengel P, Schütt K, Nofer JR, Spieker M, Orwat S, Diller GP, Strecker JK, Giannoni A, Dreher M, Randerath WJ, Boentert M, Tuleta I. Heart Failure Results in Inspiratory Muscle Dysfunction Irrespective of Left Ventricular Ejection Fraction. Respiration 2020; 100:96-108. [PMID: 33171473 DOI: 10.1159/000509940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/04/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Exercise intolerance in heart failure with reduced ejection fraction (HFrEF) or heart failure with preserved ejection fraction (HFpEF) results from both cardiac dysfunction and skeletal muscle weakness. Respiratory muscle dysfunction with restrictive ventilation disorder may be present irrespective of left ventricular ejection fraction and might be mediated by circulating pro-inflammatory cytokines. OBJECTIVE To determine lung and respiratory muscle function in patients with HFrEF/HFpEF and to determine its associations with exercise intolerance and markers of systemic inflammation. METHODS Adult patients with HFrEF (n = 22, 19 male, 61 ± 14 years) and HFpEF (n = 8, 7 male, 68 ± 8 years) and 19 matched healthy control subjects underwent spirometry, measurement of maximum mouth occlusion pressures, diaphragm ultrasound, and recording of transdiaphragmatic and gastric pressures following magnetic stimulation of the phrenic nerves and the lower thoracic nerve roots. New York Heart Association (NYHA) class and 6-min walking distance (6MWD) were used to quantify exercise intolerance. Levels of circulating interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α) were measured using ELISAs. RESULTS Compared with controls, both patient groups showed lower forced vital capacity (FVC) (p < 0.05), maximum inspiratory pressure (PImax), maximum expiratory pressure (PEmax) (p < 0.05), diaphragm thickening ratio (p = 0.01), and diaphragm strength (twitch transdiaphragmatic pressure in response to supramaximal cervical magnetic phrenic nerve stimulation) (p = 0.01). In patients with HFrEF, NYHA class and 6MWD were both inversely correlated with FVC, PImax, and PEmax. In those with HFpEF, there was an inverse correlation between amino terminal pro B-type natriuretic peptide levels and FVC (r = -0.77, p = 0.04). In all HF patients, IL-6 and TNF-α were statistically related to FVC. CONCLUSIONS Irrespective of left ventricular ejection fraction, HF is associated with respiratory muscle dysfunction, which is associated with increased levels of circulating IL-6 and TNF-α.
Collapse
Affiliation(s)
- Jens Spiesshoefer
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy, .,Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany, .,Department of Neurology with Institute for Translational Neurology, University of Muenster, Muenster, Germany,
| | - Carolin Henke
- Department of Neurology, Herz-Jesu-Krankenhaus Hiltrup, Muenster, Germany
| | - Hans Joachim Kabitz
- Department of Pneumology, Cardiology and Intensive Care Medicine, Klinikum Konstanz, Konstanz, Germany
| | - Philipp Bengel
- Clinic for Cardiology and Pneumology/Heart Center, University Medical Center Goettingen, DZHK (German Centre for Cardiovascular Research), Goettingen, Germany
| | - Katharina Schütt
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Jerzy-Roch Nofer
- Center for Laboratory Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany and Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Spieker
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Orwat
- Department of Cardiology III, University Hospital Muenster, Muenster, Germany
| | - Gerhard Paul Diller
- Department of Cardiology III, University Hospital Muenster, Muenster, Germany
| | - Jan Kolia Strecker
- Department of Neurology with Institute for Translational Neurology, University of Muenster, Muenster, Germany
| | - Alberto Giannoni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michael Dreher
- Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Winfried Johannes Randerath
- Institute for Pneumology at the University of Cologne, Solingen, Germany.,Bethanien Hospital gGmbH Solingen, Solingen, Germany
| | - Matthias Boentert
- Department of Neurology with Institute for Translational Neurology, University of Muenster, Muenster, Germany.,Department of Medicine, UKM Marienhospital Steinfurt, Steinfurt, Germany
