101
|
Løgstrup BB. Heart Failure in Rheumatic Disease: Secular Trends and Novel Insights. Rheum Dis Clin North Am 2023; 49:67-79. [PMID: 36424027 DOI: 10.1016/j.rdc.2022.08.003] [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: 11/22/2022]
Abstract
There is a significant increase in risk of heart failure in several rheumatic diseases. Common cardiovascular risk factors and inflammatory processes, present in both rheumatic diseases and heart failure, are contributing to this increase. The opportunities for using immune-based strategies to fight development of heart failure in rheumatic diseases are evolving. The diversity of inflammation calls for a tailored characterization of inflammation, enabling differentiation of inflammation and subsequent introduction of precision medicine using target-specific strategies and immunomodulatory therapy. As the field of rheuma-cardiology is still evolving, clear recommendations cannot be given yet.
Collapse
Affiliation(s)
- Brian Bridal Løgstrup
- Department of Cardiology, Institute of Clinical Medicine, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, Aarhus N 8200, Denmark.
| |
Collapse
|
102
|
Cai Z, Wu C, Xu Y, Cai J, Zhao M, Zu L. The NO-cGMP-PKG Axis in HFpEF: From Pathological Mechanisms to Potential Therapies. Aging Dis 2023; 14:46-62. [PMID: 36818566 PMCID: PMC9937694 DOI: 10.14336/ad.2022.0523] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for almost half of all heart failure (HF) cases worldwide. Unfortunately, its incidence is expected to continue to rise, and effective therapy to improve clinical outcomes is lacking. Numerous efforts currently directed towards the pathophysiology of human HFpEF are uncovering signal transduction pathways and novel therapeutic targets. The nitric oxide-cyclic guanosine phosphate-protein kinase G (NO-cGMP-PKG) axis has been described as an important regulator of cardiac function. Suppression of the NO-cGMP-PKG signalling pathway is involved in the progression of HFpEF. Therefore, the NO-cGMP-PKG signalling pathway is a potential therapeutic target for HFpEF. In this review, we aim to explore the mechanism of NO-cGMP-PKG in the progression of HFpEF and to summarize potential therapeutic drugs that target this signalling pathway.
Collapse
Affiliation(s)
- Zhulan Cai
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Cencen Wu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Yuan Xu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Jiageng Cai
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Menglin Zhao
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
| | - Lingyun Zu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, China.
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, China.
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
103
|
Xu Q, Zhao YM, He NQ, Gao R, Xu WX, Zhuo XJ, Ren Z, Wu CY, Liu LS. PCSK9: A emerging participant in heart failure. Biomed Pharmacother 2023; 158:114106. [PMID: 36535197 DOI: 10.1016/j.biopha.2022.114106] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Heart failure (HF) is a complex clinical syndrome caused by various cardiovascular diseases. Its main pathogenesis includes cardiomyocyte loss, myocardial energy metabolism disorder, and activation of cardiac inflammation. Due to the clinically unsatisfactory treatment of heart failure, different mechanisms need to be explored to provide new targets for the treatment of this disease. Proprotein convertase subtilisin/kexin type 9 (PCSK9), a gene mainly related to familial hypercholesterolemia, was discovered in 2003. Aside from regulating lipid metabolism, PCSK9 may be involved in other biological processes such as apoptosis, autophagy, pyroptosis, inflammation, and tumor immunity and related to diabetes and neurodegenerative diseases. Recently, clinical data have shown that the circulating PCSK9 level is significantly increased in patients with heart failure, and it is related to the prognosis for heart failure. Furthermore, in animal models and patients with myocardial infarction, PCSK9 in the infarct margin area was also found to be significantly increased, which further suggested that PCSK9 might be closely related to heart failure. However, the specific mechanism of how PCSK9 participates in heart failure remains to be further explored. The purpose of this review is to summarize the potential mechanism of PCSK9's involvement in heart failure, thereby providing a new treatment strategy for heart failure.
Collapse
Affiliation(s)
- Qian Xu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Yi-Meng Zhao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Nai-Qi He
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Rong Gao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Wen-Xin Xu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Xiu-Juan Zhuo
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Chun-Yan Wu
- The Third Affiliated Hospital, Department of Cardiovascular Medicine, University of South China, Hengyang, Hunan Province 421001, PR China.
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China.
| |
Collapse
|
104
|
Makrutzki-Zlotek K, Escher F, Karadeniz Z, Aleshcheva G, Pietsch H, Küchler K, Schultheiss HP, Heidecker B, Poller W, Landmesser U, Scheibenbogen C, Thevathasan T, Skurk C. FOXO3A acts as immune response modulator in human virus-negative inflammatory cardiomyopathy. HEART (BRITISH CARDIAC SOCIETY) 2023; 109:846-856. [PMID: 36702542 DOI: 10.1136/heartjnl-2022-321732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Inflammatory cardiomyopathy is characterised by inflammatory infiltrates leading to cardiac injury, left ventricular (LV) dilatation and reduced LV ejection fraction (LVEF). Several viral pathogens and autoimmune phenomena may cause cardiac inflammation.The effects of the gain of function FOXO3A single-nucleotide polymorphism (SNP) rs12212067 on inflammation and outcome were studied in a cohort of patients with inflammatory dilated cardiomyopathy (DCMi) in relation to cardiac viral presence. METHODS Distribution of the SNP was determined in virus-positive and virus-negative DCMi patients and in control subjects without myocardial pathology. Baseline and outcome data were compared in 221 virus-negative patients with detection of cardiac inflammation and reduced LVEF according to their carrier status of the SNP. RESULTS Distribution of SNP rs12212067 did not differ between virus-positive (n=22, 19.3%), virus-negative (n=45, 20.4 %) and control patients (n=18, 23.4 %), indicating the absence of susceptibility for viral infection or inflammation per se (p=0.199). Patients in the virus-negative DCMi group were characterised by reduced LVEF 35.5% (95% CI) 33.5 to 37.4) and increased LVEDD (LV end-diastolic diameter) 59.8 mm (95% CI 58.5 to 61.2). Within the group, SNP and non-SNP carriers had similarly impaired LVEF 39.2% (95% CI 34.3% to 44.0%) vs 34.5% (95% CI 32.4 to 36.5), p=0.083, and increased LVEDD 58.9 mm (95% CI 56.3 to 61.5) vs 60.1 mm (95% CI 58.6 to 61.6), p=0.702, respectively. The number of inflammatory infiltrates was not different in both SNP groups at baseline. Outcome after 6 months showed a significant improvement in LVEF and clinical symptoms in SNP rs12212067 carriers 50.9% (95% CI 45.4 to 56.3) versus non-SNP carriers 41.7% (95% CI 39.2 to 44.2), p≤0.01. The improvement in clinical symptoms and LVEF was associated with a significant reduction in cardiac inflammation (ΔCD45RO+ p≤0.05; ΔMac-1+ p≤0.05; ΔLFA-1+ p≤0.01; ΔCD54+ p≤0.01) in the SNP cohort versus non-SNP cohort, respectively. Subgroup analyses identified ΔMac-1+, ΔLFA-1+, ΔCD3+ and Δperforin+ as predictors for improvement in cardiac function in SNP-positive patients. CONCLUSION FOXO3A might act as modulator of the cardiac immune response, diminishing cardiac inflammation and injury in pathogen-negative DCMi.
