101
|
Manilall A, Mokotedi L, Gunter S, Le Roux R, Fourie S, Flanagan CA, Millen AME. Increased protein phosphatase 5 expression in inflammation-induced left ventricular dysfunction in rats. BMC Cardiovasc Disord 2022; 22:539. [PMID: 36494772 PMCID: PMC9732989 DOI: 10.1186/s12872-022-02977-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Titin phosphorylation contributes to left ventricular (LV) diastolic dysfunction. The independent effects of inflammation on the molecular pathways that regulate titin phosphorylation are unclear. METHODS We investigated the effects of collagen-induced inflammation and subsequent tumor necrosis factor-α (TNF-α) inhibition on mRNA expression of genes involved in regulating titin phosphorylation in 70 Sprague-Dawley rats. LV diastolic function was assessed with echocardiography. Circulating inflammatory markers were quantified by enzyme-linked immunosorbent assay and relative LV gene expression was assessed by Taqman® polymerase chain reaction. Differences in normally distributed variables between the groups were determined by two-way analysis of variance (ANOVA), followed by Tukey post-hoc tests. For non-normally distributed variables, group differences were determined by Kruskal-Wallis tests. RESULTS Collagen inoculation increased LV relative mRNA expression of vascular cell adhesion molecule 1 (VCAM1), pentraxin 3 (PTX3), and inducible nitric oxide synthase (iNOS) compared to controls, indicating local microvascular inflammation. Collagen inoculation decreased soluble guanylate cyclase alpha-2 (sGCα2) and soluble guanylate cyclase beta-2 (sGCβ2) expression, suggesting downregulation of nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling. Inhibiting TNF-α prevented collagen-induced changes in VCAM1, iNOS, sGCα2 and sGCβ2 expression. Collagen inoculation increased protein phosphatase 5 (PP5) expression. Like LV diastolic dysfunction, increased PP5 expression was not prevented by TNF-α inhibition. CONCLUSION Inflammation-induced LV diastolic dysfunction may be mediated by a TNF-α-independent increase in PP5 expression and dephosphorylation of the N2-Bus stretch element of titin, rather than by TNF-α-induced downregulation of NO-sGC-cGMP pathway-dependent titin phosphorylation. The steady rise in number of patients with inflammation-induced diastolic dysfunction, coupled with low success rates of current therapies warrants a better understanding of the systemic signals and molecular pathways responsible for decreased titin phosphorylation in development of LV diastolic dysfunction. The therapeutic potential of inhibiting PP5 upregulation in LV diastolic dysfunction requires investigation.
Collapse
Affiliation(s)
- Ashmeetha Manilall
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Lebogang Mokotedi
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Sulè Gunter
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Regina Le Roux
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Serena Fourie
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Colleen A. Flanagan
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Aletta M. E. Millen
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| |
Collapse
|
102
|
Edwards JJ, O’Driscoll JM. Exercise Training in Heart failure with Preserved and Reduced Ejection Fraction: A Systematic Review and Meta-Analysis. SPORTS MEDICINE - OPEN 2022; 8:76. [PMID: 35674912 PMCID: PMC9177931 DOI: 10.1186/s40798-022-00464-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/13/2022] [Indexed: 11/10/2022]
Abstract
Background While exercise training (ET) is an established tool in heart failure (HF), no research to date has analysed the efficacy of ET in both preserved (HFpEF) and reduced (HFrEF) ejection fraction phenotypes across the same clinically important parameters. Methods A comprehensive systematic search was performed to identify trials published between 1990 and May 2021. Controlled trials of adults reporting pre- and post-ET peak VO2, 6-min walk distance (6MWD), Minnesota Living with Heart Failure Questionnaire (MLHFQ), Kansas City Cardiomyopathy Questionnaire (KCCQ) and left ventricular ejection fraction (LVEF) were considered. Parameters of cardiac diastolic function, brain natriuretic peptides (BNP)/N-terminal prohormone of BNP (NTproBNP) and follow-up hospitalisation and mortality data were also analysed. Results Ninety-three studies (11 HFpEF and 82 HFrEF) were included in the final analysis, with a pooled sample size of 11,081 participants. HFpEF analysis demonstrated significant improvements in peak VO2 (weighted mean difference: 2.333 ml·min-1·kg-1, Pfixed < 0.001), 6MWD (WMD: 35.396 m, Pfixed < 0.001), MLHFQ (WMD: − 10.932, Prandom < 0.001), KCCQ (WMD: 3.709, Pfixed = 0.037) and E/e′ (WMD: − 1.709, [95% CI] = − 2.91–0.51, Prandom = 0.005). HFrEF analysis demonstrated significant improvements in peak VO2 (WMD: 3.050 ml·min-1·kg-1, Prandom < 0.001), 6MWD (WMD: 37.299 m, Prandom < 0.001), MLHFQ (WMD: − 10.932, Prandom < 0.001), LVEF (WMD: 2.677%, Prandom = 0.002) and BNP/NTproBNP (SMD: − 1.349, Prandom < 0.001). Outcome analysis was only performed in HFrEF, which found no significant changes in hospitalisation, all-cause mortality or composite end-points. Conclusion ET significantly improves exercise capacity and quality of life in both HFpEF and HFrEF patients. In HFpEF patients, ET significantly improved an important index of diastolic function, with significant improvements in LVEF and NTproBNP/BNP seen in HFrEF patients only. Such benefits did not translate into significantly reduced hospitalisation or mortality after short-term follow-up. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40798-022-00464-5.
Collapse
|
103
|
Frampton JE. Empagliflozin: A Review in Symptomatic Chronic Heart Failure. Drugs 2022; 82:1591-1602. [DOI: 10.1007/s40265-022-01778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/16/2022]
|
104
|
Zhbanov KA, Salakheeva EY, Sokolova IY, Zheleznykh EA, Zektser VY, Privalova EV, Belenkov YN, Shchendrygina AA. Neuregulin-1β, Biomarkers of Inflammation and Myocardial Fibrosis in Heart Failure Patients. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-09-05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- K. A. Zhbanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. Yu. Salakheeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. Ya. Sokolova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. A. Zheleznykh
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - V. Yu. Zektser
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. V. Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - Yu. N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | |
Collapse
|
105
|
Epidemiology, Diagnosis, Pathophysiology, and Initial Approach to Heart Failure with Preserved Ejection Fraction. Cardiol Clin 2022; 40:397-413. [DOI: 10.1016/j.ccl.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
106
|
Eidizadeh A, Schnelle M, Leha A, Edelmann F, Nolte K, Werhahn SM, Binder L, Wachter R. Biomarker profiles in heart failure with preserved vs. reduced ejection fraction: results from the DIAST-CHF study. ESC Heart Fail 2022; 10:200-210. [PMID: 36184749 PMCID: PMC9871664 DOI: 10.1002/ehf2.14167] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/29/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
AIMS Chronic heart failure (HF) is a common disease and one of the leading causes of death worldwide. Heart failure with preserved ejection fraction (HFpEF) and with reduced ejection fraction (HFrEF) are different diseases with distinct as well as comparable pathophysiologies and diverse responses to therapeutic agents. We aimed to identify possible pathobiochemical signalling pathways and biomarkers in HFpEF and HFrEF by using a broad proteomic approach. METHODS AND RESULTS A total of 180 biomarkers in the plasma of a representative subgroup (71 years old) of HFpEF (70% female) with a left ventricular ejection fraction (LVEF) ≥ 50% and HFrEF (18% female) with an LVEF ≤ 40% patients (n = 127) from the Prevalence and Clinical Course of Diastolic Dysfunction and Diastolic Heart Failure (DIAST-CHF) trial were examined and compared with a healthy control group (n = 40; 48% female). We were able to identify 35 proteins that were expressed significantly different in both HF groups compared with the control group. We determine 29 unique proteins expressed in HFpEF and 33 unique proteins in HFrEF. Significantly up-regulated trefoil factor 3 (TFF3) and down-regulated contactin-1 could be identified as previously unknown biomarkers for HF. However, TFF3 is also a predictive factor for the occurrence of a cardiovascular event in HFpEF patients. In HFpEF, serine protease 27 was found at reduced levels for the first time, which could offer a new therapeutic target. Additionally, network analyses showed a special role of platelet-derived growth factor subunit A, Dickkopf-related protein 1, and tumour necrosis factor receptor superfamily member 6 in HFpEF patients, whereas perlecan and junctional adhesion molecule A stood out in the HFrEF group. Overall, signalling pathways of metabolic processes, cellular stress, and iron metabolism seemed to be important for HFrEF, whereas for HFpEF, oxygen stress, haemostasis, cell renewal, cell migration, and cell proliferation are in the foreground. CONCLUSIONS The identified proteins and signalling pathways offer new therapeutic and diagnostic approaches for patients with chronic HF.
Collapse
Affiliation(s)
- Abass Eidizadeh
- Institute for Clinical Chemistry/Interdisciplinary UMG LaboratoryUniversity Medical Center GöttingenGöttingenGermany
| | - Moritz Schnelle
- Institute for Clinical Chemistry/Interdisciplinary UMG LaboratoryUniversity Medical Center GöttingenGöttingenGermany,DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Andreas Leha
- DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany,Department of Medical StatisticsUniversity Medical Center GöttingenGöttingenGermany
| | - Frank Edelmann
- Department of Internal Medicine and CardiologyCharité‐Universitätsmedizin Berlin, Campus Virchow KlinikumBerlinGermany,DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany,Berlin Institute of HealthBerlinGermany
| | - Kathleen Nolte
- Clinic of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | | | - Lutz Binder
- Institute for Clinical Chemistry/Interdisciplinary UMG LaboratoryUniversity Medical Center GöttingenGöttingenGermany,DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Rolf Wachter
- DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany,Clinic of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany,Clinic and Policlinic for CardiologyUniversity Hospital LeipzigLeipzigGermany
| |
Collapse
|
107
|
Zhan Q, Peng W, Wang S, Gao J. Heart Failure with Preserved Ejection Fraction: Pathogenesis, Diagnosis, Exercise, and Medical Therapies. J Cardiovasc Transl Res 2022; 16:310-326. [PMID: 36171526 DOI: 10.1007/s12265-022-10324-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for more than one-half of total heart failure cases, with a high prevalence and poor prognosis, especially in older and female patients. Patients with HFpEF are characterized by hypertension, left ventricular hypertrophy, and diastolic dysfunction, and the main symptoms are dyspnea and exercise intolerance. HFpEF is currently poorly studied, and pharmacological treatment for HFpEF is still underexplored. Accumulating clinical trials have shown that exercise could exert benefits on diastolic dysfunction and quality of life in patients with HFpEF. However, there is a high limitation for applying exercise therapy due to exercise intolerance in patients with HFpEF. Key effectors of exercise-protection could be novel therapeutic targets for developing drugs to prevent and treat HFpEF. In this review article, we provide an overview of the pathogenic factors, diagnostic methods, research animal models, the mechanisms of exercise-mediated cardiac protection, and current treatments for HFpEF.
Collapse
Affiliation(s)
- Qingyi Zhan
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Wenjing Peng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Siqi Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Juan Gao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China. .,Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
108
|
Alhajri N, Rustom M, Adegbile A, Ahmed W, Kilidar S, Afify N. Deciphering the Basis of Molecular Biology of Selected Cardiovascular Diseases: A View on Network Medicine. Int J Mol Sci 2022; 23:ijms231911421. [PMID: 36232723 PMCID: PMC9569471 DOI: 10.3390/ijms231911421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death across the world. For decades, researchers have been studying the causes of cardiovascular disease, yet many of them remain undiscovered or poorly understood. Network medicine is a recently expanding, integrative field that attempts to elucidate this issue by conceiving of disease as the result of disruptive links between multiple interconnected biological components. Still in its nascent stages, this revolutionary application of network science facilitated a number of important discoveries in complex disease mechanisms. As methodologies become more advanced, network medicine harbors the potential to expound on the molecular and genetic complexities of disease to differentiate how these intricacies govern disease manifestations, prognosis, and therapy. This is of paramount importance for confronting the incredible challenges of current and future cardiovascular disease research. In this review, we summarize the principal molecular and genetic mechanisms of common cardiac pathophysiologies as well as discuss the existing knowledge on therapeutic strategies to prevent, halt, or reverse these pathologies.