| | - Izabela Tuleta
- Department of Cardiology I, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
140
|
Ho JE, Redfield MM, Lewis GD, Paulus WJ, Lam CSP. Deliberating the Diagnostic Dilemma of Heart Failure With Preserved Ejection Fraction. Circulation 2020; 142:1770-1780. [PMID: 33136513 DOI: 10.1161/circulationaha.119.041818] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There is a lack of consensus on how we define heart failure with preserved ejection fraction (HFpEF), with wide variation in diagnostic criteria across society guidelines. This lack of uniformity in disease definition stems in part from an incomplete understanding of disease pathobiology, phenotypic heterogeneity, and natural history. We review current knowledge gaps and existing diagnostic tools and algorithms. We present a simple approach to implement these tools within the constraints of the current knowledge base, addressing separately (1) hospitalized individuals with rest congestion, where diagnosis is more straightforward; and (2) individuals with exercise intolerance, where diagnosis is more complex. Here, a potential role for advanced or provocative testing, including evaluation of hemodynamic responses to exercise is considered. More importantly, we propose focus areas for future studies to develop accurate and feasible diagnostic tools for HFpEF, including animal models that recapitulate human HFpEF, and human studies that both address a fundamental understanding of HFpEF pathobiology, and new diagnostic approaches and tools, as well. In sum, there is an urgent need to more accurately define the syndrome of HFpEF to inform diagnosis, patient selection for clinical trials, and, ultimately, future therapeutic approaches.
Collapse
Affiliation(s)
- Jennifer E Ho
- Corrigan Minehan Heart Center (J.E.H., G.D.L.), Massachusetts General Hospital, Harvard Medical School, Boston.,Cardiovascular Research Center (JE.H.), Massachusetts General Hospital, Harvard Medical School, Boston.,Division of Cardiology, Department of Medicine (J.E.H., G.D.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | | | - Gregory D Lewis
- Corrigan Minehan Heart Center (J.E.H., G.D.L.), Massachusetts General Hospital, Harvard Medical School, Boston.,Division of Cardiology, Department of Medicine (J.E.H., G.D.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Walter J Paulus
- Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, The Netherlands (W.J.P.)
| | - Carolyn S P Lam
- National Heart Centre Singapore (C.S.P.L.).,National Heart Center Singapore & Duke-NUS Medical School Singapore (C.S.P.L.).,Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands (C.S.P.L.).,The George Institute for Global Health, Sydney, Australia (C.S.P.L.)
| |
Collapse
|
141
|
Mehta A, Kondamudi N, Laukkanen JA, Wisloff U, Franklin BA, Arena R, Lavie CJ, Pandey A. Running away from cardiovascular disease at the right speed: The impact of aerobic physical activity and cardiorespiratory fitness on cardiovascular disease risk and associated subclinical phenotypes. Prog Cardiovasc Dis 2020; 63:762-774. [PMID: 33189764 DOI: 10.1016/j.pcad.2020.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023]
Abstract
Higher levels of physical activity (PA) and cardiorespiratory fitness (CRF) are associated with lower risk of incident cardiovascular disease (CVD). However, the relationship of aerobic PA and CRF with risk of atherosclerotic CVD outcomes and heart failure (HF) seem to be distinct. Furthermore, recent studies have raised concerns of potential toxicity associated with extreme levels of aerobic exercise, with higher levels of coronary artery calcium and incident atrial fibrillation noted among individuals with very high PA levels. In contrast, the relationship between PA levels and measures of left ventricular structure and function and risk of HF is more linear. Thus, personalizing exercise levels to optimal doses may be key to achieving beneficial outcomes and preventing adverse CVD events among high risk individuals. In this report, we provide a comprehensive review of the literature on the associations of aerobic PA and CRF levels with risk of adverse CVD outcomes and the preceding subclinical cardiac phenotypes to better characterize the optimal exercise dose needed to favorably modify CVD risk.