Collapse
Affiliation(s)
- Kamila Makrutzki-Zlotek
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | - Felicitas Escher
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Virchow-Klinikum, Berlin, Germany.,DZHK, German Center for Cardiovascular Research, Berlin, Germany.,IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Zehra Karadeniz
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | - Ganna Aleshcheva
- IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Heiko Pietsch
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Virchow-Klinikum, Berlin, Germany.,IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Konstanze Küchler
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | | | - Bettina Heidecker
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Poller
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany.,BIH, Berlin Institute of Health at Charité, Berlin, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin - Campus Virchow-Klinikum, Berlin, Germany
| | - Tharusan Thevathasan
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,DZHK, German Center for Cardiovascular Research, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany.,BIH, Berlin Institute of Health at Charité, Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany .,DZHK, German Center for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
105
|
Ribeiro ASF, Zerolo BE, López-Espuela F, Sánchez R, Fernandes VS. Cardiac System during the Aging Process. Aging Dis 2023:AD.2023.0115. [PMID: 37163425 PMCID: PMC10389818 DOI: 10.14336/ad.2023.0115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/15/2023] [Indexed: 05/12/2023] Open
Abstract
The aging process is accompanied by a continuous decline of the cardiac system, disrupting the homeostatic regulation of cells, organs, and systems. Aging increases the prevalence of cardiovascular diseases, thus heart failure and mortality. Understanding the cardiac aging process is of pivotal importance once it allows us to design strategies to prevent age-related cardiac events and increasing the quality of live in the elderly. In this review we provide an overview of the cardiac aging process focus on the following topics: cardiac structural and functional modifications; cellular mechanisms of cardiac dysfunction in the aging; genetics and epigenetics in the development of cardiac diseases; and aging heart and response to the exercise.
Collapse
Affiliation(s)
| | - Blanca Egea Zerolo
- Escuela de Enfermería y Fisioterapia San Juan de Dios. Universidad Pontificia Comillas, Madrid, Spain
| | - Fidel López-Espuela
- Metabolic Bone Diseases Research Group, Nursing and Occupational Therapy College, University of Extremadura, Caceres, Spain
| | - Raúl Sánchez
- Unidad de Cardiopatías Congénitas, Hospital Universitario La Paz, Madrid, Spain
| | - Vítor S Fernandes
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
106
|
Ntsekhe M, Baker JV. Cardiovascular Disease Among Persons Living With HIV: New Insights Into Pathogenesis and Clinical Manifestations in a Global Context. Circulation 2023; 147:83-100. [PMID: 36576956 DOI: 10.1161/circulationaha.122.057443] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Widespread use of contemporary antiretroviral therapy globally has transformed HIV disease into a chronic illness associated with excess risk for disorders of the heart and circulatory system. Current clinical care and research has focused on improving HIV-related cardiovascular disease outcomes, survival, and quality of life. In high-income countries, emphasis on prevention of atherosclerotic coronary artery disease over the past decade, including aggressive management of traditional risk factors and earlier initiation of antiretroviral therapy, has reduced risk for myocardial infarction among persons living with human immunodeficiency virus-1 infection. Still, across the globe, persons living with human immunodeficiency virus-1 infection on effective antiretroviral therapy treatment remain at increased risk for ischemic outcomes such as myocardial infarction and stroke relative to the persons without HIV. Unique features of HIV-related cardiovascular disease, in part, include the pathogenesis of coronary disease characterized by remodeling ectasia and unusual plaque morphology, the relative high proportion of type 2 myocardial infarction events, abnormalities of the aorta such as aneurysms and diffuse aortic inflammation, and HIV cerebrovasculopathy as a contributor to stroke risk. Literature over the past decade has also reflected a shift in the profile and prevalence of HIV-associated heart failure, with a reduced but persistent risk of heart failure with reduced ejection fraction and a growing risk of heart failure with preserved ejection fraction. Cardiac magnetic resonance imaging and autopsy data have emphasized the central importance of intramyocardial fibrosis for the pathogenesis of both heart failure with preserved ejection fraction and the increase in risk of sudden cardiac death. Still, more research is needed to better characterize the underlying mechanisms and clinical phenotype of HIV-associated myocardial disease in the current era. Across the different cardiovascular disease manifestations, a common pathogenic feature is that HIV-associated inflammation working through different mechanisms may amplify underlying pathology because of traditional risk and other host factors. The prevalence and phenotype of individual cardiovascular disease manifestations is ultimately influenced by the degree of injury from HIV disease combined with the profile of underlying cardiometabolic factors, both of which may differ substantially by region globally.