Collapse
Affiliation(s)
- Noora Alhajri
- Department of Internal Medicine, Cleveland Clinic Abu Dhabi (CCAD), Abu Dhabi P.O. Box 112412, United Arab Emirates
- Correspondence:
| | - Mohammad Rustom
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Adedayo Adegbile
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Weshah Ahmed
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Salsabeel Kilidar
- Department of Emergency Medicine, Sheikh Shakhbout Medical City SSMC, Abu Dhabi P.O. Box 11001, United Arab Emirates
| | - Nariman Afify
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
109
|
Iovanovici DC, Bungau SG, Vesa CM, Moisi M, Babes EE, Tit DM, Horvath T, Behl T, Rus M. Reviewing the Modern Therapeutical Options and the Outcomes of Sacubitril/Valsartan in Heart Failure. Int J Mol Sci 2022; 23:11336. [PMID: 36232632 PMCID: PMC9570001 DOI: 10.3390/ijms231911336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Sacubitril/valsartan (S/V) is a pharmaceutical strategy that increases natriuretic peptide levels by inhibiting neprilysin and regulating the renin-angiotensin-aldosterone pathway, blocking AT1 receptors. The data for this innovative medication are mainly based on the PARADIGM-HF study, which included heart failure with reduced ejection fraction (HFrEF)-diagnosed patients and indicated a major improvement in morbidity and mortality when S/V is administrated compared to enalapril. A large part of the observed favorable results is related to significant reverse cardiac remodeling confirmed in two prospective trials, PROVE-HF and EVALUATE-HF. Furthermore, according to a subgroup analysis from the PARAGON-HF research, S/V shows benefits in HFrEF and in many subjects having preserved ejection fraction (HFpEF), which indicated a decrease in HF hospitalizations among those with a left ventricular ejection fraction (LVEF) < 57%. This review examines the proven benefits of S/V and highlights continuing research in treating individuals with varied HF characteristics. The article analyses published data regarding both the safeness and efficacy of S/V in patients with HF, including decreases in mortality and hospitalization, increased quality of life, and reversible heart remodeling. These benefits led to the HF guidelines recommendations updating and inclusion of S/V combinations a key component of HFrEF treatment.
Collapse
Affiliation(s)
- Diana-Carina Iovanovici
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Simona Gabriela Bungau
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Madalina Moisi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Elena Emilia Babes
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Delia Mirela Tit
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Tunde Horvath
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Tapan Behl
- School of Health Sciences &Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun 248007, India
| | - Marius Rus
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
110
|
Kim TE, Kim H, Sung J, Kim DK, Lee MS, Han SW, Kim HJ, Kim SH, Ryu KH. The association between metabolic syndrome and heart failure in middle-aged male and female: Korean population-based study of 2 million individuals. Epidemiol Health 2022; 44:e2022078. [PMID: 36177980 PMCID: PMC10089711 DOI: 10.4178/epih.e2022078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Although an association is known to exist between metabolic syndrome (MetS) and heart failure (HF) risk, large longitudinal studies are limited. We investigated metabolic status as a risk factor for HF in middle-aged male and female and considered sex differences in various risk factors for HF using nationwide real-world data. METHODS Data obtained from the Korean National Health Insurance Service from 2009 to 2016 were analyzed. A total of 2,151,597 middle-aged subjects (between 50 and 59 years old) were enrolled. Subjects were divided into 3 groups (normal, pre- MetS, and MetS). Cox proportional hazard models were used to estimate the association between MetS and incident HF after adjusting for clinical risk factors. RESULTS At baseline, MetS existed in 23.77% of male and 10.58% of female. Pre-MetS and MetS increased the risk of HF: the hazard ratios of pre-MetS for incident HF were 1.508 (95% confidence interval [CI], 1.287 to 1.767) in male and 1.395 (95% CI, 1.158 to 1.681) in female, and those of MetS were 1.711 (95% CI, 1.433 to 2.044) in male and 2.144 (95% CI, 1.674 to 2.747) in female. Current smoking, a low hemoglobin level, underweight (body mass index < 18.5 kg/m2), a high creatinine level, and acute myocardial infarction were also predictors of HF in both sexes. CONCLUSIONS Pre-MetS and MetS were identified as risk factors for HF in middle-aged male and female. The effect of MetS on the occurrence of HF was stronger in female than in male. Pre-MetS was also a predictor of HF, but was associated with a lower risk than MetS.
Collapse
Affiliation(s)
- Tae-Eun Kim
- Department of Clinical Pharmacology, Konkuk University Medical Center, Seoul, Korea
| | - Hyeongsu Kim
- Department of Preventive Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - JiDong Sung
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk-Kyung Kim
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myoung-Soon Lee
- Department of Social and Preventive Medicine, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Seong Woo Han
- Division of Cardiology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea
| | - Hyun-Joong Kim
- Division of Cardiology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Sung Hea Kim
- Division of Cardiology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Kyu-Hyung Ryu
- Division of Cardiology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea
| |
Collapse
|
111
|
Walther C, Wenzel J, Schnabel RB, Heydecke G, Seedorf U, Beikler T, Borof K, Nikorowitsch J, Schrage B, Blankenberg S, Twerenbold R, Zeller T, Magnussen C, Aarabi G. Association between periodontitis and heart failure in the general population. ESC Heart Fail 2022; 9:4189-4197. [PMID: 36101477 PMCID: PMC9773719 DOI: 10.1002/ehf2.14150] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/12/2022] [Accepted: 09/04/2022] [Indexed: 01/19/2023] Open
Abstract
AIMS Data on the association between periodontitis and preclinical cardiac alterations remain scarce. The aim of the current study is to determine if periodontitis is associated with morphological and functional cardiac changes measured by transthoracic echocardiography as well as different heart failure (HF) phenotypes. METHODS Participants from the population-based Hamburg City Health Study [ClinicalTrial.gov (NCT03934957)], who underwent transthoracic echocardiography and periodontal screening were included. Periodontitis was classified according to Eke and Page (none/mild, moderate, severe). The 2021 ESC HF guidelines were applied and HF was classified into HF with preserved ejection fraction (HFpEF, ejection fraction ≥50%), HF with mid-range and reduced ejection fraction [HF(m)rEF, ejection fraction <50%], and HF in general [HFpEF and HF(m)rEF]. Due to limited size, all subjects with LVEF <50% and symptoms or signs of HF were classified as HF with reduced and mildly reduced ejection fraction [HF(m)rEF]. RESULTS Within 6209 participants with full periodontal examination, we identified an overlap of n = 167 participants with periodontitis and HF. Participants with severe periodontitis showed a higher burden of cardiovascular risk factors (men at advanced age, diabetes mellitus, hypertension) when compared with participants with none/mild periodontitis. After adjustment for age, sex, body mass index, smoking, diabetes, hypertension, atrial fibrillation, and coronary artery disease, severe periodontitis was significantly associated with HF(m)rEF (odds ratio: 3.16; 95% CI: 1.21, 8.22; P = 0.019), although no association was found for HFpEF and HF in general. CONCLUSIONS The current study demonstrated that severe periodontitis was significantly associated with HF(m)rEF, although no relevant associations were found with HFpEF and HF in general as well as echocardiographic variables. The results implicate a potential target group, who need special attention from cooperating physicians and dentists. Future studies are warranted to verify whether systemic inflammation could be the link between the two diseases.
Collapse
Affiliation(s)
- Carolin Walther
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jan‐Per Wenzel
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Epidemiological Study CenterUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany
| | - Renate B. Schnabel
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Guido Heydecke
- Department of Prosthetic Dentistry, Center for Dental and Oral MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Udo Seedorf
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Katrin Borof
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany
| | - Julius Nikorowitsch
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Epidemiological Study CenterUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany
| | - Benedikt Schrage
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Stefan Blankenberg
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Raphael Twerenbold
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Epidemiological Study CenterUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Kiel/LuebeckHamburgGermany,University Center of Cardiovascular ScienceUniversity Heart and Vascular CenterHamburgGermany
| | - Tanja Zeller
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,University Center of Cardiovascular ScienceUniversity Heart and Vascular CenterHamburgGermany
| | - Christina Magnussen
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, UKE HamburgHamburgGermany,Population Health Research DepartmentUniversity Heart and Vascular Center, UKE HamburgHamburgGermany,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Ghazal Aarabi
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
112
|
Bracun V, van Essen B, Voors AA, van Veldhuisen DJ, Dickstein K, Zannad F, Metra M, Anker S, Samani NJ, Ponikowski P, Filippatos G, Cleland JG, Lang CC, Ng LL, Shi C, de Wit S, Aboumsallem JP, Meijers WC, Klip IJT, van der Meer P, de Boer RA. Insulin-like growth factor binding protein 7 (IGFBP7), a link between heart failure and senescence. ESC Heart Fail 2022; 9:4167-4176. [PMID: 36088651 PMCID: PMC9773704 DOI: 10.1002/ehf2.14120] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023] Open
Abstract
AIMS Insulin like growth factor binding protein 7 (IGFBP7) is a marker of senescence secretome and a novel biomarker in patients with heart failure (HF). We evaluated the prognostic value of IGFBP7 in patients with heart failure and examined associations to uncover potential new pathophysiological pathways related to increased plasma IGFBP7 concentrations. METHODS AND RESULTS We have measured plasma IGFBP7 concentrations in 2250 subjects with new-onset or worsening heart failure (BIOSTAT-CHF cohort). Higher IGFBP7 plasma concentrations were found in older subjects, those with worse kidney function, history of atrial fibrillation, and diabetes mellitus type 2, and in subjects with higher number of HF hospitalizations. Higher IGFBP7 levels also correlate with the levels of several circulating biomarkers, including higher NT-proBNP, hsTnT, and urea levels. Cox regression analyses showed that higher plasma IGFBP7 concentrations were strongly associated with increased risk of all three main endpoints (hospitalization, all-cause mortality, and combined hospitalization and mortality) (HR 1.75, 95% CI 1.25-2.46; HR 1.71, 95% CI 1.39-2.11; and HR 1.44, 95% CI 1.23-1.70, respectively). IGFBP7 remained a significant predictor of these endpoints in patients with both reduced and preserved ejection fraction. Likelihood ratio test showed significant improvement of all three risk prediction models, after adding IGFBP7 (P < 0.001). A biomarker network analysis showed that IGFBP7 levels activate different pathways involved in the regulation of the immune system. Results were externally validated in BIOSTAT-CHF validation cohort. CONCLUSIONS IGFPB7 presents as an independent and robust prognostic biomarker in patients with HF, with both reduced and preserved ejection fraction. We validate the previously published data showing IGFBP7 has correlations with a number of echocardiographic markers. Lastly, IGFBP7 pathways are involved in different stages of immune system regulation, linking heart failure to senescence pathways.
Collapse
Affiliation(s)
- Valentina Bracun
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Bart van Essen
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Adriaan A. Voors
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | | | | | - Faiez Zannad
- Universite de Lorraine | InsermCentre d'Investigations CliniquesNancyFrance
| | - Marco Metra
- Department of Medical and Surgical Specialties | Radiological Sciences and Public Health | Institute of CardiologyUniversity of BresciaBresciaItaly
| | - Stefan Anker
- Department of Cardiology (CVK) and Berlin Institute of Health Center for Regenerative Therapies (BCRT) | German Centre for Cardiovascular Research (DZHK) partner site BerlinCharité UniversitätsmedizinBerlinGermany
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences | University of Leicester | Glenfield Hospital | and NIHR Leicester Biomedical Research CentreGlenfield HospitalLeicesterUnited Kingdom
| | - Piotr Ponikowski
- Department of Heart DiseasesWroclaw Medical UniversityWrocławPoland
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens | School of MedicineAttikon University HospitalAthensGreece
| | - John G.F. Cleland
- Robertson Centre for Biostatistics | Institute of Health and WellbeingUniversity of Glasgow | Imperial CollegeLondonUnited Kingdom
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine | Medical Research Institute | Ninewells Hospital & Medical SchoolUniversity of DundeeDundeeUnited Kingdom
| | - Leong L. Ng
- Department of Cardiovascular Sciences | University of Leicester | Glenfield Hospital | and NIHR Leicester Biomedical Research CentreGlenfield HospitalLeicesterUnited Kingdom
| | - Canxia Shi
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Sanne de Wit
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | | | - Wouter C. Meijers
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - IJsbrand T. Klip
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Peter van der Meer
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Rudolf A. de Boer
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
113
|
Abstract
PURPOSE OF REVIEW Hypertension (HTN) remains the most common and strongest contributing factor to the development of heart failure with preserved ejection fraction (HFpEF). In this review, we aim to summarize the pathophysiological processes linking HTN to HFpEF and highlight novel concepts in medical and device-based management of HFpEF and HTN. RECENT FINDINGS Despite the global increase in the prevalence of HFpEF, there has been limited benefit in current medication and device-based therapy for this complex syndrome. The hallmark of HFpEF is an elevated left intra-atrial and ventricular pressure and exertional dyspnea. Traditional medications used for treating HTN in patients with reduced left ventricular ejection fraction have unclear benefits in patients with HFpEF. Careful analysis of emerging medications such as angiotensin receptor-neprilysin inhibitor and sodium-glucose co-transporter-2 inhibitors showed benefit in reducing not only blood pressure but also hospitalizations in patients with HFpEF. Current data on device-based therapy aims to reduce left intra-atrial pressure, ventricular pressure and stimulate baroreceptors to lower blood pressure; however, needs further investigation. SUMMARY The nexus of HTN and HFpEF remains strong and complex. Although traditional medications for treating HFrEF did not affect long-term outcomes, novel therapies with angiotensin receptor neprilysin-inhibitor and sodium-glucose co-transporter-2 inhibitor offer promising results. Many device-based interventions in the HFpEF population are being developed with the aim to reduce left intra-atrial and ventricular pressure; however, their role in HFpEF hypertensive patients needs to be further investigated.