Collapse
Affiliation(s)
- Anurag Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nitin Kondamudi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jari A Laukkanen
- Faculty of Sport and Health Sciences, University of Jyvaskyla, Jyvaskyla, Finland
| | - Ulrik Wisloff
- K. G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Barry A Franklin
- Department of Cardiovascular Medicine, William Beaumont Hospital, Royal Oak, Michigan, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School - The University of Queensland School of Medicine, New Orleans, Louisiana, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
142
|
Brubaker PH, Avis T, Rejeski WJ, Mihalko SE, Tucker WJ, Kitzman DW. Exercise Training Effects on the Relationship of Physical Function and Health-Related Quality of Life Among Older Heart Failure Patients With Preserved Ejection Fraction. J Cardiopulm Rehabil Prev 2020; 40:427-433. [PMID: 32604218 PMCID: PMC7647941 DOI: 10.1097/hcr.0000000000000507] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE Although exercise training (ET) has been shown to improve both physical function and health-related quality of life (HRQOL) in older patients with heart failure and preserved ejection fraction (HFpEF), the relationship between changes in these important patient-centered outcome measures has not been adequately investigated. METHODS Patients (n = 116) with HFpEF (from 2 previous randomized controlled trials) were assigned to either 16 wk of endurance ET or attention control (CON). The ET in both trials consisted of ≤ 60 min of moderate-intensity endurance ET 3 time/wk. Peak exercise oxygen uptake (V˙o2peak) and other exercise capacity measures were obtained from a cardiopulmonary exercise test on an electronically braked cycle ergometer and 6-min walk test (6MWT). HRQOL was assessed using the Minnesota Living with Heart Failure (MLHF) Questionnaire and the 36-item Short Form Health Survey (SF-36). RESULTS Compared with CON, the ET group demonstrated significant improvement in measures of physical function (V˙o2peak and 6MWT) at 16 wk of follow-up. There were no significant differences observed between the groups for MLHF scores, but the ET group showed significant improvements on the SF-36. There were no significant correlations between change in any of the physical function and HRQOL measures in the ET group. CONCLUSIONS While endurance ET improved both physical function and some domains of HRQOL, the lack of significant correlations between changes in these measures suggests the effects of ET on physical function and HRQOL are largely independent of one another. Since these measures assess important and unique patient-centered outcomes in HFpEF patients, both physical function and HRQOL should be assessed in exercise-based programs and clinical trials.
Collapse
Affiliation(s)
- Peter H Brubaker
- Departments of Health and Exercise Science (Drs Brubaker, Rejeski, and Mihalko and Ms Avis) and Internal Medicine (Cardiology) (Dr Kitzman), Wake Forest University, Winston-Salem, North Carolina; and Department of Nutrition and Food Sciences, Texas Woman's University, Houston (Dr Tucker)
| | | | | | | | | | | |
Collapse
|
143
|
Sharma K, Mok Y, Kwak L, Agarwal SK, Chang PP, Deswal A, Shah AM, Kitzman DW, Wruck LM, Loehr LR, Heiss G, Coresh J, Rosamond WD, Solomon SD, Matsushita K, Russell SD. Predictors of Mortality by Sex and Race in Heart Failure With Preserved Ejection Fraction: ARIC Community Surveillance Study. J Am Heart Assoc 2020; 9:e014669. [PMID: 32924735 PMCID: PMC7792380 DOI: 10.1161/jaha.119.014669] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) accounts for half of heart failure hospitalizations, with limited data on predictors of mortality by sex and race. We evaluated for differences in predictors of all‐cause mortality by sex and race among hospitalized patients with HFpEF in the ARIC (Atherosclerosis Risk in Communities) Community Surveillance Study. Methods and Results Adjudicated HFpEF hospitalization events from 2005 to 2013 were analyzed from the ARIC Community Surveillance Study, comprising 4 US communities. Comparisons between clinical characteristics and mortality at 1 year were made by sex and race. Of 4335 adjudicated acute decompensated