1
|
Vrabie AM, Totolici S, Delcea C, Badila E. Biomarkers in Heart Failure with Preserved Ejection Fraction: A Perpetually Evolving Frontier. J Clin Med 2024; 13:4627. [PMID: 39200768 PMCID: PMC11355893 DOI: 10.3390/jcm13164627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a complex clinical syndrome, often very difficult to diagnose using the available tools. As the global burden of this disease is constantly growing, surpassing the prevalence of heart failure with reduced ejection fraction, during the last few years, efforts have focused on optimizing the diagnostic and prognostic pathways using an immense panel of circulating biomarkers. After the paradigm of HFpEF development emerged more than 10 years ago, suggesting the impact of multiple comorbidities on myocardial structure and function, several phenotypes of HFpEF have been characterized, with an attempt to find an ideal biomarker for each distinct pathophysiological pathway. Acknowledging the limitations of natriuretic peptides, hundreds of potential biomarkers have been evaluated, some of them demonstrating encouraging results. Among these, soluble suppression of tumorigenesis-2 reflecting myocardial remodeling, growth differentiation factor 15 as a marker of inflammation and albuminuria as a result of kidney dysfunction or, more recently, several circulating microRNAs have proved their incremental value. As the number of emerging biomarkers in HFpEF is rapidly expanding, in this review, we aim to explore the most promising available biomarkers linked to key pathophysiological mechanisms in HFpEF, outlining their utility for diagnosis, risk stratification and population screening, as well as their limitations.
Collapse
Affiliation(s)
- Ana-Maria Vrabie
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Stefan Totolici
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Caterina Delcea
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Elisabeta Badila
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
2
|
Gharagozloo K, Mehdizadeh M, Heckman G, Rose RA, Howlett J, Howlett SE, Nattel S. Heart Failure With Preserved Ejection Fraction in the Elderly Population: Basic Mechanisms and Clinical Considerations. Can J Cardiol 2024; 40:1424-1444. [PMID: 38604339 DOI: 10.1016/j.cjca.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/13/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) refers to a clinical condition in which the signs of heart failure, such as pulmonary congestion, peripheral edema, and increased natriuretic peptide levels, are present despite normal ejection fractions and the absence of other causes (eg, pericardial disease). The ejection fraction cutoff for the definition of HFpEF has varied in the past, but recent society guidelines have settled on a consensus of 50%. HFpEF is particularly common in the elderly population. The aim of this narrative review is to summarize the available literature regarding HFpEF in elderly patients in terms of evidence for the age dependence, specific clinical features, and underlying mechanisms. In the clinical arena, we review the epidemiology, discuss distinct clinical phenotypes typically seen in elderly patients, the importance of frailty, the role of biomarkers, and the role of medical therapies (including sodium-glucose cotransport protein 2 inhibitors, renin-angiotensin-aldosterone system blockers, angiotensin receptor/neprilysin inhibitors, diuretics, and β-adrenergic receptor blockers). We then go on to discuss the basic mechanisms implicated in HFpEF, including cellular senescence, fibrosis, inflammation, mitochondrial dysfunction, enhanced production of reactive oxygen species, abnormal cellular calcium handling, changes in microRNA signalling, insulin resistance, and sex hormone changes. Finally, we review knowledge gaps and promising areas of future investigation. Improved understanding of the specific clinical manifestations of HFpEF in elderly individuals and of the fundamental mechanisms that contribute to the age-related risk of HFpEF promises to lead to novel diagnostic and treatment approaches that will improve outcomes for this common cardiac disorder in a vulnerable population.
Collapse
Affiliation(s)
- Kimia Gharagozloo
- Montreal Heart Institute Research Center and Université de Montréal, Montréal, Quebec, Canada; McGill University Departments of Pharmacology and Therapeutics, Montréal, Quebec, Canada
| | - Mozhdeh Mehdizadeh
- Montreal Heart Institute Research Center and Université de Montréal, Montréal, Quebec, Canada; McGill University Departments of Pharmacology and Therapeutics, Montréal, Quebec, Canada
| | - George Heckman
- Schlegel Research Institute for Aging and University of Waterloo, Waterloo, Ontario, Canada
| | - Robert A Rose
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan Howlett
- Libin Cardiovascular Institute and Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Susan E Howlett
- Departments of Pharmacology and Medicine (Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Stanley Nattel
- Montreal Heart Institute Research Center and Université de Montréal, Montréal, Quebec, Canada; McGill University Departments of Pharmacology and Therapeutics, Montréal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
3
|
Andrzejczyk K, Abou Kamar S, van Ommen AM, Canto ED, Petersen TB, Valstar G, Akkerhuis KM, Cramer MJ, Umans V, Rutten FH, Teske A, Boersma E, Menken R, van Dalen BM, Hofstra L, Verhaar M, Brugts J, Asselbergs F, den Ruijter H, Kardys I. Identifying plasma proteomic signatures from health to heart failure, across the ejection fraction spectrum. Sci Rep 2024; 14:14871. [PMID: 38937570 PMCID: PMC11211454 DOI: 10.1038/s41598-024-65667-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
Circulating proteins may provide insights into the varying biological mechanisms involved in heart failure (HF) with preserved ejection fraction (HFpEF) and reduced ejection fraction (HFrEF). We aimed to identify specific proteomic patterns for HF, by comparing proteomic profiles across the ejection fraction spectrum. We investigated 4210 circulating proteins in 739 patients with normal (Stage A/Healthy) or elevated (Stage B) filling pressures, HFpEF, or ischemic HFrEF (iHFrEF). We found 2122 differentially expressed proteins between iHFrEF-Stage A/Healthy, 1462 between iHFrEF-HFpEF and 52 between HFpEF-Stage A/Healthy. Of these 52 proteins, 50 were also found in iHFrEF vs. Stage A/Healthy, leaving SLITRK6 and NELL2 expressed in lower levels only in HFpEF. Moreover, 108 proteins, linked to regulation of cell fate commitment, differed only between iHFrEF-HFpEF. Proteomics across the HF spectrum reveals overlap in differentially expressed proteins compared to stage A/Healthy. Multiple proteins are unique for distinguishing iHFrEF from HFpEF, supporting the capacity of proteomics to discern between these conditions.
Collapse
Affiliation(s)
- Karolina Andrzejczyk
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sabrina Abou Kamar
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Anne-Mar van Ommen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elisa Dal Canto
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Teun B Petersen
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gideon Valstar
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maarten Jan Cramer
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Victor Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands
| | - Frans H Rutten
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arco Teske
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Roxana Menken
- Cardiology Centers of the Netherlands, Utrecht, The Netherlands
| | - Bas M van Dalen
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Leonard Hofstra
- Cardiology Centers of the Netherlands, Utrecht, The Netherlands
| | - Marianne Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jasper Brugts
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Folkert Asselbergs
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hester den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Chiu L, Agrawal V, Armstrong D, Brittain E, Collins S, Hemnes AR, Hill JA, Lindenfeld J, Shah SJ, Stevenson LW, Wang TJ, Gupta DK. Correlates of Plasma NT-proBNP/Cyclic GMP Ratio in Heart Failure With Preserved Ejection Fraction: An Analysis of the RELAX Trial. J Am Heart Assoc 2024; 13:e031796. [PMID: 38533961 PMCID: PMC11179778 DOI: 10.1161/jaha.123.031796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Phosphodiesterases degrade cyclic GMP (cGMP), the second messenger that mediates the cardioprotective effects of natriuretic peptides. High natriuretic peptide/cGMP ratio may reflect, in part, phosphodiesterase activity. Correlates of natriuretic peptide/cGMP in patients with heart failure with preserved ejection fraction are not well understood. Among patients with heart failure with preserved ejection fraction in the RELAX (Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure With Preserved Ejection Fraction) trial, we examined (1) cross-sectional correlates of circulating NT-proBNP (N-terminal pro-B-type natriuretic peptide)/cGMP ratio, (2) whether selective phosphodiesterase-5 inhibition by sildenafil changed the ratio, and (3) whether the effect of sildenafil on 24-week outcomes varied by baseline ratio. METHODS AND RESULTS In 212 subjects, NT-proBNP/cGMP ratio was calculated at randomization and 24 weeks. Correlates of the ratio and its change were examined in multivariable proportional odds models. Whether baseline ratio modified the sildenafil effect on outcomes was examined by interaction terms. Higher NT-proBNP/cGMP ratio was associated with greater left ventricular mass and troponin, the presence of atrial fibrillation, and lower estimated glomerular filtration rate and peak oxygen consumption. Compared with placebo, sildenafil did not alter the ratio from baseline to 24 weeks (P=0.17). The effect of sildenafil on 24-week change in peak oxygen consumption, left ventricular mass, or clinical composite outcome was not modified by baseline NT-proBNP/cGMP ratio (P-interaction >0.30 for all). CONCLUSIONS Among patients with heart failure with preserved ejection fraction, higher NT-proBNP/cGMP ratio associated with an adverse cardiorenal phenotype, which was not improved by selective phosphodiesterase-5 inhibition. Other phosphodiesterases may be greater contributors than phosphodiesterase-5 to the adverse phenotype associated with a high natriuretic peptide/cGMP ratio in HFpEF. REGISTRATION INFORMATION clinicaltrials.gov. Identifier: NCT00763867.
Collapse
Affiliation(s)
- Leonard Chiu
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Vineet Agrawal
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - David Armstrong
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Evan Brittain
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Sheila Collins
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Anna R. Hemnes
- Division of Pulmonary MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology)University of Texas Southwestern Medical CenterDallasTXUSA
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - JoAnn Lindenfeld
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Lynne W. Stevenson
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Thomas J. Wang
- Department of Internal Medicine (Cardiology)University of Texas Southwestern Medical CenterDallasTXUSA
| | - Deepak K. Gupta
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|
5
|
Rao VS, Ivey-Miranda JB, Cox ZL, Moreno-Villagomez J, Testani JM. Association of Urine Galectin-3 With Cardiorenal Outcomes in Patients With Heart Failure. J Card Fail 2024; 30:340-346. [PMID: 37301248 PMCID: PMC10947725 DOI: 10.1016/j.cardfail.2023.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Approaches to distinguishing pathological cardiorenal dysfunction in heart failure (HF) from functional/hemodynamically mediated changes in serum creatinine are needed. We investigated urine galectin-3 as a candidate biomarker of renal fibrosis and a prognostic indicator of cardiorenal dysfunction phenotypes. METHODS We measured urine galectin-3 in 2 contemporary HF cohorts: the Yale Transitional Care Clinic (YTCC) cohort (n = 132) and the Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist (TOPCAT) trial (n = 434). We assessed the association of urine galectin-3 with all-cause mortality in both cohorts and the association with an established marker of renal tissue fibrosis, urinary amino-terminal propeptide of type III procollagen (PIIINP) in TOPCAT. RESULTS In the YTCC cohort, there was significant effect modification between higher urine galectin-3 and lower estimated glomerular filtration rates (eGFRs) (Pinteraction = 0.046), such that low eGFR levels had minimal prognostic importance if urine galectin-3 levels were low, but they were important and indicated high risk if urine galectin-3 levels were high. Similar observations were noted in the TOPCAT study (Pinteraction = 0.002). In TOPCAT, urine galectin-3 also positively correlated with urine PIIINP at both baseline (r = 0.43; P < 0.001) and at 12 months (r = 0.42; P < 0.001). CONCLUSIONS Urine galectin-3 levels correlated with an established biomarker of renal fibrosis in 2 cohorts and was able to differentiate high- vs low-risk phenotypes of chronic kidney disease in HF. These proof-of-concept results indicate that additional biomarker research to differentiate cardiorenal phenotypes is warranted.