Collapse
Affiliation(s)
- Abdulaziz Joury
- John Ochsner Heart and Vascular Institute, Ochsner Medical Center, New Orleans, Louisiana, USA
- King Salman Heart Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tripti Gupta
- John Ochsner Heart and Vascular Institute, Ochsner Medical Center, New Orleans, Louisiana, USA
- The University of Queensland School of Medicine, Ochsner Clinical School
| | - Selim R Krim
- John Ochsner Heart and Vascular Institute, Ochsner Medical Center, New Orleans, Louisiana, USA
- The University of Queensland School of Medicine, Ochsner Clinical School
- Section of Cardiomyopathy & Heart Transplantation, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| |
Collapse
|
114
|
Mc Causland FR, Lefkowitz MP, Claggett B, Packer M, Senni M, Gori M, Jhund PS, McGrath MM, Rouleau JL, Shi V, Swedberg K, Vaduganathan M, Zannad F, Pfeffer MA, Zile M, McMurray JJV, Solomon SD. Angiotensin-neprilysin inhibition and renal outcomes across the spectrum of ejection fraction in heart failure. Eur J Heart Fail 2022; 24:1591-1598. [PMID: 34989105 PMCID: PMC9253196 DOI: 10.1002/ejhf.2421] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/01/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
AIMS Patients with heart failure are at higher risk of progression to end-stage renal disease (ESRD), regardless of ejection fraction (EF). We assessed the renal effects of angiotensin-neprilysin inhibition in a pooled analysis of 13 195 patients with heart failure with reduced and preserved EF. METHODS AND RESULTS We combined data from PARADIGM-HF (EF ≤40%; n = 8399) and PARAGON-HF (EF ≥45%; n = 4796) in a pre-specified pooled analysis. We assessed the effect of treatment (sacubitril/valsartan vs. enalapril or valsartan) on a composite of either ≥50% reduction in estimated glomerular filtration rate (eGFR), ESRD, or death from renal causes, in addition to changes in eGFR slope. We assessed whether baseline renal function or EF modified the effect of therapy on renal outcomes. At randomization, eGFR was 68 ± 20 ml/min/1.73 m2 in PARADIGM-HF and 63 ± 19 ml/min/1.73 m2 in PARAGON-HF. The composite renal outcome occurred in 70 of 6594 patients (1.1%) in the sacubitril/valsartan group and in 123 of 6601 patients (1.9%) in the valsartan or enalapril group (hazard ratio 0.56, 95% confidence interval [CI] 0.42-0.75; p < 0.001). The mean eGFR change was -1.8 (95% CI -1.9 to -1.7) ml/min/1.73 m2 /year for the sacubitril/valsartan group, compared with -2.4 (95% CI -2.5 to -2.2) ml/min/1.73 m2 /year for the valsartan or enalapril group. The treatment effect on the composite renal endpoint was not modified by categories of baseline eGFR (p-interaction = 0.64), but was most pronounced in those with baseline EF between 30% and 60% (p-interaction = 0.001). CONCLUSIONS In patients with heart failure, sacubitril/valsartan reduced the risk of serious adverse renal outcomes and slowed decline in eGFR, compared with valsartan or enalapril, independent of baseline renal function.
Collapse
Affiliation(s)
- Finnian R. Mc Causland
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Brian Claggett
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Michele Senni
- Cardiology Division, Cardiovascular Department, Azienda Ospedaliera Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mauro Gori
- Cardiology Division, Cardiovascular Department, Azienda Ospedaliera Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Pardeep S. Jhund
- British Heart Foundation Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Martina M. McGrath
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jean L. Rouleau
- Institut de Cardiologie de Montréal, Université de Montréal, Montreal, Canada
| | - Victor Shi
- Novartis Pharmaceuticals, East Hanover, NJ, USA
| | - Karl Swedberg
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden
| | - Muthiah Vaduganathan
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Faiez Zannad
- Université de Lorraine, Inserm CIC1433, CHRU de Nancy, France
| | - Marc A. Pfeffer
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Michael Zile
- Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, South Carolina, USA
| | - John J. V. McMurray
- Cardiology Division, Cardiovascular Department, Azienda Ospedaliera Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Scott D. Solomon
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
115
|
Habimana O, Modupe Salami O, Peng J, Yi GH. Therapeutic Implications of Targeting Pyroptosis in Cardiac-related Etiology of Heart Failure. Biochem Pharmacol 2022; 204:115235. [PMID: 36044938 DOI: 10.1016/j.bcp.2022.115235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
Abstract
Heart failure remains a considerable clinical and public health problem, it is the dominant cause of death from cardiovascular diseases, besides, cardiovascular diseases are one of the leading causes of death worldwide. The survival of patients with heart failure continues to be low with 45-60% reported deaths within five years. Apoptosis, necrosis, autophagy, and pyroptosis mediate cardiac cell death. Acute cell death is the hallmark pathogenesis of heart failure and other cardiac pathologies. Inhibition of pyroptosis, autophagy, apoptosis, or necrosis reduces cardiac damage and improves cardiac function in cardiovascular diseases. Pyroptosis is a form of inflammatory deliberate cell death that is characterized by the activation of inflammasomes such as NOD-like receptors (NLR), absent in melanoma 2 (AIM2), interferon-inducible protein 16 (IFI-16), and their downstream effector cytokines: Interleukin IL-1β and IL-18 leading to cell death. Recent studies have shown that pyroptosis is also the dominant cell death process in cardiomyocytes, cardiac fibroblasts, endothelial cells, and immune cells. It plays a crucial role in the pathogenesis of cardiac diseases that contribute to heart failure. This review intends to summarize the therapeutic implications targeting pyroptosis in the main cardiac pathologies preceding heart failure.
Collapse
Affiliation(s)
- Olive Habimana
- International College, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | | | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|
116
|
Systems Pharmacology-Based Strategy to Investigate Pharmacological Mechanisms of Total Flavonoids in Dracocephalum moldavica on Chronic Heart Failure. Int J Mol Sci 2022; 23:ijms23158409. [PMID: 35955548 PMCID: PMC9368860 DOI: 10.3390/ijms23158409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Heart failure (HF) is a clinical syndrome of cardiac insufficiency caused by abnormalities in cardiac structure and function that arise for various reasons, and it is the final stage of most cardiovascular diseases’ progression. Total flavonoid extract from Dracocephalum moldavica L. (TFDM) has many pharmacological and biological roles, such as cardioprotective, neuroprotective, anti-atherogenic, antihypertensive, anti-diabetic, anti-inflammatory, antioxidant, etc. However, its effect on HF and its molecular mechanism are still unclear. In this study, we used systems pharmacology and an animal model of HF to investigate the cardioprotective effect of TFDM and its molecular mechanism. Eleven compounds in TFDM were obtained from the literature, and 114 overlapping genes related to TFDM and HF were collected from several databases. A PPI network and C-T network were established, and GO enrichment analysis and KEGG pathway analysis were performed. The top targets from the PPI network and C-T network were validated using molecular docking. The pharmacological activity was investigated in an HFpEF (heart failure with preserved ejection fraction) mouse model. This study shows that TFDM has a protective effect on HFpEF, and its protective mechanism may be related to the regulation of proinflammatory cytokines, apoptosis-related genes, fibrosis-related genes, etc. Collectively, this study offers new insights for researchers to understand the protective effect and mechanism of TFDM against HFpEF using a network pharmacology method and a murine model of HFpEF, which suggest that TFDM is a promising therapy for HFpEF in the clinic.
Collapse
|
117
|
Singh RB, Fedacko J, Pella D, Fatima G, Elkilany G, Moshiri M, Hristova K, Jakabcin P, Vaňova N. High Exogenous Antioxidant, Restorative Treatment (Heart) for Prevention of the Six Stages of Heart Failure: The Heart Diet. Antioxidants (Basel) 2022; 11:antiox11081464. [PMID: 36009183 PMCID: PMC9404840 DOI: 10.3390/antiox11081464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 01/06/2023] Open
Abstract
The exact pathophysiology of heart failure (HF) is not yet known. Western diet, characterized by highly sweetened foods, as well as being rich in fat, fried foods, red meat and processed meat, eggs, and sweet beverages, may cause inflammation, leading to oxidative dysfunction in the cardiac ultra-structure. Oxidative function of the myocardium and how oxidative dysfunction causes physio-pathological remodeling, leading to HF, is not well known. Antioxidants, such as polyphenolics and flavonoids, omega-3 fatty acids, and other micronutrients that are rich in Indo-Mediterranean-type diets, could be protective in sustaining the oxidative functions of the heart. The cardiomyocytes use glucose and fatty acids for the physiological functions depending upon the metabolic requirements of the heart. Apart from toxicity due to glucose, lipotoxicity also adversely affects the cardiomyocytes, which worsen in the presence of deficiency of endogenous antioxidants and deficiency of exogenous antioxidant nutrients in the diet. The high-sugar-and-high-fat-induced production of ceramide, advanced glycation end products (AGE) and triamino-methyl-N-oxide (TMAO) can predispose individuals to oxidative dysfunction and Ca-overloading. The alteration in the biology may start with normal cardiac cell remodeling to biological remodeling due to inflammation. An increase in the fat content of a diet in combination with inducible nitric oxide synthase (NOSi) via N-arginine methyl ester has been found to preserve the ejection fraction in HF. It is proposed that a greater intake of high exogenous antioxidant restorative treatment (HEART) diet, polyphenolics and flavonoids, as well as cessation of red meat intake and egg, can cause improvement in the oxidative function of the heart, by inhibiting oxidative damage to lipids, proteins and DNA in the cell, resulting in beneficial effects in the early stage of the Six Stages of HF. There is an unmet need to conduct cohort studies and randomized, controlled studies to demonstrate the role of the HEART diet in the treatment of HF.
Collapse
Affiliation(s)
- Ram B. Singh
- Halberg Hospital and Research Institute, Moradabad 244001, India;
| | - Jan Fedacko
- Department of Gerontology and Geriatric, Medipark, University Research Park, PJ Safarik University, 040-11 Kosice, Slovakia
- Correspondence:
| | - Dominik Pella
- Department of Cardiology, Faculty of Medicine and East Slovak, Institute for Cardiovascular Disease, PJ Safarik University, 040-11 Kosice, Slovakia;
| | - Ghizal Fatima
- Department of Biotechnology, Era University, Lucknow 226001, India;
| | - Galal Elkilany
- International College of Cardiology, Laplace, LA 90001, USA;
| | - Mahmood Moshiri
- International College of Cardiology, Richmond Hill, ON LL-9955, Canada;
| | - Krasimira Hristova
- Department of Cardiology, National University Hospital, 1000 Sofia, Bulgaria;
| | - Patrik Jakabcin
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Králové, Charles University, 10000 Prague, Czech Republic;
| | - Natalia Vaňova
- Department of Internal Medicine UPJS MF and AGEL Hospital, Research Park, PJ Safaric University, 040-11 Kosice, Slovakia;
| |
Collapse
|
118
|
Cai X, Allison MA, Ambale-Venkatesh B, Jorgensen NW, Lima JAC, Muse ED, McClelland RL, Shea S, Lebeche D. Resistin and risks of incident heart failure subtypes and cardiac fibrosis: the Multi-Ethnic Study of Atherosclerosis. ESC Heart Fail 2022; 9:3452-3460. [PMID: 35860859 DOI: 10.1002/ehf2.14064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/04/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022] Open
Abstract
AIMS Resistin is a circulating inflammatory biomarker that is associated with cardiovascular disease. We investigated the associations of resistin and incident heart failure (HF) and its subtypes, as well as specific measures of subclinical HF (myocardial fibrosis and relevant biomarkers). METHODS We analysed data from 1968 participants in the Multi-Ethnic Study of Atherosclerosis with measurements of plasma resistin levels at clinic visits from 2002 to 2005. Participants were subsequently followed for a median of 10.5 years for HF events. The associations between resistin levels and incident HF, HF with reduced ejection fraction (HFrEF), and HF with preserved ejection fraction (HFpEF) were examined using multivariable Cox proportional hazards models. Linear regression models assessed the associations between resistin levels and myocardial fibrosis from cardiac magnetic resonance imaging, as well as hs-cTnT and NT-proBNP. RESULTS The mean age of the cohort was 64.7 years, and 50.0% were female. Seventy-four participants (4%) developed incident HF during follow-up. In a Cox proportional hazards model adjusted for age, gender, education level, race/ethnicity, and traditional risk factors, higher resistin levels were significantly associated with incident HF (HR 1.44, CI 1.18-1.75, P = 0.001) and HFrEF (HR 1.47, CI 1.07-2.02, P = 0.016), but not with HFpEF (HR 1.25, CI 0.89-1.75, P = 0.195). Resistin levels showed no significant associations with myocardial fibrosis, NT-proBNP, or hs-cTnT levels. CONCLUSIONS In a multi-ethnic cohort free of cardiovascular disease at baseline, elevated resistin levels were associated with incident HF, more prominently with incident HFrEF than HFpEF, but not with subclinical myocardial fibrosis or biomarkers of HF.