heart failure cases, 1892 cases (weighted n=8987) were categorized as HFpEF. Men had an increased risk of 1‐year mortality compared with women in adjusted analysis (hazard ratio [HR], 1.27; 95% CI, 1.06–1.52 [P=0.01]). Black participants had lower mortality compared with White participants in unadjusted and adjusted analyses (HR, 0.79; 95% CI, 0.64–0.97 [P=0.02]). Age, heart rate, worsening renal function, and low hemoglobin were associated with increased mortality in all subgroups. Higher body mass index was associated with improved survival in men, with borderline interaction by sex. Higher blood pressure was associated with improved survival among all groups, with significant interaction by race. Conclusions In a diverse HFpEF population, men had worse survival compared with women, and Black participants had improved survival compared with White participants. Age, heart rate, and worsening renal function were associated with increased mortality across all subgroups; high blood pressure was associated with decreased mortality with interaction by race. These insights into sex‐ and race‐based differences in predictors of mortality may help strategize targeted management of HFpEF.
Collapse
Affiliation(s)
- Kavita Sharma
- Division of Cardiology The Johns Hopkins Hospital Baltimore MD
| | - Yejin Mok
- Department of Epidemiology Johns Hopkins Bloomberg School of Public Health Baltimore MD
| | - Lucia Kwak
- Department of Epidemiology Johns Hopkins Bloomberg School of Public Health Baltimore MD
| | | | - Patricia P Chang
- Department of Medicine University of North Carolina Chapel Hill NC
| | - Anita Deswal
- Section of Cardiology Michael E. DeBakey VA Medical Center Baylor College of Medicine Houston TX
| | - Amil M Shah
- Cardiovascular Division Brigham and Women's Hospital Boston MA
| | - Dalane W Kitzman
- Cardiology and Geriatrics Sections Department of Internal Medicine Wake Forest School of Medicine Winston-Salem NC
| | - Lisa M Wruck
- Duke Clinical Research InstituteCenter for Predictive Medicine Durham NC
| | - Laura R Loehr
- Department of Epidemiology University of North Carolina Chapel Hill NC
| | - Gerardo Heiss
- Department of Epidemiology University of North Carolina Chapel Hill NC
| | - Josef Coresh
- Department of Epidemiology Johns Hopkins Bloomberg School of Public Health Baltimore MD
| | - Wayne D Rosamond
- Department of Epidemiology University of North Carolina Chapel Hill NC
| | - Scott D Solomon
- Cardiovascular Division Brigham and Women's Hospital Boston MA
| | - Kunihiro Matsushita
- Department of Epidemiology Johns Hopkins Bloomberg School of Public Health Baltimore MD
| | | |
Collapse
|
144
|
Udelson JE, Lewis GD, Shah SJ, Zile MR, Redfield MM, Burnett J, Parker J, Seferovic JP, Wilson P, Mittleman RS, Profy AT, Konstam MA. Effect of Praliciguat on Peak Rate of Oxygen Consumption in Patients With Heart Failure With Preserved Ejection Fraction: The CAPACITY HFpEF Randomized Clinical Trial. JAMA 2020; 324:1522-1531. [PMID: 33079154 PMCID: PMC7576408 DOI: 10.1001/jama.2020.16641] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE Heart failure with preserved ejection fraction (HFpEF) is often characterized by nitric oxide deficiency. OBJECTIVE To evaluate the efficacy and adverse effects of praliciguat, an oral soluble guanylate cyclase stimulator, in patients with HFpEF. DESIGN, SETTING, AND PARTICIPANTS CAPACITY HFpEF was a randomized, double-blind, placebo-controlled, phase 2 trial. Fifty-nine sites enrolled 196 patients with heart failure and an ejection fraction of at least 40%, impaired peak rate of oxygen consumption (peak V̇o2), and at least 2 conditions associated with nitric oxide deficiency (diabetes, hypertension, obesity, or advanced age). The trial randomized patients to 1 of 3 praliciguat dose groups or a placebo group, but was refocused early to a comparison of the 40-mg praliciguat dose vs placebo. Participants were enrolled from November 15, 2017, to April 30, 2019, with final follow-up on August 19, 2019. INTERVENTIONS Patients were randomized to receive 12 weeks of treatment with 40 mg of praliciguat daily (n = 91) or placebo (n = 90). MAIN OUTCOMES AND MEASURES The primary efficacy end point was the change from baseline in peak