Collapse
Affiliation(s)
- Veena S Rao
- Division of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Juan B Ivey-Miranda
- Division of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA; Heart Failure and Heart Transplant Clinic, Hospital de Cardiologia, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Zachary L Cox
- Department of Pharmacy Practice, Lipscomb University College of Pharmacy, Nashville, TN, USA; Department of Pharmacy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julieta Moreno-Villagomez
- Division of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jeffrey M Testani
- Division of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
6
|
Smart CD, Madhur MS. The immunology of heart failure with preserved ejection fraction. Clin Sci (Lond) 2023; 137:1225-1247. [PMID: 37606086 PMCID: PMC10959189 DOI: 10.1042/cs20230226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of new heart failure diagnoses and continues to increase in prevalence in the United States. Importantly, HFpEF is a highly morbid, heterogeneous syndrome lacking effective therapies. Inflammation has emerged as a potential contributor to the pathogenesis of HFpEF. Many of the risk factors for HFpEF are also associated with chronic inflammation, such as obesity, hypertension, aging, and renal dysfunction. A large amount of preclinical evidence suggests that immune cells and their associated cytokines play important roles in mediating fibrosis, oxidative stress, metabolic derangements, and endothelial dysfunction, all potentially important processes in HFpEF. How inflammation contributes to HFpEF pathogenesis, however, remains poorly understood. Recently, a variety of preclinical models have emerged which may yield much needed insights into the causal relationships between risk factors and the development of HFpEF, including the role of specific immune cell subsets or inflammatory pathways. Here, we review evidence in animal models and humans implicating inflammation as a mediator of HFpEF and identify gaps in knowledge requiring further study. As the understanding between inflammation and HFpEF evolves, it is hoped that a better understanding of the mechanisms underlying immune cell activation in HFpEF can open up new therapeutic avenues.
Collapse
Affiliation(s)
- Charles Duncan Smart
- Department of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nashville, TN, U.S.A
| | - Meena S. Madhur
- Department of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nashville, TN, U.S.A
- Department of Medicine, Division of Cardiovascular
Medicine, Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Department of Medicine, Division of Clinical Pharmacology,
Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Vanderbilt Institute for Infection, Immunology, and
Inflammation, Nashville, TN, U.S.A
| |
Collapse
|
7
|
Hsu CC, Wang JS, Shyu YC, Fu TC, Juan YH, Yuan SS, Wang CH, Yeh CH, Liao PC, Wu HY, Hsu PH. Hypermethylation of ACADVL is involved in the high-intensity interval training-associated reduction of cardiac fibrosis in heart failure patients. J Transl Med 2023; 21:187. [PMID: 36894992 PMCID: PMC9999524 DOI: 10.1186/s12967-023-04032-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Emerging evidence suggests that DNA methylation can be affected by physical activities and is associated with cardiac fibrosis. This translational research examined the implications of DNA methylation associated with the high-intensity interval training (HIIT) effects on cardiac fibrosis in patients with heart failure (HF). METHODS Twelve HF patients were included and received cardiovascular magnetic resonance imaging with late gadolinium enhancement for cardiac fibrosis severity and a cardiopulmonary exercise test for peak oxygen consumption ([Formula: see text]O2peak). Afterwards, they underwent 36 sessions of HIIT at alternating 80% and 40% of [Formula: see text]O2peak for 30 min per session in 3-4 months. Human serum from 11 participants, as a means to link cell biology to clinical presentations, was used to investigate the exercise effects on cardiac fibrosis. Primary human cardiac fibroblasts (HCFs) were incubated in patient serum, and analyses of cell behaviour, proteomics (n = 6) and DNA methylation profiling (n = 3) were performed. All measurements were conducted after completing HIIT. RESULTS A significant increase (p = 0.009) in [Formula: see text]O2peak (pre- vs. post-HIIT = 19.0 ± 1.1 O2 ml/kg/min vs. 21.8 ± 1.1 O2 ml/kg/min) was observed after HIIT. The exercise strategy resulted in a significant decrease in left ventricle (LV) volume by 15% to 40% (p < 0.05) and a significant increase in LV ejection fraction by approximately 30% (p = 0.010). LV myocardial fibrosis significantly decreased from 30.9 ± 1.2% to 27.2 ± 0.8% (p = 0.013) and from 33.4 ± 1.6% to 30.1 ± 1.6% (p = 0.021) in the middle and apical LV myocardium after HIIT, respectively. The mean single-cell migration speed was significantly (p = 0.044) greater for HCFs treated with patient serum before (2.15 ± 0.17 μm/min) than after (1.11 ± 0.12 μm/min) HIIT. Forty-three of 1222 identified proteins were significantly involved in HIIT-induced altered HCF activities. There was significant (p = 0.044) hypermethylation of the acyl-CoA dehydrogenase very long chain (ACADVL) gene with a 4.474-fold increase after HIIT, which could activate downstream caspase-mediated actin disassembly and the cell death pathway. CONCLUSIONS Human investigation has shown that HIIT is associated with reduced cardiac fibrosis in HF patients. Hypermethylation of ACADVL after HIIT may contribute to impeding HCF activities. This exercise-associated epigenetic reprogramming may contribute to reduce cardiac fibrosis and promote cardiorespiratory fitness in HF patients. TRIAL REGISTRATION NCT04038723. Registered 31 July 2019, https://clinicaltrials.gov/ct2/show/NCT04038723 .
Collapse
Affiliation(s)
- Chih-Chin Hsu
- Department of Physical Medicine and Rehabilitation, Keelung Chang Gung Memorial Hospital, No. 200, Lane 208, Jijin 1St Rd., Anle Dist, Keelung, 204, Taiwan.
- Community Medicine Research Center, Keelung Chang Gung Memorial Hospital, Keelung, 204, Taiwan.
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
| | - Jong-Shyan Wang
- Department of Physical Medicine and Rehabilitation, Keelung Chang Gung Memorial Hospital, No. 200, Lane 208, Jijin 1St Rd., Anle Dist, Keelung, 204, Taiwan
- Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yu-Chiau Shyu
- Community Medicine Research Center, Keelung Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Tieh-Cheng Fu
- Department of Physical Medicine and Rehabilitation, Keelung Chang Gung Memorial Hospital, No. 200, Lane 208, Jijin 1St Rd., Anle Dist, Keelung, 204, Taiwan
| | - Yu-Hsiang Juan
- Department of Medical Imaging and intervention, Linkou and Taoyuan Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Shin-Sheng Yuan
- Institute of Statistical Science, Academia Sinica, Taipei, 115, Taiwan
| | - Chao-Hung Wang
- Department of Cardiology, Keelung Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Chi-Hsiao Yeh
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Division of Thoracic and Cardiovascular Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Po-Cheng Liao
- Community Medicine Research Center, Keelung Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Hsin-Yi Wu
- Instrumentation Center, National Taiwan University, Taipei, 106, Taiwan
| | - Pang-Hung Hsu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, No. 2, Beining Rd., Zhongzheng Dist., Keelung, 202, Taiwan.
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 202, Taiwan.
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
8
|
Rucker D, Joseph J. Defining the Phenotypes for Heart Failure With Preserved Ejection Fraction. Curr Heart Fail Rep 2022; 19:445-457. [PMID: 36178663 DOI: 10.1007/s11897-022-00582-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE OF REVIEW Heart failure with preserved ejection fraction (HFpEF) imposes a significant burden on society and healthcare. The lack in efficacious therapies is likely due to the significant heterogeneity of HFpEF. In this review, we define various phenotypes based on underlying comorbidities or etiologies, discuss phenotypes arrived at by novel methods, and explore therapeutic targets. RECENT FINDINGS A few studies have used machine learning methods to uncover sub-phenotypes within HFpEF in an unbiased manner based on clinical features, echocardiographic findings, and biomarker levels. We synthesized the literature and propose three broad phenotypes: (1) young, with few comorbidities, usually obese and with low natriuretic peptide levels, (2) obese with substantive cardiometabolic burden and comorbidities and impaired ventricular relaxation, (3) old, multimorbid, with high rates of atrial fibrillation, renal and coronary artery disease, chronic obstructive pulmonary disease, and left ventricular hypertrophy. We also propose potential therapeutic strategies for these phenotypes.
Collapse
Affiliation(s)
- Dane Rucker
- Department of Medicine, Boston Medical Center, Boston, MA, USA
| | - Jacob Joseph
- Massachusetts Veterans Epidemiology Research & Information Center, Veterans Affairs Boston Healthcare System, Cardiology Section (111), 1400 VFW Parkway, West Roxbury, Boston, MA, 02132, USA. .,Department of Medicine, Brigham & Women's Hospital, Boston, MA, USA.
| |
Collapse
|
9
|
Chumakova S, Urazova O, Shipulin V, Vins M, Pryakhin A, Sukhodolo I, Stelmashenko A, Litvinova L, Kolobovnikova Y, Churina E, Novitskiy V. Galectin 3 and non-classical monocytes of blood as myocardial remodeling factors at ischemic cardiomyopathy. IJC HEART & VASCULATURE 2021; 33:100766. [PMID: 33869726 PMCID: PMC8047163 DOI: 10.1016/j.ijcha.2021.100766] [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: 12/31/2020] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/19/2022]
Abstract
Aims To identify an imbalance of cardiac remodeling mediators and monocytes subpopulation in blood, distribution of myocardium macrophages in patients with ischemic cardiomyopathy (ICMP). Methods The study engaged 30 patients with ICMP, 26 patients with coronary heart disease (CHD) without ICMP, 15 healthy donors. Concentrations of TGFβ, MMP-9, MCP-1, galectin-3 were measured in plasma of blood from the coronary sinus and peripheral blood in CHD patients, as well as in peripheral blood in healthy donors, by enzyme immunoassay method. The ration of classical, intermediate, non-classical, transitional monocytes in peripheral blood of patients and healthy donors was assessed by flow cytometry (expression CD14, CD16); the content of CD68+ macrophages in myocardium – by immunohistochemistry method. Results In both samples of blood, the content of galectin-3 in patients with ICMP was higher than in CHD patients without ICMP and the level of TGFβ was comparable between the groups. At ICMP, the concentration of MMP-9 in sinus blood was higher than that in CHD patients without ICMP in whom an excess of MCP-1 in the general blood flow was determined. The density of distribution of CD68+ cells in the myocardium in patients with ICMP was higher in the perianeurysmal zone than in the right atrium appendage. ICMP was characterized by a deficiency of non-classical monocytes, and CHD without ICMP – by an excess of intermediate cells in peripheral blood. Conclusion Myocardium remodeling at ICMP is mediated by not so much TGFβ but intracardiac galectin-3, which determines the subpopulation composition of blood monocytes.
Collapse
Affiliation(s)
- S Chumakova
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - O Urazova
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - V Shipulin
- Cardiovascular Surgery Unit of CardiologyResearchInstitute, Tomsk National Medical Research Center of Russian Academy of Sciences, 111A Kievskaya Street, Tomsk 634012, Russia
| | - M Vins
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - A Pryakhin
- Cardiovascular Surgery Unit of CardiologyResearchInstitute, Tomsk National Medical Research Center of Russian Academy of Sciences, 111A Kievskaya Street, Tomsk 634012, Russia
| | - I Sukhodolo
- Morphology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - A Stelmashenko
- Morphology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - L Litvinova
- Immunology and Cell Biotechnology Center of Immanuel Kant, Baltic Federal University, 14 A. Nevskogo Street, Kaliningrad 236041, Russia
| | - Yu Kolobovnikova
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - E Churina
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia.,National Research Tomsk State University, 36 Lenina Ave, Tomsk 634050, Russia
| | - V Novitskiy
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| |
Collapse
|
10
|
Fudim M, Fathallah M, Shaw LK, James O, Samad Z, Piccini JP, Hess PL, Borges-Neto S. The prognostic value of diastolic and systolic mechanical left ventricular dyssynchrony among patients with coronary artery disease and heart failure. J Nucl Cardiol 2020; 27:1622-1632. [PMID: 31392509 DOI: 10.1007/s12350-019-01843-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/20/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Prevalence and prognostic value of diastolic and systolic dyssynchrony in patients with coronary artery disease (CAD) + heart failure (HF) or CAD alone are not well understood. METHODS We included patients with gated single-photon emission computed tomography (GSPECT) myocardial perfusion imaging (MPI) between 2003 and 2009. Patients had at least one major epicardial obstruction ≥ 50%. We assessed the association between dyssynchrony and outcomes, including all-cause and cardiovascular death. RESULTS Of the 1294 patients, HF was present in 25%. Median follow-up was 6.7 years (IQR 4.9-9.3) years with 537 recorded deaths. Patients with CAD + HF had a higher incidence of dyssynchrony than patients with CAD alone (diastolic BW 28.8% for the HF + CAD vs 14.7% for the CAD alone). Patients with CAD + HF had a lower survival than CAD alone at 10 years (33%; 95% CI 27-40 vs 59; 95% CI 55-62, P < 0.0001). With one exception, HF was found to have no statistically significant interaction with dyssynchrony measures in unadjusted and adjusted survival models. CONCLUSIONS Patients with CAD + HF have a high prevalence of mechanical dyssynchrony as measured by GSPECT MPI, and a higher mortality than CAD alone. However, clinical outcomes associated with mechanical dyssynchrony did not differ in patients with and without HF.