Collapse
Affiliation(s)
- Mpiko Ntsekhe
- Division of Cardiology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa (M.N.)
| | - Jason V Baker
- Division of Infectious Diseases, Hennepin Healthcare Research Institute, Minneapolis, MN (J.V.B.).,Department of Medicine, University of Minnesota, Minneapolis (J.V.B.)
| |
Collapse
|
107
|
Patel M, Yarlagadda H, Upadhyay S, Neupane R, Qureshi U, Raco JD, Jain R, Jain R. Disturbed Sleep is Not Good for the Heart: A Narrative Review. Curr Cardiol Rev 2023; 19:e301122211378. [PMID: 36453501 PMCID: PMC10280991 DOI: 10.2174/1573403x19666221130100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/11/2022] [Accepted: 10/31/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep-related breathing disorders, including obstructive sleep apnea (OSA) and central sleep apnea (CSA), have a major impact on cardiovascular function. It has shown an association with hypertension, coronary artery disease, cardiac arrhythmias, sudden cardiac death, and congestive heart failure (CHF). This review focuses on highlighting the relationship between sleep apnea and CHF. We discuss the underlying pathophysiology, which involves the mechanical, neurohormonal, and inflammatory mechanisms; in addition, the similarities and differentiating clinical features of OSA in patients with CHF and without CHF. We have also discussed several treatment strategies, including weight loss, continuous positive airway pressure (CPAP), supplemental oxygen therapy, theophylline, acetazolamide, mandibular advancement device, and hypoglossal nerve stimulation (HGNS). We conclude that since there are several overlapping clinical features in patients with OSA with Heart Failure (HF) and without HF, early detection and treatment are crucial to decrease the risk of HF, coronary artery disease, and stroke.
Collapse
Affiliation(s)
- Meet Patel
- Department of Internal Medicine, Tianjin Medical University, Tianjin, P.R. China
| | | | | | - Ritesh Neupane
- Department of Internal Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Umer Qureshi
- Penn State College of Medicine, Hershey, PA, USA
| | - Joseph D. Raco
- Department of Internal Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Rahul Jain
- Avalon University School of Medicine, Willemstad, Curaçao
| | - Rohit Jain
- Avalon University School of Medicine, Willemstad, Curaçao
| |
Collapse
|
108
|
Parra-Lucares A, Romero-Hernández E, Villa E, Weitz-Muñoz S, Vizcarra G, Reyes M, Vergara D, Bustamante S, Llancaqueo M, Toro L. New Opportunities in Heart Failure with Preserved Ejection Fraction: From Bench to Bedside… and Back. Biomedicines 2022; 11:70. [PMID: 36672578 PMCID: PMC9856156 DOI: 10.3390/biomedicines11010070] [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: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a growing public health problem in nearly 50% of patients with heart failure. Therefore, research on new strategies for its diagnosis and management has become imperative in recent years. Few drugs have successfully improved clinical outcomes in this population. Therefore, numerous attempts are being made to find new pharmacological interventions that target the main mechanisms responsible for this disease. In recent years, pathological mechanisms such as cardiac fibrosis and inflammation, alterations in calcium handling, NO pathway disturbance, and neurohumoral or mechanic impairment have been evaluated as new pharmacological targets showing promising results in preliminary studies. This review aims to analyze the new strategies and mechanical devices, along with their initial results in pre-clinical and different phases of ongoing clinical trials for HFpEF patients. Understanding new mechanisms to generate interventions will allow us to create methods to prevent the adverse outcomes of this silent pandemic.
Collapse
Affiliation(s)
- Alfredo Parra-Lucares
- Critical Care Unit, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- MD PhD Program, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Esteban Romero-Hernández
- MD PhD Program, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Eduardo Villa
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Sebastián Weitz-Muñoz
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Geovana Vizcarra
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Martín Reyes
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Diego Vergara
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Sergio Bustamante
- Coronary Care Unit, Cardiovascular Department, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Marcelo Llancaqueo
- Coronary Care Unit, Cardiovascular Department, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- Centro de Investigación Clínica Avanzada, Hospital Clínico, Universidad de Chile, Santiago 8380420, Chile
| |
Collapse
|
109
|
Litwin SE, East CA. Assessing clinical and biomarker characteristics to optimize the benefits of sacubitril/valsartan in heart failure. Front Cardiovasc Med 2022; 9:1058998. [PMID: 36620638 PMCID: PMC9815716 DOI: 10.3389/fcvm.2022.1058998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Of the various medical therapies for heart failure (HF), sacubitril/valsartan is a first-in-class angiotensin receptor-neprilysin inhibitor that combines sacubitril, a pro-drug that is further metabolized to the neprilysin inhibitor sacubitrilat, and the angiotensin II type 1 receptor blocker valsartan. Inhibition of neprilysin and blockade of the angiotensin II type 1 receptor with sacubitril/valsartan increases vasoactive peptide levels, increasing vasodilation, natriuresis, and diuresis. Left ventricular ejection fraction (LVEF) is widely used to classify HF, to assist with clinical decision-making, for patient selection in HF clinical trials, and to optimize the benefits of sacubitril/valsartan in HF. However, as HF is a complex syndrome that occurs on a continuum of overlapping and changing phenotypes, patient classification based solely on LVEF becomes problematic. LVEF measurement can be imprecise, have low reproducibility, and often changes over time. LVEF may not accurately reflect inherent disease heterogeneity and complexity, and the addition of alternate criteria to LVEF may improve phenotyping of HF and help guide treatment choices. Sacubitril/valsartan may work, in part, by mechanisms that are not directly related to the LVEF. For example, this drug may exert antifibrotic and neurohumoral modulatory effects through inhibition or activation of several signaling pathways. In this review, we discuss markers of cardiac remodeling, fibrosis, systemic inflammation; activation of neurohormonal pathways, including the natriuretic system and the sympathetic nervous system; the presence of comorbidities; patient characteristics; hemodynamics; and HF signs and symptoms that may all be used to (1) better understand the mechanisms of action of sacubitril/valsartan and (2) help to identify subsets of patients who might benefit from treatment, regardless of LVEF.