Collapse
Affiliation(s)
- Xinjiang Cai
- Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Specialty Training and Advanced Research (STAR) program, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew A Allison
- Department of Family Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bharath Ambale-Venkatesh
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Neal W Jorgensen
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Joao A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Evan D Muse
- Scripps Research Translational Institute and Division of Cardiovascular Diseases, Scripps Clinic, La Jolla, CA, USA
| | - Robyn L McClelland
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Steven Shea
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Physiology, University of Tennessee Health Science Center, 318H Translational Research Building, Memphis, TN, 38163, USA
| |
Collapse
|
119
|
Identification of Differential Expression Genes between Volume and Pressure Overloaded Hearts Based on Bioinformatics Analysis. Genes (Basel) 2022; 13:genes13071276. [PMID: 35886059 PMCID: PMC9318830 DOI: 10.3390/genes13071276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/27/2023] Open
Abstract
Volume overload (VO) and pressure overload (PO) are two common pathophysiological conditions associated with cardiac disease. VO, in particular, often occurs in a number of diseases, and no clinically meaningful molecular marker has yet been established. We intend to find the main differential gene expression using bioinformatics analysis. GSE97363 and GSE52796 are the two gene expression array datasets related with VO and PO, respectively. The LIMMA algorithm was used to identify differentially expressed genes (DEGs) of VO and PO. The DEGs were divided into three groups and subjected to functional enrichment analysis, which comprised GO analysis, KEGG analysis, and the protein–protein interaction (PPI) network. To validate the sequencing data, cardiomyocytes from AR and TAC mouse models were used to extract RNA for qRT-PCR. The three genes with random absolute values of LogFC and indicators of heart failure (natriuretic peptide B, NPPB) were detected: carboxylesterase 1D (CES1D), whirlin (WHRN), and WNK lysine deficient protein kinase 2 (WNK2). The DEGs in VO and PO were determined to be 2761 and 1093, respectively, in this study. Following the intersection, 305 genes were obtained, 255 of which expressed the opposing regulation and 50 of which expressed the same regulation. According to the GO and pathway enrichment studies, DEGs with opposing regulation are mostly common in fatty acid degradation, propanoate metabolism, and other signaling pathways. Finally, we used Cytoscape’s three techniques to identify six hub genes by intersecting 255 with the opposite expression and constructing a PPI network. Peroxisome proliferator-activated receptor (PPARα), acyl-CoA dehydrogenase medium chain (ACADM), patatin-like phospholipase domain containing 2 (PNPLA2), isocitrate dehydrogenase 3 (IDH3), heat shock protein family D member 1 (HSPD1), and dihydrolipoamide S-acetyltransferase (DLAT) were identified as six potential genes. Furthermore, we predict that the hub genes PPARα, ACADM, and PNPLA2 regulate VO myocardial changes via fatty acid metabolism and acyl-Coa dehydrogenase activity, and that these genes could be employed as basic biomarkers for VO diagnosis and treatment.
Collapse
|
120
|
Ferro F, Spelat R, Valente C, Contessotto P. Understanding How Heart Metabolic Derangement Shows Differential Stage Specificity for Heart Failure with Preserved and Reduced Ejection Fraction. Biomolecules 2022; 12:biom12070969. [PMID: 35883525 PMCID: PMC9312956 DOI: 10.3390/biom12070969] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is a clinical condition defined by structural and functional abnormalities in the heart that gradually result in reduced cardiac output (HFrEF) and/or increased cardiac pressures at rest and under stress (HFpEF). The presence of asymptomatic individuals hampers HF identification, resulting in delays in recognizing patients until heart dysfunction is manifested, thus increasing the chance of poor prognosis. Given the recent advances in metabolomics, in this review we dissect the main alterations occurring in the metabolic pathways behind the decrease in cardiac function caused by HF. Indeed, relevant preclinical and clinical research has been conducted on the metabolite connections and differences between HFpEF and HFrEF. Despite these promising results, it is crucial to note that, in addition to identifying single markers and reliable threshold levels within the healthy population, the introduction of composite panels would strongly help in the identification of those individuals with an increased HF risk. That said, additional research in the field is required to overcome the current drawbacks and shed light on the pathophysiological changes that lead to HF. Finally, greater collaborative data sharing, as well as standardization of procedures and approaches, would enhance this research field to fulfil its potential.
Collapse
Affiliation(s)
- Federico Ferro
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34125 Trieste, Italy
- Correspondence:
| | - Renza Spelat
- Neurobiology Sector, International School for Advanced Studies (SISSA), 34136 Trieste, Italy;
| | - Camilla Valente
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (C.V.); (P.C.)
| | - Paolo Contessotto
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (C.V.); (P.C.)
| |
Collapse
|
121
|
Chaar D, Dumont BL, Vulesevic B, Neagoe PE, Räkel A, White M, Sirois MG. Neutrophils and Circulating Inflammatory Biomarkers in Diabetes Mellitus and Heart Failure With Preserved Ejection Fraction. Am J Cardiol 2022; 178:80-88. [PMID: 35811144 DOI: 10.1016/j.amjcard.2022.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/13/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by low-grade chronic inflammation, which could be exacerbated by type 2 diabetes mellitus (DM). We hypothesized that neutrophils in patients with DM and patients with HFpEF with/without DM contribute to low-grade inflammation through the release of pro-inflammatory cytokines. Venous blood was withdrawn from patients with DM (n = 22), HFpEF (n = 15), HFpEF with DM (n = 13), and healthy controls (CTL) (n = 21). Levels of circulating cytokines and in vitro cytokines released by isolated neutrophils were assessed by enzyme-linked immunosorbent assay. Compared with CTL, there was a significant decrease in circulating nitric oxide in patients with DM (p ≤0.001), HFpEF (p ≤0.05), and HFpEF with DM (p ≤0.001) up to 44%. Circulating soluble intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 levels increased (up to 2.5-fold and 1.9-fold, respectively; p ≤0.001) in patients with HFpEF and patients with HFpEF and DM, whereas soluble E-selectin only increased in patients with HFpEF and DM (1.4-fold, p ≤0.001). Circulating vascular endothelial growth factor levels were similar in CTL and patients with DM but were decreased in patients with HFpEF with/without DM (up to 94%; p ≤0.001). Circulating C-reactive protein, interleukin (IL)-8, IL-6, and IL-receptor antagonist increased in all patient groups with a maximum of 3.3-fold, 4.7-fold, 4.8-fold, and 1.6-fold, respectively, in patients with HFpEF and patients with DM. In vitro, lipopolysaccharide increased neutrophils IL-6 release from HFpEF with DM (3.7-fold; p ≤0.001), and IL-8 release from DM and HFpEF with DM versus CTL (2.8-fold and 10.1-fold, respectively; p ≤0.001). IL-1 receptor antagonist and vascular endothelial growth factor release from HFpEF neutrophils significantly decreased up to 87.0% and 92.2%, respectively, versus CTL. Neutrophils from patients with DM and HFpEF release more cytokines than CTL. This increase in pro-inflammatory status may explain the greater event rate in patients with HFpEF and DM.
Collapse
Affiliation(s)
- Diana Chaar
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Benjamin L Dumont
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
| | - Branka Vulesevic
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | | | - Agnès Räkel
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Research Center, Center Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Michel White
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Martin G Sirois
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
122
|
Stevens TW, Khalaf FK, Soehnlen S, Hegde P, Storm K, Meenakshisundaram C, Dworkin LD, Malhotra D, Haller ST, Kennedy DJ, Dube P. Dirty Jobs: Macrophages at the Heart of Cardiovascular Disease. Biomedicines 2022; 10:1579. [PMID: 35884884 PMCID: PMC9312498 DOI: 10.3390/biomedicines10071579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the greatest public health concerns and is the leading cause of morbidity and mortality in the United States and worldwide. CVD is a broad yet complex term referring to numerous heart and vascular conditions, all with varying pathologies. Macrophages are one of the key factors in the development of these conditions. Macrophages play diverse roles in the maintenance of cardiovascular homeostasis, and an imbalance of these mechanisms contributes to the development of CVD. In the current review, we provide an in-depth analysis of the diversity of macrophages, their roles in maintaining tissue homeostasis within the heart and vasculature, and the mechanisms through which imbalances in homeostasis may lead to CVD. Through this review, we aim to highlight the potential importance of macrophages in the identification of preventative, diagnostic, and therapeutic strategies for patients with CVD.
Collapse
Affiliation(s)
- Travis W. Stevens
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Fatimah K. Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
- Department of Clinical Pharmacy, University of Alkafeel, Najaf 54001, Iraq
| | - Sophia Soehnlen
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Prajwal Hegde
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Kyle Storm
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Chandramohan Meenakshisundaram
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Lance D. Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - David J. Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Prabhatchandra Dube
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| |
Collapse
|
123
|
Similarity-based prediction of ejection fraction in heart failure patients. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
124
|
Saleh ZT, Alraoush AT, Aqel AA, Shawashi TO, Chung M, Lennie TA. Sex Differences in the Association Between Inflammation and Event-Free Survival in Patients With Heart Failure. J Cardiovasc Nurs 2022; 37:386-393. [PMID: 37707972 PMCID: PMC8733054 DOI: 10.1097/jcn.0000000000000831] [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] [Indexed: 11/26/2022]
Abstract
BACKGROUND Heart failure (HF) is associated with chronic inflammation, which is adversely associated with survival. Although sex-related differences in inflammation have been described in patients with HF, whether sex-related differences in inflammation are associated with event-free survival has not been examined. AIM The aim of this study was to determine whether the association between inflammation as indicated by tumor necrosis factor-α and event-free survival differs between men and women with HF after controlling for demographic and clinical variables. METHOD This was a secondary analysis of data from 301 male (age, 61.0 ± 11.4 years) and 137 female (age, 60.3 ± 12.1 years) patients with HF. Serum levels of soluble tumor necrosis factor receptor 1 were used to indicate inflammatory status. Patients were grouped according to median split of soluble tumor necrosis factor receptor 1 level and sex into male with low inflammation (≤1820 pg/mL) (n = 158) or high inflammation (>1820 pg/mL) (n = 143), and female with low inflammation (n = 63) or high inflammation (n = 74). Cox regression models were run separately for men and women to determine whether inflammation contributed to differences in event-free survival between sexes with HF. RESULTS There were 84 male (27.9%) and 27 female (19.7%) patients who had an event. Event-free survival in women did not differ by the severity of inflammation in the Cox regression analysis. In contrast, men with high inflammation had 1.85 times higher risk for an event compared with men with low inflammation. CONCLUSION These data provide evidence that inflammation contributed to differences in event-free survival in men but not women with HF. Clinicians should be aware that men who have higher inflammation may be at a greater risk of HF or cardiac-related events than others with HF.