Collapse
Affiliation(s)
- Marat Fudim
- Division of Cardiology, Duke Department of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA.
- Duke Clinical Research Institute, Durham, NC, USA.
| | - Mouhammad Fathallah
- Division of Cardiology, Duke Department of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Linda K Shaw
- Division of Cardiology, Duke Department of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Olga James
- Division of Nuclear Medicine, Duke Department of Radiology, Durham, NC, USA
| | - Zainab Samad
- Division of Cardiology, Duke Department of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Jonathan P Piccini
- Division of Cardiology, Duke Department of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Paul L Hess
- VA Eastern Colorado and Health Care System, Denver, CO, USA
| | | |
Collapse
|
11
|
Sabbah MS, Fayyaz AU, de Denus S, Felker GM, Borlaug BA, Dasari S, Carter RE, Redfield MM. Obese-Inflammatory Phenotypes in Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2020; 13:e006414. [PMID: 32809874 DOI: 10.1161/circheartfailure.119.006414] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Comorbidity-driven microvascular inflammation is posited as a unifying pathophysiologic mechanism for heart failure with preserved ejection fraction (HFpEF). Obesity is proinflammatory and common in HFpEF. We hypothesized that unique obesity-inflammation HFpEF phenotypes exist and are associated with differences in clinical features, fibrosis biomarkers, and functional performance. METHODS Patients (n=301) from 3 HFpEF clinical trials were studied. Unsupervised machine learning (hierarchical clustering) with obese status and 13 inflammatory biomarkers as input variables was performed. Associations of clusters with HFpEF severity and fibrosis biomarkers (PIIINP [procollagen III N-terminal peptide], CITP [C-telopeptide for type I collagen], IGFBP7 [insulin-like growth factor-binding protein-7], and GAL-3 [galectin-3]) were assessed. RESULTS Hierarchical clustering revealed 3 phenotypes: pan-inflammatory (n=129; 64% obese), noninflammatory (n=83; 55% obese), and obese high CRP (C-reactive protein; n=89; 98% obese). The pan-inflammatory phenotype had more comorbidities and heart failure hospitalizations; higher left atrial volume, NT-proBNP (N-terminal pro-B-type natriuretic peptide), and fibrosis biomarkers; and lower glomerular filtration rate, peak oxygen consumption, 6-minute walk distance, and active hours/day (P<0.05 for all). The noninflammatory phenotype had the most favorable values for all measures. The obese high CRP phenotype resembled the noninflammatory phenotype except for isolated elevation of CRP and lower functional performance. Hierarchical cluster assignment was independent of CRP genotype combinations that alter CRP levels and more biologically plausible than other clustering approaches. Multiple traditional analytic techniques confirmed and extended the hierarchical clustering findings. CONCLUSIONS Unique obesity-inflammation phenotypes exist in HFpEF and are associated with differences in comorbidity burden, HFpEF severity, and fibrosis. These data support comorbidity-driven microvascular inflammation as a pathophysiologic mechanism for many but not all HFpEF patients.
Collapse
Affiliation(s)
- Michael S Sabbah
- Department of Cardiovascular Disease (M.S.S., A.U.F., B.A.B., M.M.R.), Mayo Clinic, Rochester, MN.,Center for Regenerative Medicine (M.S.S.), Mayo Clinic, Rochester, MN
| | - Ahmed U Fayyaz
- Department of Cardiovascular Disease (M.S.S., A.U.F., B.A.B., M.M.R.), Mayo Clinic, Rochester, MN
| | - Simon de Denus
- Research Centre, Montreal Heart Institute, QC, Canada (S.d.D.).,Université de Montréal Beaulieu-Saucier Pharmacogenomics Center, QC, Canada (S.d.D.).,Department of Pharmacy, Université de Montréal, QC, Canada (S.d.D.)
| | - G Michael Felker
- Duke Clinical Research Institute, Duke University, Durham, NC (G.M.F.)
| | - Barry A Borlaug
- Department of Cardiovascular Disease (M.S.S., A.U.F., B.A.B., M.M.R.), Mayo Clinic, Rochester, MN
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL (S.D., R.E.C.)
| | - Rickey E Carter
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL (S.D., R.E.C.)
| | - Margaret M Redfield
- Department of Cardiovascular Disease (M.S.S., A.U.F., B.A.B., M.M.R.), Mayo Clinic, Rochester, MN
| |
Collapse
|
12
|
Tummalapalli SL, Zelnick LR, Andersen AH, Christenson RH, deFilippi CR, Deo R, Go AS, He J, Ky B, Lash JP, Seliger SL, Soliman EZ, Shlipak MG, Bansal N. Association of Cardiac Biomarkers With the Kansas City Cardiomyopathy Questionnaire in Patients With Chronic Kidney Disease Without Heart Failure. J Am Heart Assoc 2020; 9:e014385. [PMID: 32578483 PMCID: PMC7670503 DOI: 10.1161/jaha.119.014385] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background The Kansas City Cardiomyopathy Questionnaire (KCCQ) is a measure of heart failure (HF) health status. Worse KCCQ scores are common in patients with chronic kidney disease (CKD), even without diagnosed heart failure (HF). Elevations in the cardiac biomarkers GDF-15 (growth differentiation factor-15), galectin-3, sST2 (soluble suppression of tumorigenesis-2), hsTnT (high-sensitivity troponin T), and NT-proBNP (N-terminal pro-B-type natriuretic peptide) likely reflect subclinical HF in CKD. Whether cardiac biomarkers are associated with low KCCQ scores is not known. Methods and Results We studied participants with CKD without HF in the multicenter prospective CRIC (Chronic Renal Insufficiency Cohort) Study. Outcomes included (1) low KCCQ score <75 at year 1 and (2) incident decline in KCCQ score to <75. We used multivariable logistic regression and Cox regression models to evaluate the associations between baseline cardiac biomarkers and cross-sectional and longitudinal KCCQ scores. Among 2873 participants, GDF-15 (adjusted odds ratio 1.42 per SD; 99% CI, 1.19-1.68) and galectin-3 (1.28; 1.12-1.48) were significantly associated with KCCQ scores <75, whereas sST2, hsTnT, and NT-proBNP were not significantly associated with KCCQ scores <75 after multivariable adjustment. Of the 2132 participants with KCCQ ≥75 at year 1, GDF-15 (adjusted hazard ratio, 1.36 per SD; 99% CI, 1.12-1.65), hsTnT (1.20; 1.01-1.44), and NT-proBNP (1.30; 1.08-1.56) were associated with incident decline in KCCQ to <75 after multivariable adjustment, whereas galectin-3 and sST2 did not have significant associations with KCCQ decline. Conclusions Among participants with CKD without clinical HF, GDF-15, galectin-3, NT-proBNP, and hsTnT were associated with low KCCQ either at baseline or during follow-up. Our findings show that elevations in cardiac biomarkers reflect early symptomatic changes in HF health status in CKD patients.
Collapse
Affiliation(s)
| | - Leila R Zelnick
- Kidney Research Institute University of Washington Seattle WA.,Division of Nephrology Department of Medicine University of Washington Seattle WA
| | - Amanda H Andersen
- Biostatistics and Epidemiology and Informatics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Epidemiology Tulane University School of Public Health and Tropical Medicine New Orleans LA
| | | | | | - Rajat Deo
- Division of Cardiovascular Medicine University of Pennsylvania Philadelphia PA
| | - Alan S Go
- Department of Epidemiology and Biostatistics University of California, San Francisco San Francisco San Francisco CA.,Department of Medicine University of California San Francisco CA.,Division of Research Kaiser Permanente Northern California Oakland CA
| | - Jiang He
- Tulane University New Orleans LA
| | - Bonnie Ky
- Division of Cardiology Department of Medicine Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Biostatistics, Epidemiology & Informatics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - James P Lash
- Department of Medicine University of Illinois at Chicago IL
| | - Stephen L Seliger
- Division of Nephrology University of Maryland School of Medicine Baltimore MD
| | - Elsayed Z Soliman
- Department of Epidemiology and Prevention Epidemiological Cardiology Research Center Wake Forest University School of Medicine Winston-Salem NC
| | - Michael G Shlipak
- Kidney Health Research Collaborative University of California San Francisco CA.,San Francisco Veterans Affairs Medical Center San Francisco CA
| | - Nisha Bansal
- Kidney Research Institute University of Washington Seattle WA.,Division of Nephrology Department of Medicine University of Washington Seattle WA
| | | |
Collapse
|
13
|
The utility of growth differentiation factor-15, galectin-3, and sST2 as biomarkers for the diagnosis of heart failure with preserved ejection fraction and compared to heart failure with reduced ejection fraction: a systematic review. Heart Fail Rev 2020; 26:799-812. [PMID: 32472523 DOI: 10.1007/s10741-020-09913-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The objective was to evaluate the diagnosis of heart failure with preserved ejection fraction (HFpEF) using the biomarkers, growth differentiation factor-15 (GDF-15), galectin-3 (Gal-3), and soluble ST2 (sST2), and to determine whether they can differentiate HFpEF from heart failure with reduced ejection fraction (HFrEF). Medline and Embase databases were searched with the terms diastolic heart failure or HFpEF, biomarkers, and diagnosis, limited to years 2000 to 2019. There were significantly and consistently higher levels of GDF-15, Gal-3, and sST2 in HFpEF compared to no heart failure. Importantly, the magnitude of the increase in GDF-15 or Gal-3 and possibly sST2,correlated with a greater degree of diastolic dysfunction. There were no significant differences between GDF-15, Gal-3, and sST2 in patients with HFpEF vs HFrEF. In the studies assessing these three biomarkers, BNP was significantly greater in heart failure than controls. Furthermore, BNP was significantly higher in HFrEF compared to HFpEF. The diagnostic utility of GDF-15, Gal-3, and sST2 compared to BNP was evaluated by comparing ROC curves. The data supports the contention that to distinguish HFpEF from HFrEF, an index is needed that incorporates GDF-15, Gal-3, or sST2 as well as BNP. The three biomarkers GDF-15, Gal-3, or sST2 can identify patients with HFpEF compared to individuals without heart failure but cannot differentiate HFpEF from HFrEF. BNP is higher in and is better at differentiating HFrEF from HFpEF. Indices that incorporate GDF-15, Gal-3, or sST2 as well as BNP show promise in differentiating HFpEF from HFrEF.