Collapse
Affiliation(s)
- Sheldon E. Litwin
- Division of Cardiology, Medical University of South Carolina, Charleston, SC, United States,Ralph H. Johnson Veterans Affairs Health Network, Charleston, SC, United States,*Correspondence: Sheldon E. Litwin,
| | - Cara A. East
- Baylor Soltero Cardiovascular Research Center, Baylor Scott and White Research Institute, Dallas, TX, United States
| |
Collapse
|
110
|
Saavedra-Alvarez A, Pereyra KV, Toledo C, Iturriaga R, Del Rio R. Vascular dysfunction in HFpEF: Potential role in the development, maintenance, and progression of the disease. Front Cardiovasc Med 2022; 9:1070935. [PMID: 36620616 PMCID: PMC9810809 DOI: 10.3389/fcvm.2022.1070935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex, heterogeneous disease characterized by autonomic imbalance, cardiac remodeling, and diastolic dysfunction. One feature that has recently been linked to the pathology is the presence of macrovascular and microvascular dysfunction. Indeed, vascular dysfunction directly affects the functionality of cardiomyocytes, leading to decreased dilatation capacity and increased cell rigidity, which are the outcomes of the progressive decline in myocardial function. The presence of an inflammatory condition in HFpEF produced by an increase in proinflammatory molecules and activation of immune cells (i.e., chronic low-grade inflammation) has been proposed to play a pivotal role in vascular remodeling and endothelial cell death, which may ultimately lead to increased arterial elastance, decreased myocardium perfusion, and decreased oxygen supply to the tissue. Despite this, the precise mechanism linking low-grade inflammation to vascular alterations in the setting of HFpEF is not completely known. However, the enhanced sympathetic vasomotor tone in HFpEF, which may result from inflammatory activation of the sympathetic nervous system, could contribute to orchestrate vascular dysfunction in the setting of HFpEF due to the exquisite sympathetic innervation of both the macro and microvasculature. Accordingly, the present brief review aims to discuss the main mechanisms that may be involved in the macro- and microvascular function impairment in HFpEF and the potential role of the sympathetic nervous system in vascular dysfunction.
Collapse
Affiliation(s)
- Andrea Saavedra-Alvarez
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherine V. Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile,Facultad de la Salud, Centro de Investigación en Fisiología y Medicina de Altura (MedAlt), Universidad de Antofagasta, Antofagasta, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile,Facultad de la Salud, Centro de Investigación en Fisiología y Medicina de Altura (MedAlt), Universidad de Antofagasta, Antofagasta, Chile,*Correspondence: Rodrigo Del Rio
| |
Collapse
|
111
|
Varricchi G, Poto R, Ferrara AL, Gambino G, Marone G, Rengo G, Loffredo S, Bencivenga L. Angiopoietins, vascular endothelial growth factors and secretory phospholipase A 2 in heart failure patients with preserved ejection fraction. Eur J Intern Med 2022; 106:111-119. [PMID: 36280524 DOI: 10.1016/j.ejim.2022.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Heart failure (HF) is a growing public health burden, with high prevalence and mortality rates. A proportion of patients with HF have a normal ventricular ejection fraction (EF), referred to as HF with preserved EF (HFpEF), as opposed to patients with HF with reduced ejection fraction (HFrEF). HFpEF currently accounts for about 50% of all HF patients, and its prevalence is rising. Angiopoietins (ANGPTs), vascular endothelial growth factors (VEGFs) and secretory phospholipases A2 (sPLA2s) are proinflammatory mediators and key regulators of endothelial cells. METHODS The aim of this study was to analyze the plasma concentrations of angiogenic (ANGPT1, ANGPT2, VEGF-A) and lymphangiogenic (VEGF-C, VEGF-D) factors and the plasma activity of sPLA2 in patients with HFpEF and HFrEF compared to healthy controls. RESULTS The concentration of ANGPT1 was reduced in HFrEF compared to HFpEF patients and healthy controls. ANGPT2 levels were increased in both HFrEF and HFpEF subjects compared to controls. The ANGPT2/ANGPT1 ratio was increased in HFrEF patients compared to controls. The concentrations of both VEGF-A and VEGF-C did not differ among the three groups examined. VEGF-D was increased in both HFrEF and HFpEF patients compared to controls. Plasma activity of sPLA2 was increased in HFrEF but not in HFpEF patients compared to controls. CONCLUSIONS Our results indicate that three different classes of proinflammatory regulators of vascular permeability and smoldering inflammation are selectively altered in HFrEF or HFpEF patients. Studies involving larger cohorts of these patients will be necessary to demonstrate the clinical implications of our findings.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit, 82037, Telese, (BN), Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131, Naples, Italy; Gèrontopole de Toulouse, Institut du Vieillissement, CHU de Toulouse, 31000, Toulouse, France
| |
Collapse
|
112
|
Li C, Qin D, Hu J, Yang Y, Hu D, Yu B. Inflamed adipose tissue: A culprit underlying obesity and heart failure with preserved ejection fraction. Front Immunol 2022; 13:947147. [PMID: 36483560 PMCID: PMC9723346 DOI: 10.3389/fimmu.2022.947147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
The incidence of heart failure with preserved ejection fraction is increasing in patients with obesity, diabetes, hypertension, and in the aging population. However, there is a lack of adequate clinical treatment. Patients with obesity-related heart failure with preserved ejection fraction display unique pathophysiological and phenotypic characteristics, suggesting that obesity could be one of its specific phenotypes. There has been an increasing recognition that overnutrition in obesity causes adipose tissue expansion and local and systemic inflammation, which consequently exacerbates cardiac remodeling and leads to the development of obese heart failure with preserved ejection fraction. Furthermore, overnutrition leads to cellular metabolic reprogramming and activates inflammatory signaling cascades in various cardiac cells, thereby promoting maladaptive cardiac remodeling. Growing evidence indicates that the innate immune response pathway from the NLRP3 inflammasome, to interleukin-1 to interleukin-6, is involved in the generation of obesity-related systemic inflammation and heart failure with preserved ejection fraction. This review established the existence of obese heart failure with preserved ejection fraction based on structural and functional changes, elaborated the inflammation mechanisms of obese heart failure with preserved ejection fraction, proposed that NLRP3 inflammasome activation may play an important role in adiposity-induced inflammation, and summarized the potential therapeutic approaches.