V̇o2 in patients who completed at least 8 weeks of assigned dosing. Secondary end points included the change from baseline in 6-minute walk test distance and in ventilatory efficiency (ventilation/carbon dioxide production slope). The primary adverse event end point was the incidence of treatment-emergent adverse events (TEAEs). RESULTS Among 181 patients (mean [SD] age, 70 [9] years; 75 [41%] women), 155 (86%) completed the trial. In the placebo (n = 78) and praliciguat (n = 65) groups, changes in peak V̇o2 were 0.04 mL/kg/min (95% CI, -0.49 to 0.56) and -0.26 mL/kg/min (95% CI, -0.83 to 0.31), respectively; the placebo-adjusted least-squares between-group difference in mean change from baseline was -0.30 mL/kg/min ([95% CI, -0.95 to 0.35]; P = .37). None of the 3 prespecified secondary end points were statistically significant. In the placebo and praliciguat groups, changes in 6-minute walk test distance were 58.1 m (95% CI, 26.1-90.1) and 41.4 m (95% CI, 8.2-74.5), respectively; the placebo-adjusted least-squares between-group difference in mean change from baseline was -16.7 m (95% CI, -47.4 to 13.9). In the placebo and praliciguat groups, the placebo-adjusted least-squares between-group difference in mean change in ventilation/carbon dioxide production slope was -0.3 (95% CI, -1.6 to 1.0). There were more dizziness (9.9% vs 1.1%), hypotension (8.8% vs 0%), and headache (11% vs 6.7%) TEAEs with praliciguat compared with placebo. The frequency of serious TEAEs was similar between the groups (10% in the praliciguat group and 11% in the placebo group). CONCLUSIONS AND RELEVANCE Among patients with HFpEF, the soluble guanylate cyclase stimulator praliciguat, compared with placebo, did not significantly improve peak V̇o2 from baseline to week 12. These findings do not support the use of praliciguat in patients with HFpEF. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03254485.
Collapse
Affiliation(s)
- James E. Udelson
- Division of Cardiology and the CardioVascular Center, Tufts Medical Center, Boston, Massachusetts
| | - Gregory D. Lewis
- Massachusetts General Hospital and Harvard Medical School, Boston
| | - Sanjiv J. Shah
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Michael R. Zile
- Medical University of South Carolina and the RHJ Department of Veterans Affairs Medical Center, Charleston
| | | | | | - John Parker
- Division of Cardiology, University Health Network, Mount Sinai Hospital, Toronto, Ontario
| | | | - Phebe Wilson
- Cyclerion Therapeutics, Cambridge, Massachusetts
| | | | | | - Marvin A. Konstam
- Division of Cardiology and the CardioVascular Center, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
145
|
Williams DM, Evans M. Dapagliflozin for Heart Failure with Preserved Ejection Fraction: Will the DELIVER Study Deliver? Diabetes Ther 2020; 11:2207-2219. [PMID: 32852697 PMCID: PMC7509021 DOI: 10.1007/s13300-020-00911-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 10/29/2022] Open
Abstract
Drug therapies for people with heart failure and preserved ejection fraction (HFpEF) are often limited to diuretics to improve symptoms as no therapies demonstrate a mortality benefit in this cohort. People with diabetes have a high risk of developing HFpEF and vice versa, suggesting shared pathophysiological mechanisms exist, which in turn engenders the potential for shared treatments. Dapagliflozin is a sodium-glucose co-transporter 2 (SGLT2) inhibitor which has demonstrated significantly improved cardiovascular and hospitalisation for heart failure (HHF) outcomes in previous cardiovascular outcome trials (CVOTs). These CVOTs include the DECLARE-TIMI and DAPA-HF studies which observed significant benefits for people with heart failure and specifically those with heart failure and reduced ejection fraction (HFrEF), respectively. The ongoing DELIVER study is evaluating the use of dapagliflozin specifically in people with HFpEF, which may have enormous implications for treatment and considerable economic consequences. This will complement previous and other ongoing CVOTs evaluating dapagliflozin use. In this review we discuss the use of SGLT2 inhibitors in HFrEF and HFpEF with a focus on the DELIVER study and its potential health and economic implications.