Collapse
|
14
|
Fudim M, Kelly JP, Jones AD, AbouEzzeddine OF, Ambrosy AP, Greene SJ, Reddy YN, Anstrom KJ, Alhanti B, Lewis GD, Hernandez AF, Felker GM. Are existing and emerging biomarkers associated with cardiorespiratory fitness in patients with chronic heart failure? Am Heart J 2020; 220:97-107. [PMID: 31805424 DOI: 10.1016/j.ahj.2019.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiorespiratory fitness (CRF) is closely linked to health status and clinical outcomes in heart failure (HF) patients. We aimed to test whether biomarkers can reflect CRF and its change over time. METHODS This post hoc analysis used data from ambulatory cohorts of heart failure with reduced ejection fraction (HFrEF) (IRONOUT) and heart failure with preserved ejection fraction (HFpEF) (RELAX). Cardiopulmonary exercise testing, 6-minute walk distance (6MWD), and serum biomarkers were measured at baseline and 16- or 24-week follow-up (for IRONOUT and RELAX respectively). Biomarkers included N-terminal pro-B-type natriuretic peptide (NT-proBNP), soluble ST2, growth differentiation factor-15, and Galectin-3. RESULTS Analysis included 225 patients with HFrEF and 216 with HFpEF. Baseline peak VO2, VE/VCO2 slope, and 6MWD showed a mild correlation with the doubling of all 4 tested biomarkers in HFrEF and HFpEF. Following multivariable adjustment (including all biomarkers), the only significant association between change in biomarker and functional parameter in HFrEF was change in NT-proBNP and change in VE/VCO2 slope (3.596% increase per doubling, 95% CI 0.779-6.492, P = .012). In HFpEF, a decrease in peak VO2 was associated with an increase in NT-proBNP (-0.726 mL/min/kg per doubling, 95% CI -1.100 to -0.353, P < .001), and a decrease in 6MWD was associated with an increase in growth differentiation factor-15 (-31.606 m per doubling, 95% CI -61.404 to -1.809, P = .038). CONCLUSIONS In these ambulatory trial cohorts, NT-proBNP was associated with baseline and change in CRF in HFrEF and HFpEF. In contrast, novel biomarkers do not appear suitable as a reliable surrogate for serial assessment of exercise capacity in HF patients given lack of consistent independent association with CRF beyond traditional risk factors and NT-proBNP.
Collapse
|
15
|
Patel RB, Mehta R, Redfield MM, Borlaug BA, Hernandez AF, Shah SJ, Dubin RF. Renal Dysfunction in Heart Failure With Preserved Ejection Fraction: Insights From the RELAX Trial. J Card Fail 2020; 26:233-242. [PMID: 31931098 DOI: 10.1016/j.cardfail.2020.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/17/2019] [Accepted: 01/03/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Patients with heart failure with preserved ejection fraction (HFpEF) and chronic kidney disease (CKD) represent a high-risk phenotype. The Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure with Preserved Ejection Fraction (RELAX) trial enrolled a high proportion of CKD participants, allowing investigation into differences in HFpEF by CKD status. METHODS AND RESULTS Among 212 participants, we investigated the associations of CKD with biomarkers, cardiac structure, and exercise capacity, and identified predictors of change in estimated glomerular filtration rate (eGFR) over trial follow-up. CKD participants (eGFR ≤60 mL/min/1.73m2) were older, had more comorbidities, and had worse diastolic function. Lower eGFR was associated with higher levels of endothelin-1, N-terminal pro-B-type natriuretic peptide, aldosterone, uric acid, and biomarkers of fibrosis (P < .05 for all). Whereas lower eGFR was associated with worse peak oxygen consumption (VO2) after adjustment for demographics, clinical comorbidities, exercise modality, ejection fraction, and chronotropic index (β coefficient per 1 SD decrease in eGFR: -0.61, 95% CI: -1.01, -0.22, P = .002), this association was attenuated after further adjustment for hemoglobin (β coefficient: -0.26, 95% CI: -0.68, 0.16, P = .22). Hemoglobin mediated 35% of the association between eGFR and peak VO2. Sildenafil therapy was independently associated with worsening eGFR over the trial (β coefficient: -2.79, 95% CI: -5.34, -0.24, P = .03). CONCLUSION Renal dysfunction in HFpEF is characterized by echocardiographic and biomarker profiles indicative of more advanced disease, and reduced hemoglobin is a strong mediator of the association between renal dysfunction and low exercise capacity. Sildenafil therapy was associated with worsening of renal function in RELAX.
Collapse
Affiliation(s)
- Ravi B Patel
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Barry A Borlaug
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Adrian F Hernandez
- Division of Cardiology, Duke University Medical Center, Durham, North Carolina
| | - Sanjiv J Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ruth F Dubin
- Division of Nephrology, San Francisco VA Medical Center/University of California, San Francisco, California
| |
Collapse
|
16
|
Carnes J, Gordon G. Biomarkers in Heart Failure With Preserved Ejection Fraction: An Update on Progress and Future Challenges. Heart Lung Circ 2020; 29:62-68. [DOI: 10.1016/j.hlc.2019.05.180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/28/2022]
|
17
|
Du X, Zhao W, Nguyen M, Lu Q, Kiriazis H. β-Adrenoceptor activation affects galectin-3 as a biomarker and therapeutic target in heart disease. Br J Pharmacol 2019; 176:2449-2464. [PMID: 30756388 PMCID: PMC6592856 DOI: 10.1111/bph.14620] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a key histopathological component that drives the progression of heart disease leading to heart failure and constitutes a therapeutic target. Recent preclinical and clinical studies have implicated galectin-3 (Gal-3) as a pro-fibrotic molecule and a biomarker of heart disease and fibrosis. However, our knowledge is poor on the mechanism(s) that determine the blood level or regulate cardiac expression of Gal-3. Recent studies have demonstrated that enhanced β-adrenoceptor activity is a determinant of both circulating concentration and cardiac expression of Gal-3. Pharmacological or transgenic activation of β-adrenoceptors leads to increased blood levels of Gal-3 and up-regulated cardiac Gal-3 expression, effect that can be reversed with the use of β-adrenoceptor antagonists. Conversely, Gal-3 gene deletion confers protection against isoprenaline-induced cardiotoxicity and fibrogenesis. At the transcription level, β-adrenoceptor stimulation activates cardiac mammalian sterile-20-like kinase 1, a pivotal kinase of the Hippo signalling pathway, which is associated with Gal-3 up-regulation. Recent studies have suggested a role for the β-adrenoceptor-Hippo signalling pathway in the regulation of cardiac Gal-3 expression thereby contributing to the onset and progression of heart disease. This implies a therapeutic potential of the suppression of Gal-3 expression. In this review, we discuss the effects of β-adrenoceptor activity on Gal-3 as a biomarker and causative mediator in the setting of heart disease and point out pivotal knowledge gaps. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Physiology and Pathophysiology, School of Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wei‐Bo Zhao
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - My‐Nhan Nguyen
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - Qun Lu
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Cardiovascular Medicine, First HospitalXi'an Jiaotong University Health Science CenterXi'anChina
| | - Helen Kiriazis
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| |
Collapse
|
18
|
Reddy YNV, Lewis GD, Shah SJ, Obokata M, Abou-Ezzedine OF, Fudim M, Sun JL, Chakraborty H, McNulty S, LeWinter MM, Mann DL, Stevenson LW, Redfield MM, Borlaug BA. Characterization of the Obese Phenotype of Heart Failure With Preserved Ejection Fraction: A RELAX Trial Ancillary Study. Mayo Clin Proc 2019; 94:1199-1209. [PMID: 31272568 DOI: 10.1016/j.mayocp.2018.11.037] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/28/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To characterize the obese heart failure with preserved ejection fraction (HFpEF) phenotype in a multicenter cohort. PATIENTS AND METHODS This was a secondary analysis of the randomized clinical trial RELAX (Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure with Preserved Ejection Fraction) performed between October 1, 2008, and February 1, 2012. Patients with HFpEF were classified by body mass index (BMI) as obese (BMI≥35 kg/m2) and nonobese (BMI<30 kg/m2) for comparison. RESULTS Obese patients with HFpEF (n=81) were younger (median age, 64 [interquartile range (IQR), 67-79] years vs 73 [IQR, 56-70] years; P<.001) but had greater peripheral edema (31% [25] vs 9% [6]; P<.001), more orthopnea (76% [56] vs 53% [35]; P=.005), worse New York Heart Association class (P=.006), and more impaired quality of life (P<.001) as compared with nonobese patients with HFpEF (n=70). Despite more severe signs and symptoms, obese patients with HFpEF had lower N-terminal pro B-type natriuretic peptide level (median, 481 [IQR, 176-1183] pg/mL vs 825 [IQR, 380-1679] pg/mL [to convert to pmol/L, multiply by 0.118]; P=.007) and lower left atrial volume index (median, 38 [IQR, 31-47] mL/m2 vs 54 [IQR, 41-63] mL/m2; P<.001). Serum C-reactive protein (median, 5.0 [IQR, 2.4-9.9] mg/dL vs 2.7 [IQR, 1.6-5.4] mg/dL [to convert to mg/L, multiply by 10-3]; P<.001) and uric acid (median, 7.8 [IQR, 6.1-8.7] mg/dL vs 6.8 [IQR, 5.5-8.3] mg/dL; P=.03) levels were higher in obese HFpEF, indicating greater systemic inflammation, than in nonobese HFpEF. Peak oxygen consumption was impaired in obese HFpEF (median, 11.1 [IQR, 9.6-14.4] mL/kg per minute vs 13.1 [IQR, 11.3-14.7] mL/kg per minute; P=.008), as was submaximal exercise capacity (6-minute walk distance, 272 [IQR, 200-332] m vs 355 [IQR, 290-415] m; P<.0001). CONCLUSION Obese HFpEF is associated with decreased quality of life, worse symptoms of heart failure, greater systemic inflammation, worse exercise capacity, and higher metabolic cost of exertion as compared with nonobese HFpEF. Further study is required to understand the pathophysiology and potential distinct treatments for patients with the obese phenotype of HFpEF. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00763867.
Collapse
Affiliation(s)
| | - Gregory D Lewis
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Sanjiv J Shah
- Division of Cardiology, Northwestern University, Chicago, IL
| | - Masaru Obokata
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | | - Marat Fudim
- Division of Cardiology, Duke University, Durham, NC
| | - Jie-Lena Sun
- Division of Cardiology, Duke University, Durham, NC
| | | | | | | | - Douglas L Mann
- Division of Cardiology, Washington University, St. Louis, MO
| | | | | | - Barry A Borlaug
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN.
| |
Collapse
|
19
|
Abstract
Heart failure is a clinical condition with complex pathophysiology that involves many different processes. Diagnosis is often difficult in patients presenting for the first time with breathlessness. Many biomarkers have been identified that are elevated in heart failure and their role in assessing prognosis has also been investigated. However, at present the natriuretic peptides appear to be the gold standard biomarker against which the other biomarkers are compared. In this review we will examine the evidence behind the other biomarkers for use in heart failure patients and the current guidelines for their use.
Collapse
Affiliation(s)
- Sunil K Nadar
- Department of Medicine, Sultan Qaboos University Hospital Muscat, Oman
| | | |
Collapse
|
20
|
Stoica A, Şorodoc V, Lionte C, Jaba IM, Costache I, Anisie E, Tuchiluș C, Rusalim Petriș O, Sîrbu O, Jaba E, Ceasovschih A, Vâţă L, Şorodoc L. Acute cardiac dyspnea in the emergency department: diagnostic value of N-terminal prohormone of brain natriuretic peptide and galectin-3. J Int Med Res 2018; 47:159-172. [PMID: 30222016 PMCID: PMC6384479 DOI: 10.1177/0300060518798257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE This study was performed to determine whether a dual-biomarker approach using N-terminal prohormone of brain natriuretic peptide (NT-proBNP) and galectin-3 optimizes the diagnosis and risk stratification of acute cardiac dyspnea. Atypical clinical manifestations and overlapping pathologies require objective and effective diagnostic methods to avoid treatment delays. METHODS This prospective observational study included 208 patients who presented to the emergency department for acute dyspnea. NT-proBNP and galectin-3 were measured upon admission. The patients were divided into two groups according to the etiology of their clinical manifestations: cardiac and non-cardiac dyspnea. The patients' New York Heart Association functional class, left ventricular ejection fraction, and discharge status were assessed. RESULTS Diagnostic criteria for acute heart failure were fulfilled in 61.1% of the patients. NT-proBNP and galectin-3 were strongly and significantly correlated. Receiver operating characteristic analysis revealed similar areas under the curve for both markers in the entire group of patients as well as in the high-risk subsets of patients. CONCLUSIONS The diagnostic performance of NT-proBNP and galectin-3 is comparable for both the total population and high-risk subsets. Galectin-3 adds diagnostic value to the conventional NT-proBNP in patients with acute cardiac dyspnea, and its utility is of major interest in uncertain clinical situations.