Collapse
Affiliation(s)
- Zyad T. Saleh
- School of Nursing, The University of Jordan, Queen Rania Street, Amman, Jordan 11942
| | - Ahmad T. Alraoush
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan 22110
| | - Ahmad A. Aqel
- School of Nursing, The University of Jordan, Queen Rania Street, Amman, Jordan 11942
| | - Tagreed O. Shawashi
- School of Nursing, The University of Jordan, Queen Rania Street, Amman, Jordan 11942
| | - Misook Chung
- College of Nursing, University of Kentucky, 315 College of Nursing Building, 751 Rose Street, Lexington, KY 40536
| | - Terry A. Lennie
- College of Nursing, University of Kentucky, 315 College of Nursing Building, 751 Rose Street, Lexington, KY 40536
| |
Collapse
|
125
|
Heart failure with reduced ejection fraction and diastolic dysfunction (HrEFwDD): Time for a new clinical entity. Int J Cardiol 2022; 363:123-124. [PMID: 35760160 DOI: 10.1016/j.ijcard.2022.06.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022]
|
126
|
Fan S, Hu Y. Role of m6A Methylation in the Occurrence and Development of Heart Failure. Front Cardiovasc Med 2022; 9:892113. [PMID: 35811741 PMCID: PMC9263194 DOI: 10.3389/fcvm.2022.892113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenosine (m6A) RNA methylation is one of the most common epigenetic modifications in RNA nucleotides. It is known that m6A methylation is involved in regulation, including gene expression, homeostasis, mRNA stability and other biological processes, affecting metabolism and a variety of biochemical regulation processes, and affecting the occurrence and development of a variety of diseases. Cardiovascular disease has high morbidity, disability rate and mortality in the world, of which heart failure is the final stage. Deeper understanding of the potential molecular mechanism of heart failure and exploring more effective treatment strategies will bring good news to the sick population. At present, m6A methylation is the latest research direction, which reveals some potential links between epigenetics and pathogenesis of heart failure. And m6A methylation will bring new directions and ideas for the prevention, diagnosis and treatment of heart failure. The purpose of this paper is to review the physiological and pathological mechanisms of m6A methylation that may be involved in cardiac remodeling in heart failure, so as to explain the possible role of m6A methylation in the occurrence and development of heart failure. And we hope to help m6A methylation obtain more in-depth research in the occurrence and development of heart failure.
Collapse
|
127
|
Pilz PM, Ward JE, Chang WT, Kiss A, Bateh E, Jha A, Fisch S, Podesser BK, Liao R. Large and Small Animal Models of Heart Failure With Reduced Ejection Fraction. Circ Res 2022; 130:1888-1905. [PMID: 35679365 DOI: 10.1161/circresaha.122.320246] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) describes a heterogenous complex spectrum of pathological conditions that results in structural and functional remodeling leading to subsequent impairment of cardiac function, including either systolic dysfunction, diastolic dysfunction, or both. Several factors chronically lead to HF, including cardiac volume and pressure overload that may result from hypertension, valvular lesions, acute, or chronic ischemic injuries. Major forms of HF include hypertrophic, dilated, and restrictive cardiomyopathy. The severity of cardiomyopathy can be impacted by other comorbidities such as diabetes or obesity and external stress factors. Age is another major contributor, and the number of patients with HF is rising worldwide in part due to an increase in the aged population. HF can occur with reduced ejection fraction (HF with reduced ejection fraction), that is, the overall cardiac function is compromised, and typically the left ventricular ejection fraction is lower than 40%. In some cases of HF, the ejection fraction is preserved (HF with preserved ejection fraction). Animal models play a critical role in facilitating the understanding of molecular mechanisms of how hearts fail. This review aims to summarize and describe the strengths, limitations, and outcomes of both small and large animal models of HF with reduced ejection fraction that are currently used in basic and translational research. The driving defect is a failure of the heart to adequately supply the tissues with blood due to impaired filling or pumping. An accurate model of HF with reduced ejection fraction would encompass the symptoms (fatigue, dyspnea, exercise intolerance, and edema) along with the pathology (collagen fibrosis, ventricular hypertrophy) and ultimately exhibit a decrease in cardiac output. Although countless experimental studies have been published, no model completely recapitulates the full human disease. Therefore, it is critical to evaluate the strength and weakness of each animal model to allow better selection of what animal models to use to address the scientific question proposed.
Collapse
Affiliation(s)
- Patrick M Pilz
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA (P.M.P., E.B., R.L.).,Ludwig Boltzmann Institute at the Center for Biomedical Research, Medical University of Vienna, Austria (P.M.P., A.K., B.K.P.)
| | - Jennifer E Ward
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, MA (J.E.W., S.F., R.L.)
| | - Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Taiwan (W.-T.C.).,Department of Cardiology, Chi-Mei Medical Center, Taiwan (W.-T.C.)
| | - Attila Kiss
- Ludwig Boltzmann Institute at the Center for Biomedical Research, Medical University of Vienna, Austria (P.M.P., A.K., B.K.P.)
| | - Edward Bateh
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA (P.M.P., E.B., R.L.)
| | - Alokkumar Jha
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA (P.M.P., E.B., R.L.)
| | - Sudeshna Fisch
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, MA (J.E.W., S.F., R.L.)
| | - Bruno K Podesser
- Ludwig Boltzmann Institute at the Center for Biomedical Research, Medical University of Vienna, Austria (P.M.P., A.K., B.K.P.)
| | - Ronglih Liao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA (P.M.P., E.B., R.L.).,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, MA (J.E.W., S.F., R.L.)
| |
Collapse
|
128
|
Minotti G, Menna P, Camilli M, Salvatorelli E, Levi R. Beyond hypertension: Diastolic dysfunction associated with cancer treatment in the era of cardio-oncology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:365-409. [PMID: 35659376 DOI: 10.1016/bs.apha.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cancer patients are at an increased risk of cardiovascular events. Both old-generation cytostatics/cytotoxics and new-generation "targeted" drugs can in fact damage cardiomyocytes, endothelial cells of veins and arteries, specialized cells of the conduction system, pericardium, and valves. A new discipline, cardio-oncology, has therefore developed with the aim of protecting cancer patients from cardiovascular events, while also providing them with the best possible oncologic treatment. Anthracyclines have long been known to elicit cardiotoxicity that, depending on treatment- or patient-related factors, may progress with a variable velocity toward cardiomyopathy and systolic heart failure. However, early compromise of diastolic function may precede systolic dysfunction, and a progression of early diastolic dysfunction to diastolic rather than systolic heart failure has been documented in long-term cancer survivors. This chapter first describes general notions about hypertension in the cancer patient and then moves on reviewing the pathophysiology and clinical trajectories of diastolic dysfunction, and the molecular mechanisms of anthracycline-induced diastolic dysfunction. Diastolic dysfunction can in fact be caused and/or aggravated by hypertension. Pharmacologic foundations and therapeutic opportunities to prevent or treat diastolic dysfunction before it progresses toward heart failure are also reviewed, with a special emphasis on the mechanisms of action of drugs that raised hopes to treat diastolic dysfunction in the general population (sacubitril/valsartan, guanylyl cyclase activators, phosphodiesterase inhibitors, ranolazine, inhibitors of type-2 sodium-glucose-inked transporter). Cardio-oncologists will be confronted with the risk:benefit ratio of using these drugs in the cancer patient.
Collapse
Affiliation(s)
- Giorgio Minotti
- Department of Medicine, Campus Bio-Medico University and Fondazione Policlinico, Rome, Italy.
| | - Pierantonio Menna
- Department of Health Sciences, Campus Bio-Medico University and Fondazione Policlinico, Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome
| | - Emanuela Salvatorelli
- Department of Medicine, Campus Bio-Medico University and Fondazione Policlinico, Rome, Italy
| | - Roberto Levi
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
129
|
Rao VU, Bhasin A, Vargas J, Arun Kumar V. A multidisciplinary approach to heart failure care in the hospital: improving the patient journey. Hosp Pract (1995) 2022; 50:170-182. [PMID: 35658810 DOI: 10.1080/21548331.2022.2082776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Despite advancements in care for patients with heart failure (HF), morbidity and mortality remain high. Hospitalizations and readmissions for HF have been the focus of significant attention among health care providers and payers, with an eye towards reducing health care costs. However, considerable variability exists with regard to inpatient workflows and management for patients with HF, which represents a significant opportunity to improve care. Here we provide a summary of optimal inpatient management strategies for HF, focusing on the multidisciplinary team of emergency medicine providers, admitting hospitalists, cardiovascular consultants, pharmacists, nurses, and social workers. The patient journey serves as the template for this review article, from the initial presentation in the emergency department, to decongestion and stabilization, optimization of guideline-directed medical therapy, and discharge and appropriate disposition. Lastly, this review aims not to be proscriptive but rather to provide best practices that are clinically relevant and actionable, with the goal of improving care for patients during the sentinel hospitalization for HF.
Collapse
Affiliation(s)
- Vijay U Rao
- Indiana Heart Physicians,Franciscan Health, Indianapolis, IN, USA
| | - Atul Bhasin
- Department of Internal Medicine, CentraState Medical Center, Freehold, and Hackensack Meridian Health Hospice, Wall, NJ, USA
| | - Jesus Vargas
- University of Pennsylvania Medical Center Harrisburg Hospital, Harrisburg, PA, USA
| | - Vijaya Arun Kumar
- Department of Emergency Medicine, Wayne State University, School of Medicine, Detroit, MI, USA
| |
Collapse
|
130
|
Sonawane AR, Aikawa E, Aikawa M. Connections for Matters of the Heart: Network Medicine in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:873582. [PMID: 35665246 PMCID: PMC9160390 DOI: 10.3389/fcvm.2022.873582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/19/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular diseases (CVD) are diverse disorders affecting the heart and vasculature in millions of people worldwide. Like other fields, CVD research has benefitted from the deluge of multiomics biomedical data. Current CVD research focuses on disease etiologies and mechanisms, identifying disease biomarkers, developing appropriate therapies and drugs, and stratifying patients into correct disease endotypes. Systems biology offers an alternative to traditional reductionist approaches and provides impetus for a comprehensive outlook toward diseases. As a focus area, network medicine specifically aids the translational aspect of in silico research. This review discusses the approach of network medicine and its application to CVD research.
Collapse
Affiliation(s)
- Abhijeet Rajendra Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
131
|
Marstein HS, Witczak BN, Godang K, Schwartz T, Flatø B, Bollerslev J, Sjaastad I, Sanner H. Adipose tissue distribution is associated with cardio-metabolic alterations in adult patients with juvenile-onset dermatomyositis. Rheumatology (Oxford) 2022; 62:SI196-SI204. [PMID: 35575380 PMCID: PMC9949708 DOI: 10.1093/rheumatology/keac293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Primary aims were to compare adipose tissue distribution in adult patients with juvenile-onset DM (JDM), with matched controls. Secondary aims were to explore how adipose tissue distribution is associated with cardio-metabolic status (cardiac dysfunction and metabolic syndrome) in patients. METHODS Thirty-nine JDM patients (all aged ≥18 y, mean age 31.7 y and 51% female) were examined mean 22.7 y (s.d. 8.9 y) after disease onset and compared with 39 age/sex-matched controls. In patients, disease activity and lipodystrophy were assessed by validated tools and use of prednisolone noted. In all participants, dual-energy X-ray absorptiometry (DXA) and echocardiography were used to measure visceral adipose tissue (VAT)(g) and cardiac function, respectively. Risk factors for metabolic syndrome were measured and associations with adipose tissue distribution explored. For primary and secondary aims, respectively, P-values ≤0.05 and ≤0.01 were considered significant. RESULTS Patients exhibited a 2.4-fold increase in VAT, and reduced HDL-cholesterol values compared with controls (P-values ≤ 0.05). Metabolic syndrome was found in 25.7% of the patients and none of the controls. Cardiac dysfunction (systolic and/or diastolic) was found in 23.7% of patients and 8.1% of controls (P = 0.07). In patients, VAT levels were correlated with age, disease duration and occurrence of metabolic syndrome and cardiac dysfunction. Occurrence of lipodystrophy (P = 0.02) and male sex (P = 0.04) tended to be independently associated with cardiac dysfunction. CONCLUSION Adults with JDM showed more central adiposity and cardio-metabolic alterations than controls. Further, VAT was found increased with disease duration, which was associated with development of cardio-metabolic syndrome.