Collapse
Affiliation(s)
- Chenyu Li
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Donglu Qin
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Jiarui Hu
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Yang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Die Hu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Bilian Yu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China,*Correspondence: Bilian Yu,
| |
Collapse
|
113
|
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
|
114
|
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]
|
115
|
Song Y, Zhang Y, Zhang X, Hu S, Wang J, Deng G, Zhou Z. AMPK/Sirt1-mediated inflammation is positively correlated with myocardial fibrosis during ageing. Acta Cardiol 2022; 77:826-835. [PMID: 36378531 DOI: 10.1080/00015385.2022.2119667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Cardiovascular disease is the leading cause of death in the world, and it increases dramatically with ageing. The objective of this study was to elucidate age-dependent molecular changes of inflammation and its correlation with the progression of myocardial fibrosis. METHODS Methods: Male SD rats aged 3, 6, 9 and 24 months were used in this study. H&E staining was used to assessed histo-morphological changes in different ages. Masson's trichrome staining was used to evaluate myocardial fibrosis. Immunofluorescence as well as western blot was carried out to detect the expression of vimentin. Real-time PCR was used to detect the level of pro-inflammatory chemokines MCP-1, IL1β, TNFα and IL-6. Western blotting was also carried out to detect p-AMPK, Sirt1, AC-NF-κB expression. RESULTS Myocardial pathological changes and fibrosis are positively correlated with age. Ageing rats showed an enhanced expression of inflammatory factors and the activation of cardiac fibroblasts increases. Meanwhile, the expression of p-AMPK, Sirt1 and downstream AC-NF-κB increased significantly during ageing. Furthermore, the 15-24 months of age in rats is the fastest changing stage of increased inflammation and decreased Sirt1 activity. CONCLUSIONS Ageing is an independent risk factor for the occurrence and development of myocardial fibrosis. During ageing, myocardial fibroblasts are activated, accompanied by an increase in extracellular matrix deposition. The inflammation mediated by AMPK/Sirt1/NF-κB signalling pathway is closely positively correlated with the activation of myocardial fibroblasts and the progression of myocardial fibrosis.
Collapse
Affiliation(s)
- Yanan Song
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| | - Yaqing Zhang
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| | - Xulan Zhang
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| | - Shanshan Hu
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| | - Jin'er Wang
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| | - Gaigai Deng
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| | - Zhiyong Zhou
- Department of Pharmacy, Third-grade Pharmacological Laboratory on Traditional Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Yichang, China.,Department of Pharmacy, Medical College of China Three Gorges University, Yichang, China
| |
Collapse
|
116
|
Safabakhsh S, Al-Shaheen A, Swiggum E, Mielniczuk L, Tremblay-Gravel M, Laksman Z. Arrhythmic Sudden Cardiac Death in Heart Failure With Preserved Ejection Fraction: Mechanisms, Genetics, and Future Directions. CJC Open 2022; 4:959-969. [PMID: 36444369 PMCID: PMC9700220 DOI: 10.1016/j.cjco.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an increasingly recognized disorder. Many clinical trials have failed to demonstrate benefit in patients with HFpEF but have recognized alarming rates of sudden cardiac death (SCD). Genetic testing has become standard in the workup of patients with otherwise unexplained cardiac arrest, but the genetic architecture of HFpEF, and the overlap of a genetic predisposition to HFpEF and arrhythmias, is poorly understood. An understanding of the genetics of HFpEF and related SCD has the potential to redefine and generate novel diagnostic, prognostic, and therapeutic tools. In this review, we examine recent pathophysiological and clinical advancements in our understanding of HFpEF, which reinforce the heterogeneity of the condition. We also discuss data describing SCD events in patients with HFpEF and review the current literature on genetic underpinnings of HFpEF. Mechanisms of arrhythmogenesis which may lead to SCD in this population are also explored. Lastly, we outline several areas of promise for experimentation and clinical trials that have the potential to further advance our understanding of and contribute to improved clinical care of this patient population.
Collapse
Affiliation(s)
- Sina Safabakhsh
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Elizabeth Swiggum
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa Mielniczuk
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Zachary Laksman
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
117
|
Shuaishuai D, Jingyi L, Zhiqiang Z, Guanwei F. Sex differences and related estrogenic effects in heart failure with preserved ejection fraction. Heart Fail Rev 2022:10.1007/s10741-022-10274-2. [PMID: 36190606 DOI: 10.1007/s10741-022-10274-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2022] [Indexed: 11/04/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an essential subtype of heart failure accounting for 40% of the total. However, the related pathological mechanism and drug therapy research have been stagnant for a long time. The direct cause of this dilemma is the heterogeneity of HFpEF. And some researchers believe that there is no common pathway to reach the origin of HFpEF; others argue that there is an unidentified unified pathophysiological process hidden beneath the ice surface. Aside from the debate, a series of clinical studies have shown that hypertension and obesity play a fundamental role in the pathogenesis of HFpEF. These results imply that there may be two parallel pathological processes interweaved in one disease, manifested as multiple coexistent pathological phenomena, like a shadow. Meanwhile, the prevalence of HFpEF in women is higher than in men in any given age group, especially prominent in elderly patients. These pathological processes and epidemiological data reflect gender differences, reminding us to shift our attention to estrogen. This article will review the parallel pathogenesis of HFpEF, and also introduce sex differences and the potential effect of estrogen in this condition below.