Collapse
Affiliation(s)
- Alexandra Stoica
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Victoriţa Şorodoc
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Cătălina Lionte
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Irina M Jaba
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania
| | - Irina Costache
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | | | - Cristina Tuchiluș
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Ovidiu Rusalim Petriș
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Oana Sîrbu
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Elisabeta Jaba
- 3 Statistics Department, FEAA, "Al. I. Cuza" University Iași, Romania
| | - Alexandr Ceasovschih
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Luminiţa Vâţă
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| | - Laurenţiu Şorodoc
- 1 Gr. T. Popa University of Medicine and Pharmacy, Iași, Romania.,2 Sf. Spiridon Emergency Hospital Iași, Romania
| |
Collapse
|
21
|
Ansari U, Behnes M, Hoffmann J, Natale M, Fastner C, El-Battrawy I, Rusnak J, Kim SH, Lang S, Hoffmann U, Bertsch T, Borggrefe M, Akin I. Galectin-3 Reflects the Echocardiographic Grades of Left Ventricular Diastolic Dysfunction. Ann Lab Med 2018; 38:306-315. [PMID: 29611380 PMCID: PMC5895859 DOI: 10.3343/alm.2018.38.4.306] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/19/2017] [Accepted: 01/15/2018] [Indexed: 01/19/2023] Open
Abstract
Background The level of Galectin-3 (Gal-3) protein purportedly reflects an ongoing cardiac fibrotic process and has been associated with ventricular remodeling, which is instrumental in the development of heart failure with preserved ejection fraction (HFpEF) syndrome. The aim of this study was to investigate the potential use of Gal-3 in improved characterization of the grades of diastolic dysfunction as defined by echocardiography. Methods Seventy HFpEF patients undergoing routine echocardiography were prospectively enrolled in the present monocentric study. Blood samples for measurements of Gal-3 and amino-terminal pro-brain natriuretic peptide (NT-proBNP) were collected within 24 hours pre- or post-echocardiographic examination. The classification of patients into subgroups based on diastolic dysfunction grade permitted detailed statistical analyses of the derived data. Results The Gal-3 serum levels of all patients corresponded to echocardiographic indices, suggesting HFpEF (E/A, P=0.03 and E/E', P=0.02). Gal-3 was also associated with progressive diastolic dysfunction, and increased levels corresponded to the course of disease (P=0.012). Detailed analyses of ROC curves suggested that Gal-3 levels could discriminate patients with grade III diastolic dysfunction (area under the curve [AUC]=0.770, P=0.005). Conclusions Gal-3 demonstrates remarkable effectiveness in the diagnosis of patients suffering from severe grade diastolic dysfunction. Increasing levels of Gal-3 possibly reflect the progressive course of HFpEF, as classified by the echocardiographic grades of diastolic dysfunction.
Collapse
Affiliation(s)
- Uzair Ansari
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim.
| | - Michael Behnes
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Julia Hoffmann
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Michele Natale
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Christian Fastner
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Ibrahim El-Battrawy
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Jonas Rusnak
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Seung Hyun Kim
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Siegfried Lang
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Ursula Hoffmann
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Martin Borggrefe
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| | - Ibrahim Akin
- First Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany: DZHK (German Center for Cardiovascular Research) partner site Mannheim
| |
Collapse
|
22
|
High-sensitivity C-reactive protein in heart failure with preserved ejection fraction. PLoS One 2018; 13:e0201836. [PMID: 30114262 PMCID: PMC6095520 DOI: 10.1371/journal.pone.0201836] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/17/2018] [Indexed: 11/19/2022] Open
Abstract
Background Microvascular inflammation may contribute to the pathogenesis of both heart failure with preserved ejection fraction (HFpEF) and pulmonary hypertension (PH). We investigated whether the inflammation biomarker C-reactive protein (CRP) was associated with clinical characteristics, disease severity or PH in HFpEF. Methods Patients in the Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Diastolic Heart failure (RELAX) trial had baseline high-sensitivity CRP levels measured (n = 214). Clinical characteristics, exercise performance, echocardiographic variables and biomarkers of neurohumoral activation, fibrosis and myocardial necrosis were assessed. Patients with normal (≤3mg/L) versus high (>3mg/L) CRP levels were compared. Results The median CRP level was 3.69mg/L. CRP was elevated in 57% of patients. High CRP levels were associated with younger age, higher body mass index (BMI), chronic obstructive pulmonary disease (COPD), lower peak oxygen consumption and higher endothelin-1 and aldosterone levels. CRP increased progressively with the number of comorbidities (0.7mg/L per increment in comorbidity number, P = 0.02). Adjusting for age, BMI and statin use, high CRP levels were additionally associated with atrial fibrillation, right ventricular dysfunction, and higher N-terminal pro-B-type natriuretic peptide levels (P<0.05 for all). CRP was not associated with PH or left ventricular function. CRP did not identify responders to sildenafil(P-value for interaction 0.13). Conclusions In HFpEF, high CRP is associated with greater comorbidity burden and some markers of disease severity but CRP was normal in 40% of patients. These findings support the presence of comorbidity-driven systemic inflammation in HFpEF but also the need to study other biomarkers which may better reflect the presence of systemic inflammation.
Collapse
|
23
|
Oikonomou E, Vogiatzi G, Tsalamandris S, Mourouzis K, Siasos G, Lazaros G, Skotsimara G, Marinos G, Vavuranakis M, Tousoulis D. Non-natriuretic peptide biomarkers in heart failure with preserved and reduced ejection fraction. Biomark Med 2018; 12:783-797. [PMID: 29865857 DOI: 10.2217/bmm-2017-0376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Heart failure (HF) with reduced and preserved ejection fraction constitutes two entities with distinct pathogenetic backgrounds sharing common features. Beyond natriuretic peptides, several novel biomarkers have been proven useful in the diagnosis, prognosis and treatment of HF. Biomarkers of myocardial fibrosis have a low diagnostic yield in subjects with acute HF but may add prognostic information, especially in patients with HF and preserved ejection fraction. Biomarkers of renal impairment identify subjects with worse prognosis independently of left ventricle ejection fraction while inflammatory markers have not been proven useful in patients with systolic or diastolic impairment. In this review article, we summarize the main differences and application of non-natriuretic peptide biomarkers in HF patients with preserved and reduced ejection fraction.
Collapse
Affiliation(s)
- Evangelos Oikonomou
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Georgia Vogiatzi
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Sotiris Tsalamandris
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Konstantinos Mourouzis
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Gerasimos Siasos
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - George Lazaros
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Georgia Skotsimara
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - George Marinos
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Athens, 11528, Greece
| |
Collapse
|
24
|
Michalska-Kasiczak M, Bielecka-Dabrowa A, von Haehling S, Anker SD, Rysz J, Banach M. Biomarkers, myocardial fibrosis and co-morbidities in heart failure with preserved ejection fraction: an overview. Arch Med Sci 2018; 14:890-909. [PMID: 30002709 PMCID: PMC6040115 DOI: 10.5114/aoms.2018.76279] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is steadily increasing. Its diagnosis remains difficult and controversial and relies mostly on non-invasive echocardiographic detection of left ventricular diastolic dysfunction and elevated filling pressures. The large phenotypic heterogeneity of HFpEF from pathophysiologic al underpinnings to clinical manifestations presents a major obstacle to the development of new therapies targeted towards specific HF phenotypes. Recent studies suggest that natriuretic peptides have the potential to improve the diagnosis of early HFpEF, but they still have significant limitations, and the cut-off points for diagnosis and prognosis in HFpEF remain open to debate. The purpose of this review is to present potential targets of intervention in patients with HFpEF, starting with myocardial fibrosis and methods of its detection. In addition, co-morbidities are discussed as a means to treat HFpEF according to cut-points of biomarkers that are different from usual. Biomarkers and approaches to co-morbidities may be able to tailor therapies according to patients' pathophysiological needs. Recently, soluble source of tumorigenicity 2 (sST2), growth differentiation factor 15 (GDF-15), galectin-3, and other cardiac markers have emerged, but evidence from large cohorts is still lacking. Furthermore, the field of miRNA is a very promising area of research, and further exploration of miRNA may offer diagnostic and prognostic applications and insight into the pathology, pointing to new phenotype-specific therapeutic targets.
Collapse
Affiliation(s)
- Marta Michalska-Kasiczak
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
- Department of Endocrine Disorders and Bone Metabolism, 1 Chair of Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Agata Bielecka-Dabrowa
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
| | - Stefan D. Anker
- Division of Cardiology and Metabolism – Heart Failure, Cachexia and Sarcopenia, Department of Cardiology, Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
25
|
Nguyen MN, Su Y, Vizi D, Fang L, Ellims AH, Zhao WB, Kiriazis H, Gao XM, Sadoshima J, Taylor AJ, McMullen JR, Dart AM, Kaye DM, Du XJ. Mechanisms responsible for increased circulating levels of galectin-3 in cardiomyopathy and heart failure. Sci Rep 2018; 8:8213. [PMID: 29844319 PMCID: PMC5973942 DOI: 10.1038/s41598-018-26115-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Galectin-3 is a biomarker of heart disease. However, it remains unknown whether increase in galectin-3 levels is dependent on aetiology or disease-associated conditions and whether diseased heart releases galectin-3 into the circulation. We explored these questions in mouse models of heart disease and in patients with cardiomyopathy. All mouse models (dilated cardiomyopathy, DCM; fibrotic cardiomyopathy, ischemia-reperfusion, I/R; treatment with β-adrenergic agonist isoproterenol) showed multi-fold increases in cardiac galectin-3 expression and preserved renal function. In mice with fibrotic cardiomyopathy, I/R or isoproterenol treatment, plasma galectin-3 levels and density of cardiac inflammatory cells were elevated. These models also exhibited parallel changes in cardiac and plasma galectin-3 levels and presence of trans-cardiac galectin-3 gradient, indicating cardiac release of galectin-3. DCM mice showed no change in circulating galectin-3 levels nor trans-cardiac galectin-3 gradient or myocardial inflammatory infiltration despite a 50-fold increase in cardiac galectin-3 content. In patients with hypertrophic cardiomyopathy or DCM, plasma galectin-3 increased only in those with renal dysfunction and a trans-cardiac galectin-3 gradient was not present. Collectively, this study documents the aetiology-dependency and diverse mechanisms of increment in circulating galectin-3 levels. Our findings highlight cardiac inflammation and enhanced β-adrenoceptor activation in mediating elevated galectin-3 levels via cardiac release in the mechanism.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Yidan Su
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Donna Vizi
- Department of Cardiovascular Medicine, the Alfred Hospital, Melbourne, Australia
| | - Lu Fang
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiovascular Medicine, the Alfred Hospital, Melbourne, Australia
| | - Andris H Ellims
- Department of Cardiovascular Medicine, the Alfred Hospital, Melbourne, Australia
| | - Wei-Bo Zhao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine Rutgers, New Jersey Medical School, New Jersey, USA
| | - Andrew J Taylor
- Department of Cardiovascular Medicine, the Alfred Hospital, Melbourne, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Anthony M Dart
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia.,Department of Cardiovascular Medicine, the Alfred Hospital, Melbourne, Australia
| | - David M Kaye
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia.,Department of Cardiovascular Medicine, the Alfred Hospital, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia. .,Central Clinical School, Monash University, Melbourne, Australia.
| |
Collapse
|
26
|
Navarro-Alvarez N, Goncalves B, Andrews AR, Wang Z, Wang Z, Harrington E, Shah J, Sachs DH, Eliaz I, Huang CA. The effects of galectin-3 depletion apheresis on induced skin inflammation in a porcine model. J Clin Apher 2018; 33:486-493. [PMID: 29572917 DOI: 10.1002/jca.21624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/26/2018] [Accepted: 03/01/2018] [Indexed: 12/20/2022]
Abstract
Galectin-3 (Gal-3), a β-galactoside-binding lectin that is expressed in mammalian cells, is known to modulate several biological functions such as cell-cell adhesion, macrophage activation, angiogenesis, metastasis, and fibrosis. The goal of this study was to evaluate the ability of Gal-3 depletion apheresis using an adsorption column with immobilized anti-Gal-3-antibody to reduce inflammation induced by Complete Freund's Adjuvant injection in a skin inflammation porcine model. Here, we report that plasma perfusion by apheresis through a Gal-3 binding immuno-affinity column reduces plasma Gal-3 levels to below limits of quantitative detection, and results in significant decrease in skin inflammation, including degree and duration of inflammatory lesions. Human plasma was tested ex vivo and found to be efficiently depleted using the anti-Gal-3 affinity column. This study demonstrates the potential of Gal-3 depletion apheresis as a therapeutic method for inflammation-mediated disease, supporting continued research in this area for clinical application.