Collapse
Affiliation(s)
- Henriette S Marstein
- Correspondence to: Henriette S. Marstein, Institute for Experimental Medical Research, Oslo University Hospital, Ullevål PB 4956 Nydalen, NO-0424 Oslo, Norway. E-mail:
| | - Birgit N Witczak
- Institute for Experimental Medical Research, University of Oslo and Oslo University Hospital,KG Jebsen Centre for Cardiac Research, University of Oslo
| | | | - Thomas Schwartz
- Institute for Experimental Medical Research, University of Oslo and Oslo University Hospital,Department of Health Sciences, Oslo New University College,KG Jebsen Centre for Cardiac Research, University of Oslo
| | - Berit Flatø
- Institute for Clinical Medicine, Medical Faculty, University of Oslo,Department of Rheumatology, Oslo University Hospital, Rikshospitalet
| | - Jens Bollerslev
- Department of Endocrinology, Oslo University Hospital,Institute for Clinical Medicine, Medical Faculty, University of Oslo
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, University of Oslo and Oslo University Hospital,KG Jebsen Centre for Cardiac Research, University of Oslo,Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Helga Sanner
- Department of Health Sciences, Oslo New University College,Department of Rheumatology, Oslo University Hospital, Rikshospitalet
| |
Collapse
|
132
|
Carris NW, Mhaskar R, Coughlin E, Bracey E, Tipparaju SM, Halade GV. Novel biomarkers of inflammation in heart failure with preserved ejection fraction: analysis from a large prospective cohort study. BMC Cardiovasc Disord 2022; 22:221. [PMID: 35568817 PMCID: PMC9107006 DOI: 10.1186/s12872-022-02656-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) is a syndrome with a heterogeneous cluster of causes, including non-resolving inflammation, endothelial dysfunction, and multi-organ defects. The present study’s objective was to identify novel predictors of HFpEF. Methods The study analyzed the Multi-Ethnic Study of Atherosclerosis (MESA) to assess the association of specific markers of inflammation with new onset of HFpEF (interleukin-2 [IL-2], matrix metalloproteinase 3 [MMP3], large low-density lipoprotein cholesterol [LDL-C], and medium high-density lipoprotein cholesterol [HDL-C]). The study included men and women 45 to 84 years of age without cardiovascular disease at baseline. The primary outcome was the multivariate association of the hypothesized markers of inflammation with new-onset of HFpEF versus participants without new-onset heart failure. Participants with missing data were excluded. Results The present analysis included 6814 participants, 53% female, with a mean age of 62 years. Among the entire cohort, HFpEF was diagnosed in 151 (2.2%) participants and heart failure with reduced ejection fraction (HFrEF) was diagnosed in 146 (2.1%) participants. Participants were followed for the outcome of heart failure for a median 13.9 years. Baseline IL-2 was available for 2861 participants. The multivariate analysis included 2792 participants. Of these, 2668 did not develop heart failure, 62 developed HFpEF, 47 developed HFrEF, and 15 developed unclassified heart failure. In the multivariate regression model, IL-2 was associated with new-onset HFpEF (OR, 1.00058; 95% confidence interval, 1.00014 to 1.00102, p = 0.009) but not new-onset HFrEF. In multivariate analysis, MMP3, large LDL-C, and medium HDL-C were not associated with HFpEF or HFrEF. Conclusion These findings portend IL-2 as an important component of suboptimal inflammation in the pathogenesis of HFpEF. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02656-z.
Collapse
Affiliation(s)
- Nicholas W Carris
- Taneja College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd MDC 30, Tampa, FL, 33612, USA.
| | - Rahul Mhaskar
- Morsani College of Medicine, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Emily Coughlin
- Morsani College of Medicine, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Easton Bracey
- Taneja College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd MDC 30, Tampa, FL, 33612, USA
| | - Srinivas M Tipparaju
- Taneja College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd MDC 30, Tampa, FL, 33612, USA
| | - Ganesh V Halade
- Morsani College of Medicine, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA.
| |
Collapse
|
133
|
Marra AM, Bencivenga L, D'Assante R, Rengo G, Cittadini A. Heart failure with preserved ejection fraction: Squaring the circle between comorbidities and cardiovascular abnormalities. Eur J Intern Med 2022; 99:1-6. [PMID: 35033395 DOI: 10.1016/j.ejim.2022.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/22/2021] [Accepted: 01/05/2022] [Indexed: 12/23/2022]
Abstract
Heart Failure with preserved Ejection Fraction (HFpEF) is nowadays considered a major healthcare issue. According to forecasts two third of all Heart Failure patients will belong to this phenotype by year 2050, overwhelming those affected by Heart Failure with reduced Ejection Fraction (HFrEF). Both epidemiological and mechanistic studies support the concept that HFpEF represents true HF although aggravated by a collection of comorbidities. There is urgent need of improving its phenotyping due to the high degree of disease heterogeneity within HFpEF that lead to the failure of randomized clinical trials in demonstrating a remarkable impact of drugs in improving its morbidity and mortality.
Collapse
Affiliation(s)
- Alberto Maria Marra
- Department of Translational Medical Sciences, University of Naples "Federico II", Napoli, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), Federico II University, Naples, Italy; Center for Pulmonary Hypertension, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Leonardo Bencivenga
- Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France; Department of Advanced Biomedical Sciences, University of Naples "Federico II", Napoli, Italy
| | - Roberta D'Assante
- Department of Translational Medical Sciences, University of Naples "Federico II", Napoli, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples "Federico II", Napoli, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese, Italy
| | - Antonio Cittadini
- Department of Translational Medical Sciences, University of Naples "Federico II", Napoli, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), Federico II University, Naples, Italy.
| |
Collapse
|
134
|
Noll NA, Riley LA, Moore CS, Zhong L, Bersi MR, West JD, Zent R, Merryman WD. Loss of talin in cardiac fibroblasts results in augmented ventricular cardiomyocyte hypertrophy in response to pressure overload. Am J Physiol Heart Circ Physiol 2022; 322:H857-H866. [PMID: 35333120 PMCID: PMC9018049 DOI: 10.1152/ajpheart.00632.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/22/2022]
Abstract
Pressure overload of the heart is characterized by concentric hypertrophy and interstitial fibrosis. Cardiac fibroblasts (CFs) in the ventricular wall become activated during injury and synthesize and compact the extracellular matrix, which causes interstitial fibrosis and stiffening of the ventricular heart walls. Talin1 (Tln1) and Talin2 (Tln2) are mechanosensitive proteins that participate in focal adhesion transmission of signals from the extracellular environment to the actin cytoskeleton of CFs. The aim of the present study was to determine whether the removal of Tln1 and Tln2 from CFs would reduce interstitial fibrosis and cardiac hypertrophy. Twelve-week-old male and female Tln2-null (Tln2-/-) and Tln2-null, CF-specific Tln1 knockout (Tln2-/-;Tln1CF-/-) mice were given angiotensin-II (ANG II) (1.5 mg/kg/day) or saline through osmotic pumps for 8 wk. Cardiomyocyte area and measures of heart thickness were increased in the male ANG II-infused Tln2-/-;Tln1CF-/- mice, whereas there was no increase in interstitial fibrosis. Systolic blood pressure was increased in the female Tln2-/-;Tln1CF-/- mice after ANG II infusion compared with the Tln2-/- mice. However, there was no increase in cardiac hypertrophy in the Tln2-/-;Tln1CF-/- mice, which was seen in the Tln2-/- mice. Collectively, these data indicate that in male mice, the absence of Tln1 and Tln2 in CFs leads to cardiomyocyte hypertrophy in response to ANG II, whereas it results in a hypertrophy-resistant phenotype in female mice. These findings have important implications for the role of mechanosensitive proteins in CFs and their impact on cardiomyocyte function in the pathogenesis of hypertension and cardiac hypertrophy.NEW & NOTEWORTHY The role of talins has been previously studied in cardiomyocytes; however, these mechanotransductive proteins that are members of the focal adhesion complex have not been examined in cardiac fibroblasts previously. We hypothesized that loss of talins in cardiac fibroblasts would reduce interstitial fibrosis in the heart with a pressure overload model. However, we found that although loss of talins did not alter fibrosis, it did result in cardiomyocyte and ventricular hypertrophy.
Collapse
Affiliation(s)
- Natalie A Noll
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Lance A Riley
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Christy S Moore
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lin Zhong
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mathew R Bersi
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - James D West
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Roy Zent
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
135
|
van Ham WB, Kessler EL, Oerlemans MI, Handoko ML, Sluijter JP, van Veen TA, den Ruijter HM, de Jager SC. Clinical Phenotypes of Heart Failure With Preserved Ejection Fraction to Select Preclinical Animal Models. JACC Basic Transl Sci 2022; 7:844-857. [PMID: 36061340 PMCID: PMC9436760 DOI: 10.1016/j.jacbts.2021.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/31/2021] [Indexed: 11/21/2022]
Abstract
To better define HFpEF clinically, patients are nowadays often clustered into phenogroups, based on their comorbidities and symptoms Many animal models claim to mimic HFpEF, but phenogroups are not yet regularly used to cluster them HFpEF animals models often lack reports of clinical symptoms of HF, therefore mainly presenting as extended models of LVDD, clinically seen as a prestate of HFpEF We investigated if clinically relevant phenogroups can guide selection of animal models aiming at better defined animal research
At least one-half of the growing heart failure population consists of heart failure with preserved ejection fraction (HFpEF). The limited therapeutic options, the complexity of the syndrome, and many related comorbidities emphasize the need for adequate experimental animal models to study the etiology of HFpEF, as well as its comorbidities and pathophysiological changes. The strengths and weaknesses of available animal models have been reviewed extensively with the general consensus that a “1-size-fits-all” model does not exist, because no uniform HFpEF patient exists. In fact, HFpEF patients have been categorized into HFpEF phenogroups based on comorbidities and symptoms. In this review, we therefore study which animal model is best suited to study the different phenogroups—to improve model selection and refinement of animal research. Based on the published data, we extrapolated human HFpEF phenogroups into 3 animal phenogroups (containing small and large animals) based on reports and definitions of the authors: animal models with high (cardiac) age (phenogroup aging); animal models focusing on hypertension and kidney dysfunction (phenogroup hypertension/kidney failure); and models with hypertension, obesity, and type 2 diabetes mellitus (phenogroup cardiometabolic syndrome). We subsequently evaluated characteristics of HFpEF, such as left ventricular diastolic dysfunction parameters, systemic inflammation, cardiac fibrosis, and sex-specificity in the different models. Finally, we scored these parameters concluded how to best apply these models. Based on our findings, we propose an easy-to-use classification for future animal research based on clinical phenogroups of interest.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Elise L. Kessler
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University of Utrecht, Utrecht, the Netherlands
| | | | - M. Louis Handoko
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Joost P.G. Sluijter
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University of Utrecht, Utrecht, the Netherlands
| | - Toon A.B. van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hester M. den Ruijter
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia C.A. de Jager
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Address for correspondence: Dr Saskia C.A. de Jager, Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
136
|
Yoshida T, Shibata A, Tanihata A, Hayashi H, Yamaguchi Y, Kitada R, Ehara S, Izumiya Y, Yoshiyama M. Thigh Intramuscular Fat on Prognosis of Patients With Nonischemic Cardiomyopathy. Am J Cardiol 2022; 169:113-119. [PMID: 35067348 DOI: 10.1016/j.amjcard.2021.12.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 11/01/2022]
Abstract
Skeletal muscle atrophy is an independent prognostic predictor for patients with chronic heart failure, and the concept of sarcopenia is drawing attention. Furthermore, the importance of not only muscle mass but also ectopic fat has been pointed out. However, there is a lack of consensus on the implications of ectopic fat for the prognosis in patients with nonischemic cardiomyopathy. We investigated whether ectopic fat in the thigh affects the prognosis of nonischemic cardiomyopathy. This prospective study recruited 145 patients diagnosed with nonischemic cardiomyopathy between September 2017 and January 2020. Finally, 93 patients with a reduced ejection fraction were enrolled. The clinical end points were cardiovascular death or unexpected rehospitalization because of a cardiac event. Using computed tomography, the percentage of intramuscular fat (%IMF) in the thigh was measured in all patients. Patients were divided into 2 groups based on the median %IMF. The results of Spearman's correlation coefficient analysis revealed a correlation among %IMF and peak oxygen uptake (Spearman r = -0.221, p = 0.036). Kaplan-Meier analysis results showed significantly higher risk of adverse events in the high %IMF group (log-rank p = 0.013). Multivariate Cox regression analysis results revealed the %IMF as an independent factor for adverse events (hazard ratio 1.361; 95% confidence interval 1.043 to 1.745; p = 0.018). In conclusion, %IMF may have adverse consequences such as increased cardiac events in patients with nonischemic cardiomyopathy with a reduced ejection fraction.