Collapse
Affiliation(s)
- Deng Shuaishuai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Lin Jingyi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Zhao Zhiqiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Fan Guanwei
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China. .,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
118
|
Genetzakis E, Gilchrist J, Kassiou M, Figtree GA. Development and clinical translation of P2X7 receptor antagonists: A potential therapeutic target in coronary artery disease? Pharmacol Ther 2022; 237:108228. [DOI: 10.1016/j.pharmthera.2022.108228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/17/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022]
|
119
|
Larson KF, Malik A, Brozovich FV. Aging and Heart Failure with Preserved Ejection Fraction. Compr Physiol 2022; 12:3813-3822. [PMID: 35950652 DOI: 10.1002/cphy.c210035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Heart failure is a clinical syndrome characterized by the inability of the cardiovascular system to provide adequate cardiac output at normal filling pressures. This results in a clinical syndrome characterized by dyspnea, edema, and decreased exertional tolerance. Heart failure with preserved ejection fraction (HFpEF) is an increasingly common disease, and the incidence of HFpEF increases with age. There are a variety of factors which contribute to the development of HFpEF, including the presence of hypertension, diabetes, obesity, and other pro-inflammatory states. These comorbid conditions result in changes at the biochemical and cell signaling level which ultimately lead to a disease with a great deal of phenotypic heterogeneity. In general, the physiologic dysfunction of HFpEF is characterized by vascular stiffness, increased cardiac filling pressures, pulmonary hypertension, and impaired volume management. The normal and abnormal processes associated with aging serve as an accelerant in this process, resulting in the hypothesis that HFpEF represents a form of presbycardia. In this article, we aim to review the processes importance of aging in the development of HFpEF by examining the disease and its causes from the biochemical to physiologic level. © 2022 American Physiological Society. Compr Physiol 12: 1-10, 2022.
Collapse
Affiliation(s)
- Kathryn F Larson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Awais Malik
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Frank V Brozovich
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Physiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
120
|
Budde H, Hassoun R, Mügge A, Kovács Á, Hamdani N. Current Understanding of Molecular Pathophysiology of Heart Failure With Preserved Ejection Fraction. Front Physiol 2022; 13:928232. [PMID: 35874547 PMCID: PMC9301384 DOI: 10.3389/fphys.2022.928232] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/20/2022] [Indexed: 12/15/2022] Open
Abstract
Heart Failure (HF) is the most common cause of hospitalization in the Western societies. HF is a heterogeneous and complex syndrome that may result from any dysfunction of systolic or diastolic capacity. Abnormal diastolic left ventricular function with impaired relaxation and increased diastolic stiffness is characteristic of heart failure with preserved ejection fraction (HFpEF). HFpEF accounts for more than 50% of all cases of HF. The prevalence increases with age: from around 1% for those aged <55 years to >10% in those aged 70 years or over. Nearly 50% of HF patients have HFrEF and the other 50% have HFpEF/HFmrEF, mainly based on studies in hospitalized patients. The ESC Long-Term Registry, in the outpatient setting, reports that 60% have HFrEF, 24% have HFmrEF, and 16% have HFpEF. To some extent, more than 50% of HF patients are female. HFpEF is closely associated with co-morbidities, age, and gender. Epidemiological evidence suggests that HFpEF is highly represented in older obese women and proposed as 'obese female HFpEF phenotype'. While HFrEF phenotype is more a male phenotype. In addition, metabolic abnormalities and hemodynamic perturbations in obese HFpEF patients appear to have a greater impact in women then in men (Sorimachi et al., European J of Heart Fail, 2022, 22). To date, numerous clinical trials of HFpEF treatments have produced disappointing results. This outcome suggests that a "one size fits all" approach to HFpEF may be inappropriate and supports the use of tailored, personalized therapeutic strategies with specific treatments for distinct HFpEF phenotypes. The most important mediators of diastolic stiffness are the cardiomyocytes, endothelial cells, and extracellular matrix (ECM). The complex physiological signal transduction networks that respond to the dual challenges of inflammatory and oxidative stress are major factors that promote the development of HFpEF pathologies. These signalling networks contribute to the development of the diseases. Inhibition and/or attenuation of these signalling networks also delays the onset of disease. In this review, we discuss the molecular mechanisms associated with the physiological responses to inflammation and oxidative stress and emphasize the nature of the contribution of most important cells to the development of HFpEF via increased inflammation and oxidative stress.
Collapse
Affiliation(s)
- Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Andreas Mügge
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Árpád Kovács
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
121
|
Gu S, Mickael C, Kumar R, Lee MH, Sanders L, Kassa B, Harral J, Williams J, Hansen KC, Stenmark KR, Tuder RM, Graham BB. The role of macrophages in right ventricular remodeling in experimental pulmonary hypertension. Pulm Circ 2022; 12:e12105. [PMID: 35874852 PMCID: PMC9297026 DOI: 10.1002/pul2.12105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Right ventricular (RV) failure is the primary cause of death in pulmonary hypertension (PH), but the mechanisms of RV failure are not well understood. We hypothesized macrophages in the RV contribute to the RV response in PH. We induced PH in mice with hypoxia (FiO2 10%) and Schistosoma mansoni exposure, and in rats with SU5416-hypoxia. We quantified cardiac macrophages in mice using flow cytometry. Parabiosis between congenic CD45.1/.2 mice or Cx3cr1-green fluorescent protein and wild-type mice was used to quantify circulation-derived macrophages in experimental PH conditions. We administered clodronate liposomes to Sugen hypoxia (SU-Hx) exposed rats to deplete macrophages and evaluated the effect on the extracellular matrix (ECM) and capillary network in the RV. In hypoxia exposed mice, the overall number of macrophages did not significantly change but two macrophage subpopulations increased. Parabiosis identified populations of RV macrophages that at steady state is derived from the circulation, with one subpopulation that significantly increased with PH stimuli. Clodronate treatment of SU-Hx rats resulted in a change in the RV ECM, without altering the RV vasculature, and correlated with improved RV function. Populations of RV macrophages increase and contribute to RV remodeling in PH, including through regulation of the RV ECM.