Collapse
Affiliation(s)
- David M Williams
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK.
| | - Marc Evans
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK
| |
Collapse
|
146
|
Huusko J, Tuominen S, Studer R, Corda S, Proudfoot C, Lassenius M, Ukkonen H. Recurrent hospitalizations are associated with increased mortality across the ejection fraction range in heart failure. ESC Heart Fail 2020; 7:2406-2417. [PMID: 32667143 PMCID: PMC7524224 DOI: 10.1002/ehf2.12792] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/22/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
AIMS The proportion of patients hospitalized for heart failure (HF) with preserved left ventricular ejection fraction (LVEF) is rising, but no approved treatment exists, in part owing to incomplete characterization of this particular HF phenotype. In order to better define the characteristics of HF phenotypes in Finland, a large cohort with 12 years' follow-up time was analysed. METHODS AND RESULTS Patients diagnosed between 2005 and 2017 at the Hospital District of Southwest Finland were stratified according to LVEF measure and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels. For this retrospective registry study, previously diagnosed HF patients were defined as follows: patients with reduced ejection fraction (HFrEF; LVEF ≤ 40%; n = 4042), mid-range ejection fraction (HFmrEF; LVEF > 40-50% and NT-proBNP ≥ 125 pg/mL; n = 1468), and preserved ejection fraction (HFpEF; LVEF > 50% and NT-proBNP ≥ 125 pg/mL; n = 3122) and followed up for 15 022, 4962, and 10 097 patient-years, respectively. Cardiovascular (CV) hospitalization and mortality, influence of pre-selected covariates on hospitalization and mortality, and the proportion of HFpEF and HFmrEF patients with a drop in LVEF to HFrEF phenotype were analysed. All data were extracted from the electronic patient register. HFrEF patients were rehospitalized slightly earlier than HFpEF/HFmrEF patients, but the second, third, and fourth rehospitalization rates did not differ between the subgroups. Female gender and better kidney function were associated with reduced rehospitalizations in HFmrEF and HFrEF, with a non-significant trend in HFpEF. Each additional hospitalization was associated with a two-fold increased risk of death and 2.2- to 2.3-fold increased risk of CV death. All-cause mortality was higher in patients with HFpEF. Although CV mortality was less frequent in HFpEF patients, it was associated with increased NT-proBNP concentrations at index in all patient groups. During the 10 years following the index date, 26% of HFmrEF patients and 10% of HFpEF patients progressed to an HFrEF phenotype. CONCLUSIONS These findings suggest that disease progression, in terms of increased frequency of hospitalizations, and the relationship between increased number of hospitalizations and mortality are similar by LVEF phenotypes. These data highlight the importance of effective treatments that can reduce hospitalizations and suggest a role for monitoring NT-proBNP levels in the management of HFpEF patients in particular.