Collapse
|
137
|
Zelnick LR, Shlipak MG, Soliman EZ, Anderson A, Christenson R, Kansal M, Deo R, He J, Jaar BG, Weir MR, Rao P, Cohen DL, Cohen JB, Feldman HI, Go A, Bansal N. Prediction of Incident Heart Failure in CKD: The CRIC Study. Kidney Int Rep 2022; 7:708-719. [PMID: 35497796 PMCID: PMC9039424 DOI: 10.1016/j.ekir.2022.01.1067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Heart failure (HF) is common in chronic kidney disease (CKD); identifying patients with CKD at high risk for HF may guide clinical care. We assessed the prognostic value of cardiac biomarkers and echocardiographic variables for 10-year HF prediction compared with a published clinical HF prediction equation in a cohort of participants with CKD. Methods We studied 2147 Chronic Renal Insufficiency Cohort (CRIC) participants without prior HF with complete clinical, cardiac biomarker (N-terminal brain natriuretic peptide [NT-proBNP] and high sensitivity troponin-T [hsTnT]), and echocardiographic data (left ventricular mass [LVM] and left ventricular ejection fraction [LVEF] data). We compared the discrimination of the 11-variable Atherosclerosis Risk in Communities (ARIC) HF prediction equation with LVM, LVEF, hsTnT, and NT-proBNP to predict 10-year risk of hospitalization for HF using a Fine and Gray modeling approach. We separately evaluated prediction of HF with preserved and reduced LVEF (LVEF ≥50% and <50%, respectively). We assessed discrimination with internally valid C-indices using 10-fold cross-validation. Results Participants' mean (SD) age was 59 (11) years, 53% were men, 43% were Black, and mean (SD) estimated glomerular filtration rate (eGFR) was 44 (16) ml/min per 1.73 m2. A total of 324 incident HF hospitalizations occurred during median (interquartile range) 10.0 (5.7-10.0) years of follow-up. The ARIC HF model with clinical variables had a C-index of 0.68. Echocardiographic variables predicted HF (C-index 0.70) comparably to the published ARIC HF model, while NT-proBNP and hsTnT together (C-index 0.73) had significantly better discrimination (P = 0.004). A model including cardiac biomarkers, echocardiographic variables, and clinical variables had a C-index of 0.77. Discrimination of HF with preserved LVEF was lower than for HF with reduced LVEF for most models. Conclusion The ARIC HF prediction model for 10-year HF risk had modest discrimination among adults with CKD. NT-proBNP and hsTnT discriminated better than the ARIC HF model and at least as well as a model with echocardiographic variables. HF clinical prediction models tailored to adults with CKD are needed. Until then, measurement of NT-proBNP and hsTnT may be a low-burden approach to predicting HF in this population, as they offer moderate discrimination.
Collapse
Affiliation(s)
- Leila R. Zelnick
- Kidney Research Institute, Department of Medicine (Nephrology), University of Washington, Seattle, Washington, USA
| | - Michael G. Shlipak
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Elsayed Z. Soliman
- Department of Internal Medicine (Cardiology), Wake Forest University, Winston-Salem, North Carolina, USA
| | - Amanda Anderson
- Department of Epidemiology, Tulane University, New Orleans, Louisiana, USA
| | - Robert Christenson
- Department of Pathology, University of Maryland, Baltimore, Maryland, USA
| | - Mayank Kansal
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Rajat Deo
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, Louisiana, USA
| | - Bernard G. Jaar
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew R. Weir
- Department of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Panduranga Rao
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Debbie L. Cohen
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordana B. Cohen
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harold I. Feldman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan Go
- Kaiser Permanente Northern California, Oakland, California, USA
| | - Nisha Bansal
- Kidney Research Institute, Department of Medicine (Nephrology), University of Washington, Seattle, Washington, USA
| |
Collapse
|
138
|
Mongirdienė A, Skrodenis L, Varoneckaitė L, Mierkytė G, Gerulis J. Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies. Biomedicines 2022; 10:602. [PMID: 35327404 PMCID: PMC8945343 DOI: 10.3390/biomedicines10030602] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
With respect to structural and functional cardiac disorders, heart failure (HF) is divided into HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Oxidative stress contributes to the development of both HFrEF and HFpEF. Identification of a broad spectrum of reactive oxygen species (ROS)-induced pathways in preclinical models has provided new insights about the importance of ROS in HFrEF and HFpEF development. While current treatment strategies mostly concern neuroendocrine inhibition, recent data on ROS-induced metabolic pathways in cardiomyocytes may offer additional treatment strategies and targets for both of the HF forms. The purpose of this article is to summarize the results achieved in the fields of: (1) ROS importance in HFrEF and HFpEF pathophysiology, and (2) treatments for inhibiting ROS-induced pathways in HFrEF and HFpEF patients. ROS-producing pathways in cardiomyocytes, ROS-activated pathways in different HF forms, and treatment options to inhibit their action are also discussed.
Collapse
Affiliation(s)
- Aušra Mongirdienė
- Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, LT-50161 Kaunas, Lithuania
| | - Laurynas Skrodenis
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| | - Leila Varoneckaitė
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| | - Gerda Mierkytė
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| | - Justinas Gerulis
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| |
Collapse
|
139
|
Kuna J, Żuber Z, Chmielewski G, Gromadziński L, Krajewska-Włodarczyk M. Role of Distinct Macrophage Populations in the Development of Heart Failure in Macrophage Activation Syndrome. Int J Mol Sci 2022; 23:2433. [PMID: 35269577 PMCID: PMC8910409 DOI: 10.3390/ijms23052433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is one of the few entities in rheumatology with the potential to quickly cause multiple organ failure and loss of life, and as such, requires urgent clinical intervention. It has a broad symptomatology, depending on the organs it affects. One especially dangerous aspect of MAS's course of illness is myocarditis leading to acute heart failure and possibly death. Research in recent years has proved that macrophages settled in different organs are not a homogenous group, with particular populations differing in both structure and function. Within the heart, we can determine two major groups, based on the presence of the C-C 2 chemokine receptor (CCR2): CCR2+ and CCR2-. There are a number of studies describing their function and the changes in the population makeup between normal conditions and different illnesses; however, to our knowledge, there has not been one touching on the matter of changes occurring in the populations of heart macrophages during MAS and their possible consequences. This review summarizes the most recent knowledge on heart macrophages, the influence of select cytokines (those particularly significant in the development of MAS) on their activity, and both the immediate and long-term consequences of changes in the makeup of specific macrophage populations-especially the loss of CCR2- cells that are responsible for regenerative processes, as well as the substitution of tissue macrophages by the highly proinflammatory CCR2+ macrophages originating from circulating monocytes. Understanding the significance of these processes may lead to new discoveries that could improve the therapeutic methods in the treatment of MAS.
Collapse
Affiliation(s)
- Jakub Kuna
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Zbigniew Żuber
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Kraków University, 30-705 Kraków, Poland;
| | - Grzegorz Chmielewski
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| |
Collapse
|
140
|
Kobak KA, Zarzycka W, Chiao YA. Age and Sex Differences in Heart Failure With Preserved Ejection Fraction. FRONTIERS IN AGING 2022; 3:811436. [PMID: 35821846 PMCID: PMC9261310 DOI: 10.3389/fragi.2022.811436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/13/2022] [Indexed: 11/29/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multi-organ disorder that represents about 50% of total heart failure (HF) cases and is the most common form of HF in the elderly. Because of its increasing prevalence caused by the aging population, high mortality and morbidity, and very limited therapeutic options, HFpEF is considered as one of the greatest unmet medical needs in cardiovascular medicine. Despite its complex pathophysiology, numerous preclinical models have been established in rodents and in large animals to study HFpEF pathophysiology. Although age and sex differences are well described in HFpEF population, there are knowledge gaps in sex- and age-specific differences in established preclinical models. In this review, we summarize various strategies that have been used to develop HFpEF models and discuss the knowledge gaps in sex and age differences in HFpEF.
Collapse
|
141
|
Rabinovich-Nikitin I, Kirshenbaum LA. Mef2 Regulated Cardiac Hypertrophy and Heart Failure in Hypertension. Trends Cardiovasc Med 2022; 33:213-214. [PMID: 35092849 DOI: 10.1016/j.tcm.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada; Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
142
|
Left atrial structure and function in heart failure with reduced (HFrEF) versus preserved ejection fraction (HFpEF): systematic review and meta-analysis. Heart Fail Rev 2022; 27:1933-1955. [PMID: 35079942 PMCID: PMC9388424 DOI: 10.1007/s10741-021-10204-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 01/10/2023]
Abstract
Left atrial (LA) structure and function in heart failure with reduced (HFrEF) versus preserved ejection fraction (HFpEF) is only established in small studies. Therefore, we conducted a systematic review of LA structure and function in order to find differences between patients with HFrEF and HFpEF. English literature on LA structure and function using echocardiography was reviewed to calculate pooled prevalence and weighted mean differences (WMD). A total of 61 studies, comprising 8806 patients with HFrEF and 9928 patients with HFpEF, were included. The pooled prevalence of atrial fibrillation (AF) was 34.4% versus 42.8% in the acute inpatient setting, and 20.1% versus 33.1% in the chronic outpatient setting when comparing between HFrEF and HFpEF. LA volume index (LAVi), LA reservoir global longitudinal strain (LAGLSR), and E/e’ was 59.7 versus 52.7 ml/m2, 9.0% versus 18.9%, and 18.5 versus 14.0 in the acute inpatient setting, and 48.3 versus 38.2 ml/m2, 12.8% versus 23.4%, and 16.9 versus 13.5 in the chronic outpatient setting when comparing HFrEF versus HFpEF, respectively. The relationship between LAVi and LAGLSR was significant in HFpEF, but not in HFrEF. Also, in those studies that directly compared patients with HFrEF versus HFpEF, those with HFrEF had worse LAGLSR [WMD = 16.3% (22.05,8.61); p < 0.001], and higher E/e’ [WMD = −0.40 (−0.56, −0.24); p < 0.05], while LAVi was comparable. When focusing on acute hospitalized patients, E/e’ was comparable between patients with HFrEF and HFpEF. Despite the higher burden of AF in HFpEF, patients with HFrEF had worse LA global function. Left atrial myopathy is not specifically related to HFpEF.
Collapse
|
143
|
Mongirdienė A, Liobikas J. Phenotypic and Functional Heterogeneity of Monocyte Subsets in Chronic Heart Failure Patients. BIOLOGY 2022; 11:195. [PMID: 35205062 PMCID: PMC8869357 DOI: 10.3390/biology11020195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
Chronic heart failure (CHF) results when the heart cannot consistently supply the body's tissues with oxygen and required nutrients. CHF can be categorized as heart failure (HF) with preserved ejection fraction (HFpEF) or HF with reduced ejection fraction (HFrEF). There are different causes and mechanisms underlying HF pathogenesis; however, inflammation can be regarded as one of the factors that promotes both HFrEF and HFpEF. Monocytes, a subgroup of leukocytes, are known to be cellular mediators in response to cardiovascular injury and are closely related to inflammatory reactions. These cells are a vital component of the immune system and are the source of macrophages, which participate in cardiac tissue repair after injury. However, these monocytes are not as homogenous as thought and can present different functions under different cardiovascular disease conditions. In addition, there is still an open question regarding whether the functions of monocytes and macrophages should be regarded as causes or consequences in CHF development. Therefore, the aim of this work was to summarize current studies on the functions of various monocyte subsets in CHF with a focus on the role of a certain monocyte subset in HFpEF and HFrEF patients, as well as the subsets' relationship to inflammatory markers.
Collapse
Affiliation(s)
- Aušra Mongirdienė
- Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, LT50161 Kaunas, Lithuania
| | - Julius Liobikas
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, LT50162 Kaunas, Lithuania
| |
Collapse
|
144
|
Singh RB, Sozzi FB, Fedacko J, Hristova K, Fatima G, Pella D, Cornelissen G, Isaza A, Pella D, Singh J, Carugo S, Nanda NC, Elkilany GEN. Pre-heart failure at 2D- and 3D-speckle tracking echocardiography: A comprehensive review. Echocardiography 2022; 39:302-309. [PMID: 35026047 DOI: 10.1111/echo.15284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 11/20/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic heart failure (CHF) has different stages and includes pre-HF (PHF), a state of high risk of developing myocardial dysfunction and advanced CHF. Some major behavioral risk factors of PHF might predispose to biological risk factors such as obesity, diabetes mellitus, dyslipidemia, hypertension, myocardial infarction, and cardiomyopathy. These risk factors damage the myocytes leading to fibrosis, apoptosis, cardiac hypertrophy, along with alterations in cardiomyocyte' size and shape. A condition of physiological subcellular remodeling resulting into a pathological state might be developed, conducting to PHF. Both PHF and heart failure (HF) are associated with the activation of phospholipases and protease, mitochondrial dysfunction, oxidative stress and development of intra-cellular free Ca2+ [Ca2+ ]i overloading to an elevation in diastolic [Ca2+ ]i . Simultaneously, cardiac gene expression is activated leading to further molecular, structural and biochemical changes of the myocardium. The sub-cellular remodeling may be intimately involved in the transition of cardiac hypertrophy to heart failure. 2D- and 3D-speckle tracking echocardiography (STE) have been used to quantify regional alterations of longitudinal strain and area strain, through their polar projection, which permits a further assessment of both sites and degrees of myocardial damage. The examination of strain can identify sub-clinical cardiac dysfunction or cardiomyocyte remodeling. During remodeling of the myocardium cardiac strain is attenuated, therefore it is an indicator of disease assessment.