Collapse
Affiliation(s)
- Sue Gu
- Department of Medicine, Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Cardiovascular Pulmonary Research LabUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Claudia Mickael
- Department of Medicine, Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of Medicine, Program in Translational Lung ResearchUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Rahul Kumar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Michael H. Lee
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Linda Sanders
- Department of Medicine, Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of Medicine, Program in Translational Lung ResearchUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Biruk Kassa
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Julie Harral
- Cardiovascular Pulmonary Research LabUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Jason Williams
- Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Kirk C. Hansen
- Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research LabUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Rubin M. Tuder
- Department of Medicine, Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of Medicine, Program in Translational Lung ResearchUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Brian B. Graham
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
122
|
Zhang X, Zhang Y, Sun A, Ge J. The effects of nicotinamide adenine dinucleotide in cardiovascular diseases: Molecular mechanisms, roles and therapeutic potential. Genes Dis 2022; 9:959-972. [PMID: 35685463 PMCID: PMC9170600 DOI: 10.1016/j.gendis.2021.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 12/23/2022] Open
Abstract
Recently, cardiovascular diseases (CVDs) were identified as the leading cause of mortality, imposing a heavy burden on health care systems and the social economy. Nicotinamide adenine dinucleotide (NAD+), as a pivotal co-substrate for a range of different enzymes, is involved in many signal transduction pathways activated in CVDs. Emerging evidence has shown that NAD+ can exert remediating effects on CVDs by regulating metabolism, maintaining redox homeostasis and modulating the immune response. In fact, NAD+ might delay ageing through sirtuin and non-sirtuin pathways and thus contribute to interventions for age-related diseases such as CVDs. Considering that robust clinical studies of NAD+ are ongoing, we discuss current challenges and the future translational potential of NAD+ based on existing studies and our understanding. Despite some remaining gaps in its clinical application, NAD+ has been shown to have broad prospects and pan-effects, making it a suitable prophylactic drug for CVDs.
Collapse
Affiliation(s)
- Xiaokai Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, PR China
| | - Yang Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, PR China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, PR China.,Institute of Biomedical Sciences, Fudan University, Shanghai 200032, PR China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, PR China.,Institute of Biomedical Sciences, Fudan University, Shanghai 200032, PR China
| |
Collapse
|
123
|
Dehe L, Mousa SA, Shaqura M, Shakibaei M, Schäfer M, Treskatsch S. Naltrexone-Induced Cardiac Function Improvement is Associated With an Attenuated Inflammatory Response and Lipid Perioxidation in Volume Overloaded Rats. Front Pharmacol 2022; 13:873169. [PMID: 35847039 PMCID: PMC9280420 DOI: 10.3389/fphar.2022.873169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
In previous studies, upregulation of myocardial opioid receptors as well as the precursors of their endogenous ligands were detected in the failing heart due to chronic volume overload. Moreover, opioid receptor blockade by naltrexone improved left ventricular function. In parallel, inflammatory processes through cytokines have been confirmed to play an important role in the pathogenesis of different forms of heart failure. Thus, the present study examined the systemic and myocardial inflammatory response to chronic volume overload and its modulation by chronic naltrexone therapy. Chronic volume overload was induced in male Wistar rats by applying an infrarenal aortocaval fistula (ACF) for 28 days during which the selective opioid receptor antagonist naltrexone (n = 6) or vehicle (n = 6) were administered via a subcutaneously implanted Alzet minipump. The ultrastructural, morphometric and hemodynamic characterization of ACF animals were performed using an intraventricular conductance catheter in vivo and electron microscopy in vitro. Co-localization of mu-, delta- and kappa-opioid receptor subtypes (MOR, DOR, and KOR respectively) with the voltage gated L-type Ca2+ channel (Cav1.2), the ryanodine receptor (RyR), and mitochondria in cardiomyocytes as well as IL-6, IL-12, TNF-alpha, and Malondialdehyde (MDA) were determined using double immunofluorescence confocal microscopy, RT-PCR and ELISA, respectively. In rat left ventricular myocardium, three opioid receptor subtypes MOR, DOR, and KOR colocalized with Cav1.2, RyR and mitochondria suggesting a modulatory role of the excitation-contraction coupling. In rats with ACF-induced volume overload, signs of heart failure and myocardial ultrastructural damage, chronic naltrexone therapy improved cardiac function and reversed the systemic and myocardial inflammatory cytokine expression as well as lipid peroxidation. In conclusion, antagonism of the cardiodepressive effects of the myocardial opioid system does not only improve left ventricular function but also blunts the inflammatory response and lipid peroxidation.