Collapse
|
147
|
Liu LYM, Yun CH, Kuo JY, Lai YH, Sung KT, Yuan PJ, Tsai JP, Huang WH, Lin YH, Hung TC, Chen YJ, Su CH, Tsai CT, Yeh HI, Hung CL. Aortic Root Remodeling as an Indicator for Diastolic Dysfunction and Normative Ranges in Asians: Comparison and Validation with Multidetector Computed Tomography. Diagnostics (Basel) 2020; 10:diagnostics10090712. [PMID: 32961874 PMCID: PMC7555013 DOI: 10.3390/diagnostics10090712] [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: 08/28/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Background: The aortic root diameter (AoD) has been shown to be a marker of cardiovascular risk and heart failure (HF). Data regarding the normal reference ranges in Asians and their correlates with diastolic dysfunction using contemporary guidelines remain largely unexplored. Methods: Among 5343 consecutive population-based asymptomatic Asians with echocardiography evaluations for aortic root diameter (without/with indexing, presented as AoD/AoDi) were related to cardiac structure/function and N-terminal pro-brain B-type natriuretic peptide (Nt-ProBNP), with 245 participants compared with multidetector computed tomography (MDCT)-based aortic root geometry. Results: Advanced age, hypertension, higher diastolic blood pressure, and lower body fat all contributed to greater AoD/AoDi. The highest correlation between echo-based aortic diameter and the MDCT-derived measures was found at the level of the aortic sinuses of Valsalva (r = 0.80, p < 0.001). Age- and sex-stratified normative ranges of AoD/AoDi were provided in 3646 healthy participants. Multivariate linear regressions showed that AoDi was associated with a higher NT-proBNP, more unfavorable left ventricular (LV) remodeling, worsened LV systolic annular velocity (TDI-s′), a higher probability of presenting with LV hypertrophy, and abnormal LV diastolic indices except tricuspid regurgitation velocity by contemporary diastolic dysfunction (DD) criteria (all p < 0.05). AoDi superimposed on key clinical variables significantly expanded C-statistic from 0.71 to 0.84 (p for ∆AUROC: < 0.001). These associations were broadly weaker for AoD. Conclusion: In our large asymptomatic Asian population, echocardiography-defined aortic root dilation was associated with aging and hypertension and were correlated modestly with computed tomography measures. A larger indexed aortic diameter appeared to be a useful indicator in identifying baseline abnormal diastolic dysfunction.
Collapse
Affiliation(s)
- Lawrence Yu-min Liu
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- Division of Cardiology, Department of Internal Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
| | - Chun-Ho Yun
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Department of Radiology, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Jen-Yuan Kuo
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Yau-Huei Lai
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- Division of Cardiology, Department of Internal Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
| | - Kuo-Tzu Sung
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Po-Jung Yuan
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- Division of Cardiology, Department of Internal Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
| | - Jui-Peng Tsai
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Wen-Hung Huang
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Yueh-Hung Lin
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Ta-Chuan Hung
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Ying-Ju Chen
- Telehealth Center, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan;
| | - Cheng-Huang Su
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Cheng-Ting Tsai
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
- Correspondence: (C.-T.T.); (C.-L.H.)
| | - Hung-I Yeh
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Chung-Lieh Hung
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; (L.Y.-m.L.); (C.-H.Y.); (J.-Y.K.); (Y.-H.L.); (K.-T.S.); (P.-J.Y.); (J.-P.T.); (W.-H.H.); (Y.-H.L.); (T.-C.H.); (C.-H.S.); (H.-I.Y.)