Collapse
Affiliation(s)
- Ram B Singh
- Internal Medicine/Cardiology, TsimTsoum Institute, Krakow, Poland & Halberg Hospital, Moradabad, India
| | - Fabiola B Sozzi
- Cardiology Unit, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, Milan, IT
| | - Jan Fedacko
- Faculty of Medicine, PJ Safaric University, Kosice, Slovakia
| | - Krasimira Hristova
- Department of Intensive Cardiology, National Heart Hospital, Sofia, Bulgaria
| | - Ghizal Fatima
- Department of biotechnology, Era's Medical College and Hospital, Lucknow, India
| | - Daniel Pella
- Faculty of Medicine, PJ Safaric University, Kosice, Slovakia
| | - Germaine Cornelissen
- Halberg Chronobiology Center, University of Minnesota Medical School, Minneapolis, USA
| | - Adrian Isaza
- Adrian Isaza (PhD) Department of medicine, Everglade University, Tampa, Florida, USA
| | - Dominik Pella
- Department of Cardiology, East Slovak Institute of Medical Sciences, Kosice, Slovakia
| | - Jaipaul Singh
- School of Natural Sciences, University of Central Lancashire, Preston, UK
| | - Stefano Carugo
- Cardiology Unit, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, Milan, IT
| | - Navin C Nanda
- Dpt. of Medicine and Cardiovascular Disease, University of Alabama, Birmingham, Alabama, USA
| | - Galal E Nagib Elkilany
- Department of Cardiology, Gulf Medical University-Thumbay Hospitals, Fujairah, United Arab Emirates, 1002, UAE
| |
Collapse
|
145
|
Berezin AA, Fushtey IM, Berezin AE. Discriminative Utility of Apelin-to-NT-Pro-Brain Natriuretic Peptide Ratio for Heart Failure with Preserved Ejection Fraction among Type 2 Diabetes Mellitus Patients. J Cardiovasc Dev Dis 2022; 9:23. [PMID: 35050233 PMCID: PMC8779441 DOI: 10.3390/jcdd9010023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/01/2022] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Apelin is a regulatory vasoactive peptide, which plays a pivotal role in adverse cardiac remodeling and heart failure (HF) with reduced ejection fraction. The purpose of the study was to investigate whether serum levels of apelin is associated with HF with preserved election fraction (HFpEF) in patients with T2DM. METHODS The study retrospectively involved 101 T2DM patients aged 41 to 62 years (48 patients with HFpEF and 28 non-HFpEF patients). The healthy control group consisted of 25 individuals with matched age and sex. Data collection included demographic and anthropometric information, hemodynamic performances and biomarkers of the disease. Transthoracic B-mode echocardiography, Doppler and TDI were performed at baseline. Serum levels of N-terminal pro-brain natriuretic peptide (NT-proBNP) and apelin were measured by ELISA in all patients at the study entry. RESULTS Unadjusted multivariate logistic model yielded the only apelin to NT-proBNP ratio (OR = 1.44; p = 0.001), BMI > 34 кг/м2 (OR = 1.07; p = 0.036), NT-proBNP > 458 pmol/mL (OR = 1.17; p = 0.042), LAVI > 34 mL/m2 (OR = 1.06; p = 0.042) and E/e' > 11 (OR = 1.04; p = 0.044) remained to be strong predictors for HFpEF. After obesity adjustment, multivariate logistic regression showed that the apelin to NT-proBNP ratio < 0.82 × 10-2 units remained sole independent predictor for HFpEF (OR = 1.44; 95% CI: 1.18-2.77; p = 0.001) HFpEF in T2DM patients. In conclusion, we found that apelin to NT-proBNP ratio < 0.82 × 10-2 units better predicted HFpEF in T2DM patients than apelin and NT-proBNP alone. This finding could open new approach for CV risk stratification of T2DM at higher risk of HF.
Collapse
Affiliation(s)
- Alexander A. Berezin
- Internal Medicine Department, Medical Academy of Postgraduate Education, 69096 Zaporozhye, Ukraine; (A.A.B.); (I.M.F.)
| | - Ivan M. Fushtey
- Internal Medicine Department, Medical Academy of Postgraduate Education, 69096 Zaporozhye, Ukraine; (A.A.B.); (I.M.F.)
| | - Alexander E. Berezin
- Internal Medicine Department, State Medical University, 69096 Zaporozhye, Ukraine
| |
Collapse
|
146
|
Cornwell JD, McDermott JC. MEF2 in cardiac hypertrophy in response to hypertension. Trends Cardiovasc Med 2022; 33:204-212. [PMID: 35026393 DOI: 10.1016/j.tcm.2022.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/14/2022]
Abstract
Hypertension is a globally prevalent pathological condition and an underlying risk factor for the development of cardiac hypertrophy leading to heart failure. Myocyte enhancer factor 2 (Mef2) has been identified as one of the primary effectors of morphological changes in the hypertensive heart, as part of a complex network of molecular signaling controlling cardiac gene expression. Experimental chronic pressure-overload models that mimic hypertension in the mammalian heart lead to the activation of various pathological mechanisms that result in structural changes leading to debilitating cardiac hypertrophy and ultimately heart failure. The purpose here is to survey the literature implicating Mef2 in hypertension induced cardiac hypertrophy, towards illuminating points of interest for understanding and potentially treating heart failure.
Collapse
Affiliation(s)
- James D Cornwell
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; Muscle Health Research Centre (MHRC), York University, Toronto, ON M3J 1P3, Canada; Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
147
|
Elevated Serum Fibroblast Growth Factor 21 Is Relevant to Heart Failure Patients with Reduced Ejection Fraction. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7138776. [PMID: 35069790 PMCID: PMC8767358 DOI: 10.1155/2022/7138776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022]
Abstract
Objective The aim of this study was to evaluate the roles of fibroblast growth factor 21 (FGF21) in heart failure patients with reduced ejection fraction and its association with Heart Failure with reduced Ejection Fraction (HFrEF). Methods The level of FGF21 was measured by enzyme-linked immunosorbent assay (ELISA) in 199 subjects enrolled in this study, including 128 subjects with HFrEF and 71 control subjects. The mean follow-up time was 13.36 months. The left ventricular end-diastolic diameter (LVEDD) and left ventricular ejection fraction (LVEF) percentage were evaluated by the 2D echocardiography. Serum brain natriuretic peptide (BNP) was measured in the routine clinical laboratory. Results The serum FGF21 level was evidently higher in patients with HFrEF than in the control group (228.72 ± 24.04 vs. 171.60 ± 12.98, p < 0.001). After 1 year of follow-up, 61 patients (47.66%) with heart failure were readmitted to the hospital, including 8 deaths (13.11%). The AUC of the receiver operating characteristic (ROC) curve for the predictive value of FGF21 for prognosis was 0.964. Kaplan-Meier analysis results showed that there were significant differences in the 1-year mortality and heart failure readmission events between the grouped subjects. A poor prognosis was correlated with the serum level of FGF21, BNP, LVEDD, and LVEF, which was confirmed by the univariate Cox analysis. Conclusion FGF21 was independently associated with an increased risk of mortality and readmission HFrEF patients. Therefore, FGF21 has the potential to be a biomarker for the progression of HFrEF in patients.
Collapse
|
148
|
Main A, Boguslavskyi A, Howie J, Kuo CW, Rankin A, Burton FL, Smith GL, Hajjar R, Baillie GS, Campbell KS, Shattock MJ, Fuller W. Dynamic but discordant alterations in zDHHC5 expression and palmitoylation of its substrates in cardiac pathologies. Front Physiol 2022; 13:1023237. [PMID: 36277202 PMCID: PMC9581287 DOI: 10.3389/fphys.2022.1023237] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
S-palmitoylation is an essential lipid modification catalysed by zDHHC-palmitoyl acyltransferases that regulates the localisation and activity of substrates in every class of protein and tissue investigated to date. In the heart, S-palmitoylation regulates sodium-calcium exchanger (NCX1) inactivation, phospholemman (PLM) inhibition of the Na+/K+ ATPase, Nav1.5 influence on membrane excitability and membrane localisation of heterotrimeric G-proteins. The cell surface localised enzyme zDHHC5 palmitoylates NCX1 and PLM and is implicated in injury during anoxia/reperfusion. Little is known about how palmitoylation remodels in cardiac diseases. We investigated expression of zDHHC5 in animal models of left ventricular hypertrophy (LVH) and heart failure (HF), along with HF tissue from humans. zDHHC5 expression increased rapidly during onset of LVH, whilst HF was associated with decreased zDHHC5 expression. Paradoxically, palmitoylation of the zDHHC5 substrate NCX1 was significantly reduced in LVH but increased in human HF, while palmitoylation of the zDHHC5 substrate PLM was unchanged in all settings. Overexpression of zDHHC5 in rabbit ventricular cardiomyocytes did not alter palmitoylation of its substrates or overall cardiomyocyte contractility, suggesting changes in zDHHC5 expression in disease may not be a primary driver of pathology. zDHHC5 itself is regulated by post-translational modifications, including palmitoylation in its C-terminal tail. We found that in HF palmitoylation of zDHHC5 changed in the same manner as palmitoylation of NCX1, suggesting additional regulatory mechanisms may be involved. This study provides novel evidence that palmitoylation of cardiac substrates is altered in the setting of HF, and that expression of zDHHC5 is dysregulated in both hypertrophy and HF.
Collapse
Affiliation(s)
- Alice Main
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andri Boguslavskyi
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom
| | - Jacqueline Howie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chien-Wen Kuo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Aileen Rankin
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Francis L Burton
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Roger Hajjar
- Flagship Pioneering, Cambridge, MA, United States
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, United States
| | - Michael J Shattock
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom
| | - William Fuller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
149
|
Sanhueza-Olivares F, Troncoso MF, Pino-de la Fuente F, Martinez-Bilbao J, Riquelme JA, Norambuena-Soto I, Villa M, Lavandero S, Castro PF, Chiong M. A potential role of autophagy-mediated vascular senescence in the pathophysiology of HFpEF. Front Endocrinol (Lausanne) 2022; 13:1057349. [PMID: 36465616 PMCID: PMC9713703 DOI: 10.3389/fendo.2022.1057349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is one of the most complex and most prevalent cardiometabolic diseases in aging population. Age, obesity, diabetes, and hypertension are the main comorbidities of HFpEF. Microvascular dysfunction and vascular remodeling play a major role in its development. Among the many mechanisms involved in this process, vascular stiffening has been described as one the most prevalent during HFpEF, leading to ventricular-vascular uncoupling and mismatches in aged HFpEF patients. Aged blood vessels display an increased number of senescent endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). This is consistent with the fact that EC and cardiomyocyte cell senescence has been reported during HFpEF. Autophagy plays a major role in VSMCs physiology, regulating phenotypic switch between contractile and synthetic phenotypes. It has also been described that autophagy can regulate arterial stiffening and EC and VSMC senescence. Many studies now support the notion that targeting autophagy would help with the treatment of many cardiovascular and metabolic diseases. In this review, we discuss the mechanisms involved in autophagy-mediated vascular senescence and whether this could be a driver in the development and progression of HFpEF.
Collapse
Affiliation(s)
- Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Francisco Pino-de la Fuente
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Javiera Martinez-Bilbao
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Jaime A. Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Monica Villa
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Pablo F. Castro
- Advanced Center for Chronic Diseases, Faculty of Medicine, Pontifical University Catholic of Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- *Correspondence: Mario Chiong,
| |
Collapse
|
150
|
Hsu Y, Huang K, Cheng K. Resuscitating the Field of Cardiac Regeneration: Seeking Answers from Basic Biology. Adv Biol (Weinh) 2021; 6:e2101133. [PMID: 34939372 DOI: 10.1002/adbi.202101133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is one of the leading causes for hospital admissions worldwide. HF patients are classified based on the chronic changes in left ventricular ejection fraction (LVEF) as preserved (LVEF ≥ 50%), reduced (LVEF ≤ 40%), or mid-ranged (40% < LVEF < 50%) HFs. Treatments nowadays can prevent HFrEF progress, whereas only a few of the treatments have been proven to be effective in improving the survival of HFpEF. In this review, numerous mediators involved in the pathogenesis of HF are summarized. The regional upstream signaling and their diagnostic and therapeutic potential are also discussed. Additionally, the recent challenges and development in cardiac regenerative therapy that hold opportunities for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Yaching Hsu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|