Collapse
Affiliation(s)
- Nalu Navarro-Alvarez
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Beatriz Goncalves
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alec R Andrews
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhaohui Wang
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhirui Wang
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Edward Harrington
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jigesh Shah
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David H Sachs
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, California
| | - Christene A Huang
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Abstract
Abstract
Heart failure is nowadays a common condition associated with high mortality and increased healthcare-related costs. Over the years, the research on heart failure management has been extensive in order to better diagnose and treat the condition. Since the progression of left ventricular dysfunction is a consequence of myocardial inflammation, apopotosis, and fibrosis leading to myocardium remodelling, several molecules that are involved in the inflammation pathways have been explored as possible biomarkers for the condition. The study of biomarkers and their key roles in inflammation could allow early identification of patients with heart failure, improve prognostic assessment, and provide a target for future therapies. Among currently studied biomarkers, extensive research has been conducted on galectin-3, a galactoside-binding lectin, which is synthetised and secreted when cardiomyocytes and fibroblasts are submitted to mechanical stress. Accordingly, it has been hypothesised that galectin-3 could be a promoter of left ventricular dysfunction. Galectin-3 has been shown to mediate inflammation by several different pathways which are further detailed in the current review. Also, we aimed to provide a comprehensive overview of existing evidence on the utility of galectin-3 in clinical settings associated with heart failure.
Collapse
|
28
|
Dencker M, Arvidsson D, Karlsson MK, Wollmer P, Andersen LB, Thorsson O. Galectin-3 levels relate in children to total body fat, abdominal fat, body fat distribution, and cardiac size. Eur J Pediatr 2018; 177:461-467. [PMID: 29327139 PMCID: PMC5816767 DOI: 10.1007/s00431-017-3079-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/27/2022]
Abstract
UNLABELLED Galectin-3 has recently been proposed as a novel biomarker for cardiovascular disease in adults. The purpose of this investigation was to assess relationships between galectin-3 levels and total body fat, abdominal fat, body fat distribution, aerobic fitness, blood pressure, left ventricular mass, left atrial size, and increase in body fat over a 2-year period in a population-based sample of children. Our study included 170 children aged 8-11 years. Total fat mass and abdominal fat were measured by dual-energy x-ray absorptiometry (DXA). Body fat distribution was expressed as abdominal fat/total fat mass. Maximal oxygen uptake was assessed by indirect calorimetry during a maximal exercise test and scaled to body mass. Systolic and diastolic blood pressure and pulse pressure were measured. Left atrial size, left ventricular mass, and relative wall thickness were measured by echocardiography. Frozen serum samples were analyzed for galectin-3 by the Proximity Extension Assay technique. A follow-up DXA scan was performed in 152 children 2 years after the baseline exam. Partial correlations, with adjustment for sex and age, between galectin-3 versus body fat measurements indicated weak to moderate relationships. Moreover, left atrial size, left ventricular mass, and relative wall thickness and pulse pressure were also correlated with galectin-3. Neither systolic blood pressure nor maximal oxygen uptake was correlated with galectin-3. There was also a correlation between galectin-3 and increase in total body fat over 2 years, while no such correlations were found for the other fat measurements. CONCLUSION More body fat and abdominal fat, more abdominal body fat distribution, more left ventricular mass, and increased left atrial size were all associated with higher levels of galectin-3. Increase in total body fat over 2 years was also associated with higher levels of galectin-3. What is Known: • Galectin-3 has been linked to obesity and been proposed to be a novel biomarker for cardiovascular disease in adults. • Information on this subject in children is very scarce. What is New: • The present study demonstrates a relationship between galectin-3 levels and total body fat, abdominal fat, body fat distribution, cardiac size and geometry, and increase in total body fat over 2 years in young children.
Collapse
Affiliation(s)
- Magnus Dencker
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, 205 02, Malmö, Sweden.
| | - Daniel Arvidsson
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, 205 02 Malmö, Sweden ,Center for Health and Performance, Department of Food and Nutrition, and Sports Science, University of Gothenburg, Gothenburg, Sweden
| | - Magnus K. Karlsson
- Department of Clinical Sciences and Orthopaedics, Clinical and Molecular Osteoporosis Research Unit, Skåne University Hospital, Lund University, Lund, Sweden
| | - Per Wollmer
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, 205 02 Malmö, Sweden
| | - Lars B. Andersen
- Department of Teacher Education and Sport, Sogn and Fjordane University College, Sogndal, Norway ,Department of Sports Medicine, Norwegian School of Sport Sciences, Oslo, Norway
| | - Ola Thorsson
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, 205 02 Malmö, Sweden
| |
Collapse
|
29
|
Senthong V, Kirsop JL, Tang WHW. Clinical Phenotyping of Heart Failure with Biomarkers: Current and Future Perspectives. Curr Heart Fail Rep 2017; 14:106-116. [PMID: 28205040 DOI: 10.1007/s11897-017-0321-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Heart failure (HF) is a complex clinical syndrome with diverse risk factors and etiologies, differing underlying pathophysiology, and large phenotypic heterogeneity. RECENT FINDINGS Advances in imaging techniques coupled with clinical trials that targeted only in those with impaired left ventricular ejection fraction (LVEF) have largely shaped the current management strategy for HF that focuses predominantly in patients with systolic HF. In contrast, there are no effective treatments for HF with preserved ejection fraction (HFpEF). Instead of this "one-size-fits-all" approach to treatment, better precision to define HF phenotypic classifications may lead to more efficient and effective HF disease management. CONCLUSION Integrating variables-including clinical variables, HF biomarkers, imaging, genotypes, metabolomics, and proteomics-can identify different pathophysiologies, lead to more precise phenotypic classification, and warrant investigation in future clinical trials.
Collapse
Affiliation(s)
- Vichai Senthong
- Department of Cardiovascular Medicine, Heart and Vascular Institute, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44915, USA.,Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jennifer L Kirsop
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland, OH, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44915, USA. .,Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland, OH, USA. .,Center for Clinical Genomics, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
30
|
Dong R, Zhang M, Hu Q, Zheng S, Soh A, Zheng Y, Yuan H. Galectin-3 as a novel biomarker for disease diagnosis and a target for therapy (Review). Int J Mol Med 2017; 41:599-614. [PMID: 29207027 PMCID: PMC5752178 DOI: 10.3892/ijmm.2017.3311] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/29/2017] [Indexed: 01/03/2023] Open
Abstract
Galectin-3 is a member of the galectin family, which are β‑galactoside‑binding lectins with ≥1 evolutionary conserved carbohydrate‑recognition domain. It binds proteins in a carbohydrate‑dependent and ‑independent manner. Galectin‑3 is predominantly located in the cytoplasm; however, it shuttles into the nucleus and is secreted onto the cell surface and into biological fluids including serum and urine. It serves important functions in numerous biological activities including cell growth, apoptosis, pre‑mRNA splicing, differentiation, transformation, angiogenesis, inflammation, fibrosis and host defense. Numerous previous studies have indicated that galectin‑3 may be used as a diagnostic or prognostic biomarker for certain types of heart disease, kidney disease and cancer. With emerging evidence to support the function and application of galectin‑3, the current review aims to summarize the latest literature regarding the biomarker characteristics and potential therapeutic application of galectin‑3 in associated diseases.
Collapse
Affiliation(s)
- Rui Dong
- Department of Pediatric Hepatobiliary Surgery, Children's Hospital of Fudan University and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 200433, P.R. China
| | - Min Zhang
- Medical College, Xizang Minzu University, Xianyang, Shaanxi 712000, P.R. China
| | - Qunying Hu
- Medical College, Xizang Minzu University, Xianyang, Shaanxi 712000, P.R. China
| | - Shan Zheng
- Department of Pediatric Hepatobiliary Surgery, Children's Hospital of Fudan University and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 200433, P.R. China
| | - Andrew Soh
- Medical Scientific Affairs, Abbott Diagnostics Division, Abbott Laboratories, Shanghai 200032, P.R. China
| | - Yijie Zheng
- Medical Scientific Affairs, Abbott Diagnostics Division, Abbott Laboratories, Shanghai 200032, P.R. China
| | - Hui Yuan
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
31
|
Afsar B, Rossignol P, van Heerebeek L, Paulus WJ, Damman K, Heymans S, van Empel V, Sag A, Maisel A, Kanbay M. Heart failure with preserved ejection fraction: a nephrologist-directed primer. Heart Fail Rev 2017; 22:765-773. [DOI: 10.1007/s10741-017-9619-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Van Tassell BW, Buckley LF, Carbone S, Trankle CR, Canada JM, Dixon DL, Abouzaki N, Oddi-Erdle C, Biondi-Zoccai G, Arena R, Abbate A. Interleukin-1 blockade in heart failure with preserved ejection fraction: rationale and design of the Diastolic Heart Failure Anakinra Response Trial 2 (D-HART2). Clin Cardiol 2017; 40:626-632. [PMID: 28475816 DOI: 10.1002/clc.22719] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 02/05/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of confirmed HF cases in the United States. However, there are no highly effective evidence-based treatments currently available for these patients. Inflammation correlates positively with adverse outcomes in HF patients. Interleukin (IL)-1, a prototypical inflammatory cytokine, has been implicated as a driver of diastolic dysfunction in preclinical animal models and a pilot clinical trial. The Diastolic Heart Failure Anakinra Response Trial 2 (D-HART2) is a phase 2, 2:1 randomized, double-blind, placebo-controlled clinical trial that will test the hypothesis that IL-1 blockade with anakinra (recombinant human IL-1 receptor antagonist) improves (1) cardiorespiratory fitness, (2) objective evidence of diastolic dysfunction, and (3) elevated inflammation in patients with HFpEF (http://www.ClinicalTrials.gov NCT02173548). The co-primary endpoints will be placebo-corrected interval changes in peak oxygen consumption and ventilatory efficiency at week 12. In addition, secondary and exploratory analyses will investigate the effects of IL-1 blockade on cardiac structure and function, systemic inflammation, endothelial function, quality of life, body composition, nutritional status, and clinical outcomes. The D-HART2 clinical trial will add to the growing body of evidence on the role of inflammation in cardiovascular disease, specifically focusing on patients with HFpEF.