- MacKay Junior College of Medicine, Nursing, and Management, Taipei City 11260, Taiwan
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan
- Telehealth Center, MacKay Memorial Hospital, Zhongshan North Road, Taipei City 10449, Taiwan;
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 25245, Taiwan
- Correspondence: (C.-T.T.); (C.-L.H.)
| |
Collapse
|
148
|
Oxidative Stress and Inflammatory Modulation of Ca 2+ Handling in Metabolic HFpEF-Related Left Atrial Cardiomyopathy. Antioxidants (Basel) 2020; 9:antiox9090860. [PMID: 32937823 PMCID: PMC7555173 DOI: 10.3390/antiox9090860] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
Metabolic syndrome-mediated heart failure with preserved ejection fraction (HFpEF) is commonly accompanied by left atrial (LA) cardiomyopathy, significantly affecting morbidity and mortality. We evaluate the role of reactive oxygen species (ROS) and intrinsic inflammation (TNF-α, IL-10) related to dysfunctional Ca2+ homeostasis of LA cardiomyocytes in a rat model of metabolic HFpEF. ZFS-1 obese rats showed features of HFpEF and atrial cardiomyopathy in vivo: increased left ventricular (LV) mass, E/e’ and LA size and preserved LV ejection fraction. In vitro, LA cardiomyocytes exhibited more mitochondrial-fission (MitoTracker) and ROS-production (H2DCF). In wildtype (WT), pro-inflammatory TNF-α impaired cellular Ca2+ homeostasis, while anti-inflammatory IL-10 had no notable effect (confocal microscopy; Fluo-4). In HFpEF, TNF-α had no effect on Ca2+ homeostasis associated with decreased TNF-α receptor expression (western blot). In addition, IL-10 substantially improved Ca2+ release and reuptake, while IL-10 receptor-1 expression was unaltered. Oxidative stress in metabolic syndrome mediated LA cardiomyopathy was increased and anti-inflammatory treatment positively affected dysfunctional Ca2+ homeostasis. Our data indicates, that patients with HFpEF-related LA dysfunction might profit from IL-10 targeted therapy, which should be further explored in preclinical trials.
Collapse
|
149
|
Billingsley HE, Hummel SL, Carbone S. The role of diet and nutrition in heart failure: A state-of-the-art narrative review. Prog Cardiovasc Dis 2020; 63:538-551. [PMID: 32798501 PMCID: PMC7686142 DOI: 10.1016/j.pcad.2020.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 08/09/2020] [Indexed: 02/07/2023]
Abstract
Heart Failure (HF) incidence is increasing steadily worldwide, while prognosis remains poor. Though nutrition is a lifestyle factor implicated in prevention of HF, little is known about the effects of macro- and micronutrients as well as dietary patterns on the progression and treatment of HF. This is reflected in a lack of nutrition recommendations in all major HF scientific guidelines. In this state-of-the-art review, we examine and discuss the implications of evidence contained in existing randomized control trials as well as observational studies covering the topics of sodium restriction, dietary patterns and caloric restriction as well as supplementation of dietary fats and fatty acids, protein and amino acids and micronutrients in the setting of pre-existing HF. Finally, we explore future directions and discuss knowledge gaps regarding nutrition therapies for the treatment of HF.
Collapse
Affiliation(s)
- Hayley E Billingsley
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, United States of America; VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Scott L Hummel
- University of Michigan Frankel Cardiovascular Center, Ann Arbor, MI, United States of America; Ann Arbor Veterans Affairs Health System, Ann Arbor, MI, United States of America
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, United States of America; VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States of America.
| |
Collapse
|
150
|
Lima FV, Kennedy KF, Sheikh W, French A, Parulkar A, Sharma E, Henien S, Wu M, Chu A. Thirty‐day readmissions after atrial fibrillation catheter ablation in patients with heart failure. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2020; 43:930-940. [DOI: 10.1111/pace.14013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/02/2020] [Accepted: 07/12/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Fabio V. Lima
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Kevin F. Kennedy
- Mid America Heart and Vascular Institute St. Luke's Hospital Kansas City Missouri
| | - Wasiq Sheikh
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Amy French
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Anshul Parulkar
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Esseim Sharma
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Shady Henien
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Michael Wu
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| | - Antony Chu
- Cardiovascular Institute Warren Alpert School of Medicine, Brown University 593 Eddy Street, APC 814 Providence RI 02903
| |
Collapse
|