Collapse
Affiliation(s)
- Benjamin W Van Tassell
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond.,VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Leo F Buckley
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond.,VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Salvatore Carbone
- VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond.,Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Cory R Trankle
- VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Justin M Canada
- VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Dave L Dixon
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond.,VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Nayef Abouzaki
- VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Claudia Oddi-Erdle
- VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - Giuseppe Biondi-Zoccai
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago
| | - Antonio Abbate
- VCU Johnson Center for Pulmonary and Critical Care Research, Virginia Commonwealth University, Richmond.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond
| |
Collapse
|
33
|
Clinical Correlates and Prognostic Value of Plasma Galectin-3 Levels in Degenerative Aortic Stenosis: A Single-Center Prospective Study of Patients Referred for Invasive Treatment. Int J Mol Sci 2017; 18:ijms18050947. [PMID: 28468272 PMCID: PMC5454860 DOI: 10.3390/ijms18050947] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/24/2017] [Accepted: 04/25/2017] [Indexed: 01/06/2023] Open
Abstract
Galectin-3 (Gal-3), a β-galactoside-binding lectin, has been implicated in myocardial fibrosis, development of left ventricular (LV) dysfunction and transition from compensated LV hypertrophy to overt heart failure (HF), being a novel prognostic marker in HF. Risk stratification is crucial for the choice of the optimal therapy in degenerative aortic stenosis (AS), affecting elderly subjects with coexistent diseases. Our aim was to assess correlates and prognostic value of circulating Gal-3 in real-world patients with degenerative AS referred for invasive treatment. Gal-3 levels were measured at admission in 80 consecutive patients with symptomatic degenerative AS (mean age: 79 ± 8 years; aortic valve area (AVA) index: 0.4 ± 0.1 cm²/m²). The therapeutic strategy was chosen following a dedicated multidisciplinary team-oriented approach, including surgical valve replacement (n = 11), transcatheter valve implantation (n = 19), balloon aortic valvuloplasty (BAV) (n = 25) and optimal medical therapy (n = 25). Besides routine echocardiographic indices, valvulo-arterial impedance (Zva), an index of global LV afterload, was computed. There were 22 deaths over a median follow-up of 523 days. Baseline Gal-3 correlated negatively with estimated glomerular filtration rate (eGFR) (r = -0.61, p < 0.001) and was unrelated to age, symptomatic status, AVA index, LV ejection fraction, LV mass index or Zva. For the study group as a whole, Gal-3 tended to predict mortality (Gal-3 >17.8 vs. Gal-3 <17.8 ng/mL; hazard ratio (HR): 2.03 (95% confidence interval, 0.88-4.69), p = 0.09), which was abolished upon adjustment for eGFR (HR: 1.70 (0.61-4.73), p = 0.3). However, in post-BAV patients multivariate-adjusted pre-procedural Gal-3 was associated with worse survival (HR: 7.41 (1.52-36.1), p = 0.01) regardless of eGFR. In conclusion, the inverse eGFR-Gal-3 relationship underlies a weak association between Gal-3 and adverse outcome in patients with degenerative AS referred for invasive therapy irrespective of type of treatment employed. In contrast, pre-procedural Gal-3 appears an independent mortality predictor in high-risk AS patients undergoing BAV.
Collapse
|
34
|
Besler C, Lang D, Urban D, Rommel KP, von Roeder M, Fengler K, Blazek S, Kandolf R, Klingel K, Thiele H, Linke A, Schuler G, Adams V, Lurz P. Plasma and Cardiac Galectin-3 in Patients With Heart Failure Reflects Both Inflammation and Fibrosis. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003804. [DOI: 10.1161/circheartfailure.116.003804] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 02/07/2017] [Indexed: 12/20/2022]
Abstract
Background—
Galectin (Gal)-3 is a β-galactoside-binding lectin and currently intensely studied as a biomarker in heart failure. Gal-3 also exerts proinflammatory effects, at least in extracardiac tissues. Objective of this study was to characterize the relationship of plasma and myocardial Gal-3 levels with cardiac fibrosis and inflammation in patients with nonischemic dilated cardiomyopathy and inflammatory cardiomyopathy (iCMP).
Methods and Results—
Endomyocardial biopsies and blood samples were obtained from patients with newly diagnosed cardiomyopathy and clinical suspicion of myocarditis. According to histopathologic findings, patients were classified as having dilated cardiomyopathy (n=40) or iCMP (n=75). Cardiac fibrosis was assessed histologically on endomyocardial biopsy sections. In patients with iCMP, myocardial Gal-3 expression significantly correlated with inflammatory cell count on endomyocardial biopsy (
r
=0.56;
P
<0.05). In contrast, an inverse association was observed between myocardial Gal-3 expression and cardiac fibrosis in patients with iCMP (
r
=−0.59;
P
<0.05). In patients with dilated cardiomyopathy, myocardial Gal-3 expression correlated with cardiac fibrosis on left ventricular biopsy (
P
=0.63;
P
<0.01). Of note, in both groups, plasma Gal-3 levels did not correlate with myocardial Gal-3 levels or left ventricular fibrosis, whereas a positive correlation between plasma Gal-3 levels and inflammatory cell count on endomyocardial biopsy was observed in patients with iCMP.
Conclusions—
The present study suggests that myocardial Gal-3 can be considered as a possible marker for both cardiac inflammation and fibrosis, depending on the pathogenesis of heart failure. However, circulating concentrations of Gal-3 do not seem to reflect endomyocardial Gal-3 levels or cardiac fibrosis.
Collapse
Affiliation(s)
- Christian Besler
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - David Lang
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Daniel Urban
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Karl-Philipp Rommel
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Maximilian von Roeder
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Karl Fengler
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Stephan Blazek
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Reinhard Kandolf
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Karin Klingel
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Holger Thiele
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Axel Linke
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Gerhard Schuler
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Volker Adams
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| | - Philipp Lurz
- From the Department of Internal Medicine/Cardiology, University of Leipzig-Heart Center, Germany (C.B., D.L., D.U., K.-P.R., M.v.R., K.F., S.B., A.L., G.S., V.A., P.L.); Institute for Pathology and Neuropathology, Department of Molecular Pathology, University Hospital Tübingen, Germany (R.K., K.K.); and Medical Clinic II, University Heart Center Lübeck, University Hospital Schleswig-Holstein, Germany (H.T.)
| |
Collapse
|
35
|
|
36
|
The potential of targeting epigenetic regulators for the treatment of fibrotic cardiac diseases. Future Med Chem 2016; 8:1533-6. [PMID: 27556930 DOI: 10.4155/fmc-2016-0144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
37
|
Abstract
Despite >100 clinical trials, only 2 new drugs had been approved by the US Food and Drug Administration for the treatment of chronic heart failure in more than a decade: the aldosterone antagonist eplerenone in 2003 and a fixed dose combination of hydralazine-isosorbide dinitrate in 2005. In contrast, 2015 has witnessed the Food and Drug Administration approval of 2 new drugs, both for the treatment of chronic heart failure with reduced ejection fraction: ivabradine and another combination drug, sacubitril/valsartan or LCZ696. Seemingly overnight, a range of therapeutic possibilities, evoking new physiological mechanisms, promise great hope for a disease that often carries a prognosis worse than many forms of cancer. Importantly, the newly available therapies represent a culmination of basic and translational research that actually spans many decades. This review will summarize newer drugs currently being used in the treatment of heart failure, as well as newer strategies increasingly explored for their utility during the stages of the heart failure syndrome.
Collapse
Affiliation(s)
- Anjali Tiku Owens
- From the Cardiovascular Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Susan C Brozena
- From the Cardiovascular Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Mariell Jessup
- From the Cardiovascular Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia.
| |
Collapse
|
38
|
Zile MR, Jhund PS, Baicu CF, Claggett BL, Pieske B, Voors AA, Prescott MF, Shi V, Lefkowitz M, McMurray JJV, Solomon SD. Plasma Biomarkers Reflecting Profibrotic Processes in Heart Failure With a Preserved Ejection Fraction: Data From the Prospective Comparison of ARNI With ARB on Management of Heart Failure With Preserved Ejection Fraction Study. Circ Heart Fail 2016; 9:CIRCHEARTFAILURE.115.002551. [PMID: 26754625 DOI: 10.1161/circheartfailure.115.002551] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Heart failure with preserved ejection fraction is a clinical syndrome that has been associated with changes in the extracellular matrix. The purpose of this study was to determine whether profibrotic biomarkers accurately reflect the presence and severity of disease and underlying pathophysiology and modify response to therapy in patients with heart failure with preserved ejection fraction. METHODS AND RESULTS Four biomarkers, soluble form of ST2 (an interleukin-1 receptor family member), galectin-3, matrix metalloproteinase-2, and collagen III N-terminal propeptide were measured in the Prospective Comparison of ARNI With ARB on Management of Heart Failure With Preserved Ejection Fraction (PARAMOUNT) trial at baseline, 12 and 36 weeks after randomization to valsartan or LCZ696. We examined the relationship between baseline biomarkers, demographic and echocardiographic characteristics, change in primary (change in N-terminal pro B-type natriuretic peptide) and secondary (change in left atrial volume) end points. The median (interquartile range) value for soluble form of ST2 (33 [24.6-48.1] ng/mL) and galectin 3 (17.8 [14.1-22.8] ng/mL) were higher, and for matrix metalloproteinase-2 (188 [155.5-230.6] ng/mL) lower, than in previously published referent controls; collagen III N-terminal propeptide (5.6 [4.3-6.9] ng/mL) was similar to referent control values. All 4 biomarkers correlated with severity of disease as indicated by N-terminal pro B-type natriuretic peptide, E/E', and left atrial volume. Baseline biomarkers did not modify the response to LCZ696 for lowering N-terminal pro B-type natriuretic peptide; however, left atrial volume reduction varied by baseline level of soluble form of ST2 and galectin 3; patients with values less than the observed median (<33 ng/mL soluble form of ST2 and <17.8 ng/mL galectin 3) had reduction in left atrial volume, those above median did not. Although LCZ696 reduced N-terminal pro B-type natriuretic peptide, levels of the other 4 biomarkers were not affected over time. CONCLUSIONS In patients with heart failure with preserved ejection fraction, biomarkers that reflect collagen homeostasis correlated with the presence and severity of disease and underlying pathophysiology, and may modify the structural response to treatment. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00887588.
Collapse
Affiliation(s)
- Michael R Zile
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.).
| | - Pardeep S Jhund
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Catalin F Baicu
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Brian L Claggett
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Burkert Pieske
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Adriaan A Voors
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Margaret F Prescott
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Victor Shi
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Martin Lefkowitz
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - John J V McMurray
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | - Scott D Solomon
- From the Department of Medicine, Medical University of South Carolina, Charleston (M.R.Z., C.F.B.); RHJ Department of Veterans Affairs Medical Center, Charleston, SC (M.R.Z., C.F.B.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (P.S.J., B.L.C., S.D.S.); BHF Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (P.S.J., J.J.V.M.M.); Department of Cardiology, Medical University of Graz, Graz, Austria (B.P.); Department of Internal Medicine and Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany (B.P.); Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany (B.P.); Department of Cardiology, University of Groningen, Groningen, The Netherlands (A.A.V.); and Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., V.S., M.L.)
| | | |
Collapse
|
39
|
Horn MA, Trafford AW. Aging and the cardiac collagen matrix: Novel mediators of fibrotic remodelling. J Mol Cell Cardiol 2016; 93:175-85. [PMID: 26578393 PMCID: PMC4945757 DOI: 10.1016/j.yjmcc.2015.11.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 01/05/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide and there is a pressing need for new therapeutic strategies to treat such conditions. The risk of developing cardiovascular disease increases dramatically with age, yet the majority of experimental research is executed using young animals. The cardiac extracellular matrix (ECM), consisting predominantly of fibrillar collagen, preserves myocardial integrity, provides a means of force transmission and supports myocyte geometry. Disruptions to the finely balanced control of collagen synthesis, post-synthetic deposition, post-translational modification and degradation may have detrimental effects on myocardial functionality. It is now well established that the aged heart is characterized by fibrotic remodelling, but the mechanisms responsible for this are incompletely understood. Furthermore, studies using aged animal models suggest that interstitial remodelling with disease may be age-dependent. Thus with the identification of new therapeutic strategies targeting fibrotic remodelling, it may be necessary to consider age-dependent mechanisms. In this review, we discuss remodelling of the cardiac collagen matrix as a function of age, whilst highlighting potential novel mediators of age-dependent fibrotic pathways.
Collapse
Affiliation(s)
- Margaux A Horn
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, 3.06 Core Technology Facility, 46 Grafton Street, Manchester M13 9NT, United Kingdom.
| | - Andrew W Trafford
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, 3.06 Core Technology Facility, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| |
Collapse
|
40
|
Franssen C, González Miqueo A. The role of titin and extracellular matrix remodelling in heart failure with preserved ejection fraction. Neth Heart J 2016; 24:259-67. [PMID: 26886920 PMCID: PMC4796057 DOI: 10.1007/s12471-016-0812-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterised by a high incidence of metabolic comorbidities that share the potential to induce both systemic and coronary microvascular inflammation and oxidative stress. These pathophysiological alterations contribute to increased passive stiffness of the myocardium and to diastolic dysfunction, both hallmarks of HFpEF. Passive myocardial stiffness depends mainly on two components: the extracellular matrix (ECM) and the cardiomyocytes. Quantitative and qualitative changes in collagen metabolism leading to myocardial fibrosis determine the ECM-based stiffness of the myocardium. Different noninvasive diagnostic tools to assess myocardial fibrosis are being developed, some of which have demonstrated to correlate with clinical status and prognosis. Cardiomyocytes mainly alter the passive stiffness through alterations in the giant myofilament titin, which serves as a spring. By modifying its phosphorylation state or by direct oxidative effects, titin determines cardiomyocyte-based passive stiffness. Probably the relative importance of cardiomyocyte-based changes is more important in the beginning of the disease, whereas ECM-based changes become more prominent in the more advanced stages. The present review focuses on these changes in ECM and cardiomyocytes in HFpEF and their potential prognostic and therapeutic implications.
Collapse
Affiliation(s)
- C Franssen
- ICaR-VU, VU University Medical Center, Van der Boechorststraat 7, 1081, BT Amsterdam, The Netherlands.
| | - A González Miqueo
- Center for Applied Medical Research, University of Navarra, Program of Cardiovascular Diseases, Pamplona, Spain
| |
Collapse
|
41
|
Abstract
Biomarkers are widely used and studied in heart failure. Most studies have described the utility and performance of biomarkers in sub-studies of randomised clinical trials, where the vast majority of the patients suffered from heart failure with reduced ejection fraction (HFrEF), and not with preserved ejection fraction (HFpEF). As a result, there is a scarcity of data describing the levels, dynamics, clinical and biochemical correlates, and biology of biomarkers in patients suffering from HFpEF, whereas HFpEF is in fact a very frequent clinical entity. This article discusses the value of different biomarkers in HFpEF. We describe various aspects of natriuretic peptide measurements in HFpEF patients, with a focus on diagnosis, prognosis and the risk prediction of developing heart failure. Further, we will discuss several emerging biomarkers such as galectin-3 and suppression of tumorigenicity 2, and recently discovered ones such as growth differentiation factor-15 and syndecan-1.
Collapse
|
42
|
Harvey A, Montezano AC, Lopes RA, Rios F, Touyz RM. Vascular Fibrosis in Aging and Hypertension: Molecular Mechanisms and Clinical Implications. Can J Cardiol 2016; 32:659-68. [PMID: 27118293 PMCID: PMC4906153 DOI: 10.1016/j.cjca.2016.02.070] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 02/08/2023] Open
Abstract
Aging is the primary risk factor underlying hypertension and incident cardiovascular disease. With aging, the vasculature undergoes structural and functional changes characterized by endothelial dysfunction, wall thickening, reduced distensibility, and arterial stiffening. Vascular stiffness results from fibrosis and extracellular matrix (ECM) remodelling, processes that are associated with aging and are amplified by hypertension. Some recently characterized molecular mechanisms underlying these processes include increased expression and activation of matrix metalloproteinases, activation of transforming growth factor-β1/SMAD signalling, upregulation of galectin-3, and activation of proinflammatory and profibrotic signalling pathways. These events can be induced by vasoactive agents, such as angiotensin II, endothelin-1, and aldosterone, which are increased in the vasculature during aging and hypertension. Complex interplay between the “aging process” and prohypertensive factors results in accelerated vascular remodelling and fibrosis and increased arterial stiffness, which is typically observed in hypertension. Because the vascular phenotype in a young hypertensive individual resembles that of an elderly otherwise healthy individual, the notion of “early” or “premature” vascular aging is now often used to describe hypertension-associated vascular disease. We review the vascular phenotype in aging and hypertension, focusing on arterial stiffness and vascular remodelling. We also highlight the clinical implications of these processes and discuss some novel molecular mechanisms of fibrosis and ECM reorganization.
Collapse
Affiliation(s)
- Adam Harvey
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Rheure Alves Lopes
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Francisco Rios
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland.
| |
Collapse
|
43
|
Tang WHW, Grodin JL. Novel Biomarkers of Heart Failure: Do They Have Incremental Clinical Utility? J Card Fail 2016; 22:263-4. [PMID: 26923642 DOI: 10.1016/j.cardfail.2016.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
Affiliation(s)
- W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio.
| | - Justin L Grodin
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
44
|
Ter Maaten JM, Damman K, Verhaar MC, Paulus WJ, Duncker DJ, Cheng C, van Heerebeek L, Hillege HL, Lam CSP, Navis G, Voors AA. Connecting heart failure with preserved ejection fraction and renal dysfunction: the role of endothelial dysfunction and inflammation. Eur J Heart Fail 2016; 18:588-98. [PMID: 26861140 DOI: 10.1002/ejhf.497] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/09/2016] [Accepted: 01/12/2015] [Indexed: 12/17/2022] Open
Abstract
Renal dysfunction in heart failure with preserved ejection fraction (HFpEF) is common and is associated with increased mortality. Impaired renal function is also a risk factor for developing HFpEF. A new paradigm for HFpEF, proposing a sequence of events leading to myocardial remodelling and dysfunction in HFpEF, was recently introduced, involving inflammatory, microvascular, and cardiac components. The kidney might play a key role in this systemic process. Renal impairment causes metabolic and systemic derangements in circulating factors, causing an activated systemic inflammatory state and endothelial dysfunction, which may lead to cardiomyocyte stiffening, hypertrophy, and interstitial fibrosis via cross-talk between the endothelium and cardiomyocyte compartments. Here, we review the role of endothelial dysfunction and inflammation to explain the link between renal dysfunction and HFpEF, which allows for identification of new early risk markers, prognostic factors, and unique targets for intervention.
Collapse
Affiliation(s)
- Jozine M Ter Maaten
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Kevin Damman
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marianne C Verhaar
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands
| | - Walter J Paulus
- Department of Physiology, Cardiology, Pathology, and Surgery, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Dirk J Duncker
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Caroline Cheng
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands.,Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Loek van Heerebeek
- Department of Physiology, Cardiology, Pathology, and Surgery, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Hans L Hillege
- University of Groningen, Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolyn S P Lam
- National Heart Centre Singapore and, Duke-National University of Singapore Graduate School Medicine, Singapore
| | - Gerjan Navis
- University of Groningen, Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriaan A Voors
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Samson R, Jaiswal A, Ennezat PV, Cassidy M, Le Jemtel TH. Clinical Phenotypes in Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2016; 5:e002477. [PMID: 26811159 PMCID: PMC4859363 DOI: 10.1161/jaha.115.002477] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Rohan Samson
- Tulane University Heart and Vascular InstituteTulane University School of MedicineNew OrleansLA
| | - Abhishek Jaiswal
- Tulane University Heart and Vascular InstituteTulane University School of MedicineNew OrleansLA
| | - Pierre V. Ennezat
- Department of CardiologyCentre Hospitalier Universitaire de GrenobleGrenoble Cedex 09France
| | - Mark Cassidy
- Tulane University Heart and Vascular InstituteTulane University School of MedicineNew OrleansLA
| | - Thierry H. Le Jemtel
- Tulane University Heart and Vascular InstituteTulane University School of MedicineNew OrleansLA
| |
Collapse
|
46
|
D'Elia E, Vaduganathan M, Gori M, Gavazzi A, Butler J, Senni M. Role of biomarkers in cardiac structure phenotyping in heart failure with preserved ejection fraction: critical appraisal and practical use. Eur J Heart Fail 2015; 17:1231-9. [PMID: 26493383 DOI: 10.1002/ejhf.430] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 09/28/2015] [Accepted: 09/28/2015] [Indexed: 12/28/2022] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a heterogeneous clinical syndrome characterized by cardiovascular, metabolic, and pro-inflammatory diseases associated with advanced age and extracardiac comorbidities. All of these conditions finally lead to impairment of myocardial structure and function. The large phenotypic heterogeneity of HFpEF from pathophysiological underpinnings presents a major hurdle to HFpEF therapy. The new therapeutic approach in HFpEF should be targeted to each HF phenotype, instead of the 'one-size-fits-all' approach, which has not been successful in clinical trials. Unless the structural and biological determinants of the failing heart are deeply understood, it will be impossible to appropriately differentiate HFpEF patients, identify subtle myocardial abnormalities, and finally reverse abnormal cardiac function. Based on evidence from endomyocardial biopsies, some of the specific cardiac structural phenotypes to be targeted in HFpEF may be represented by myocyte hypertrophy, interstitial fibrosis, myocardial inflammation associated with oxidative stress, and coronary disease. Once the diagnosis of HFpEF has been established, a potential approach could be to use a panel of biomarkers to identify the main cardiac structural HFpEF phenotypes, guiding towards more appropriate therapeutic strategies. Accordingly, the purpose of this review is to investigate the potential role of biomarkers in identifying different cardiac structural HFpEF phenotypes and to discuss the merits of a biomarker-guided strategy in HFpEF.
Collapse
Affiliation(s)
- Emilia D'Elia
- Dipartimento Cardiovascolare, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Muthiah Vaduganathan
- Brigham and Women's Hospital Heart and Vascular Center and Harvard Medical School, Boston, MA, USA
| | - Mauro Gori
- Dipartimento Cardiovascolare, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Antonello Gavazzi
- FROM Fondazione per la Ricerca, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Javed Butler
- Cardiology Division, Stony Brook University, Stony Brook, NY, USA
| | - Michele Senni
- Dipartimento Cardiovascolare, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
47
|
Alehagen U, Benson L, Edner M, Dahlström U, Lund LH. Association Between Use of Statins and Mortality in Patients With Heart Failure and Ejection Fraction of ≥50%. Circ Heart Fail 2015; 8:862-70. [DOI: 10.1161/circheartfailure.115.002143] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 07/29/2015] [Indexed: 11/16/2022]
Abstract
Background—
The pathophysiology of heart failure with preserved ejection fraction is poorly understood, but may involve a systemic proinflammatory state. Therefore, statins might improve outcomes in patients with heart failure with preserved ejection fraction defined as ≥50%.
Methods and Results—
Of 46 959 unique patients in the prospective Swedish Heart Failure Registry, 9140 patients had heart failure and ejection fraction ≥50% (age 77±11 years, 54.0% women), and of these, 3427 (37.5%) were treated with statins. Propensity scores for statin treatment were derived from 40 baseline variables. The association between statin use and primary (all-cause mortality) and secondary (separately, cardiovascular mortality, and combined all-cause mortality or cardiovascular hospitalization) end points was assessed with Cox regressions in a population matched 1:1 based on age and propensity score. In the matched population, 1-year survival was 85.1% for statin-treated versus 80.9% for untreated patients (hazard ratio, 0.80; 95% confidence interval, 0.72–0.89;
P
<0.001). Statins were also associated with reduced cardiovascular death (hazard ratio, 0.86; 95% confidence interval, 0.75–0.98;
P
=0.026) and composite all-cause mortality or cardiovascular hospitalization (hazard ratio, 0.89; 95% confidence interval, 0.82–0.96;
P
=0.003).
Conclusions—
In heart failure with ejection fraction ≥50%, the use of statins was associated with improved outcomes. The mechanisms should be evaluated and the effects tested in a randomized trial.
Collapse
Affiliation(s)
- Urban Alehagen
- From the Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden (U.A., U.D.); Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden (L.B.); and Department of Cardiology, Karolinska University Hospital and Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden (M.E., L.H.L.)
| | - Lina Benson
- From the Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden (U.A., U.D.); Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden (L.B.); and Department of Cardiology, Karolinska University Hospital and Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden (M.E., L.H.L.)
| | - Magnus Edner
- From the Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden (U.A., U.D.); Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden (L.B.); and Department of Cardiology, Karolinska University Hospital and Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden (M.E., L.H.L.)
| | - Ulf Dahlström
- From the Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden (U.A., U.D.); Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden (L.B.); and Department of Cardiology, Karolinska University Hospital and Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden (M.E., L.H.L.)
| | - Lars H. Lund
- From the Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden (U.A., U.D.); Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden (L.B.); and Department of Cardiology, Karolinska University Hospital and Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden (M.E., L.H.L.)
| |
Collapse
|
48
|
van Empel V, Brunner-La Rocca HP. Inflammation in HFpEF: Key or circumstantial? Int J Cardiol 2015; 189:259-63. [DOI: 10.1016/j.ijcard.2015.04.110] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/04/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022]
|