Collapse
Affiliation(s)
- Lukas Dehe
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Shaaban A. Mousa
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
- *Correspondence: Shaaban A. Mousa,
| | - Mohammed Shaqura
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilians-Universität München, München, Germany
| | - Michael Schäfer
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Sascha Treskatsch
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
124
|
Zhang M, Shu H, Chen C, He Z, Zhou Z, Wang DW. Epoxyeicosatrienoic acid: A potential therapeutic target of heart failure with preserved ejection fraction. Biomed Pharmacother 2022; 153:113326. [PMID: 35759865 DOI: 10.1016/j.biopha.2022.113326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/02/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) reduces the quality of life, costs substantial medical resources, and has a high mortality. However, we lack an effective therapy for HFpEF due to our limited knowledge of its mechanism. Therefore, it is crucial to explore novel therapeutics, such as those with endogenous protective roles, and seek new targeted therapies. Epoxyeicosatrienoic acids (EETs) are endogenous bioactive metabolites of arachidonic acids produced by cytochrome P450 (CYP) epoxygenases. EETs can function as endogenous cardioprotective factors with potent inhibitory roles in inflammation, endothelial dysfunction, cardiac remodeling, and fibrosis, which are the fundamental mechanisms of HFpEF. This suggests that EETs have the potential function to protect against HFpEF. Therefore, we present an overview of the ever-expanding world of EETs and how they might help alleviate the pathophysiology underlying HFpEF to provide new insights for research in this field.
Collapse
Affiliation(s)
- Min Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Zuowen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Zhou Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
125
|
Delacroix C, Hulot JS. [Integrins in cardiac fibrosis]. Med Sci (Paris) 2022; 38:438-444. [PMID: 35608466 DOI: 10.1051/medsci/2022055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
For the last 20 years, integrins have been a therapeutic target of interest in the treatment of fibrotic diseases, particularly regarding the integrins of the αV family. Initially developed as anti-cancer drugs but with modest benefits, inhibitors of integrins (such as the anti-αV cilengitide) have shown interesting anti-fibrotic effects in different organs including the heart. Cardiac fibrosis is defined as an accumulation of stiff extracellular matrix in the myocardium, and ultimately leads to heart failure, one of the leading causes of mortality worldwide. Understanding the determinants of cardiac fibrosis and the involvement of integrins is a major matter of public health. This review presents the current knowledge on the different types of cardiac fibrosis and their etiologies, and report on first data supporting specific integrin inhibition therapy as a novel anti-fibrotic strategy, in particular to treat cardiac fibrosis.
Collapse
Affiliation(s)
- Clément Delacroix
- Paris Centre de recherche cardiovasculaire (PARCC), Inserm U.970, 56 rue Leblanc, 75015 Paris, France
| | - Jean-Sébastien Hulot
- Paris Centre de recherche cardiovasculaire (PARCC), Inserm U.970, 56 rue Leblanc, 75015 Paris, France
| |
Collapse
|
126
|
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: 23] [Impact Index Per Article: 7.7] [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
|
127
|
Jin X, Hung CL, Tay WT, Soon D, Sim D, Sung KT, Loh SY, Lee S, Jaufeerally F, Ling LH, Richards AM, van Melle JP, Voors AA, Lam CSP. Epicardial Adipose Tissue related to Left Atrial and Ventricular Function in Heart Failure with Preserved (HFpEF) versus Reduced and Mildly Reduced Ejection Fraction (HFrEF/HFmrEF). Eur J Heart Fail 2022; 24:1346-1356. [PMID: 35475591 DOI: 10.1002/ejhf.2513] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/11/2022] [Accepted: 04/14/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Different associations between epicardial adipose tissue (EAT) and cardiac function have been suggested in patients with heart failure with preserved (HFpEF) versus reduced and mildly reduced ejection fraction (HFrEF/HFmrEF). However, few studies have directly compared the association between EAT and left atrial (LA) and ventricular (LV) function in patients with HFpEF and HFrEF/HFmrEF. METHODS We studied EAT thickness using transthoracic echocardiography in a multicenter cohort of 149 community-dwelling controls without HF, 99 patients with HFpEF, and 366 patients with HFrEF/HFmrEF. EAT thickness was averaged from parasternal long-axis and short-axis views, respectively, and off-line speckle tracking analysis was performed to quantify LA and LV function. Data were validated in an independent cohort of 626 controls, 243 patients with HFpEF, and 180 patients with HFrEF/HFmrEF. For LV function, LV global longitudinal strain (GLS) was measured in both derivation and validation cohorts. For the LA function, LAGLS at reservoir, contractile and conduit phase were measured in the derivation cohort, and only LAGLS at reservoir phase was measured in the validation cohort. RESULTS In the derivation cohort, EAT thickness was lower in HFrEF/HFmrEF (7.3±2.5mm) compared to HFpEF (8.3±2.6mm, p<0.05) and controls (7.9±1.8mm, p<0.05). Greater EAT thickness was associated with better LV and contractile LA function in HFrEF/HFmrEF, but not in HFpEF (p for interaction < 0.05). These findings were confirmed in the validation cohort, where EAT thickness was lower in HFrEF/HFmrEF (6.7±1.4mm) compared to HFpEF (9.6±2.8mm; p<0.05) and controls (7.7±2.3mm; p<0.05). Greater EAT thickness was associated with better LV and reservoir LA function in patients with HFrEF/HFmrEF but worse LV and reservoir LA function in patients with HFpEF (p for interaction <0.05). Thickened EAT (EAT thickness >10mm) was associated with LA dysfunction (LAGLS at reservoir phase<23%) in HFpEF, but not in HFrEF/HFmrEF. CONCLUSION EAT thickness is greater in patient with HFpEF than HFrEF/HFmrEF. Increased EAT thickness is associated with worse LA and LV function in HFpEF but the opposite in HFrEF/HFmrEF. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xuanyi Jin
- National Heart Centre Singapore, Singapore, Singapore.,Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Chung-Lieh Hung
- Division of Cardiology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan.,Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Wan Ting Tay
- National Heart Centre Singapore, Singapore, Singapore
| | | | - David Sim
- National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Kuo-Tzu Sung
- Division of Cardiology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan.,Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, 25245, Taiwan
| | | | | | - Fazlur Jaufeerally
- Duke-NUS Medical School, Singapore, Singapore.,Singapore General Hospital, Singapore
| | - Lieng Hsi Ling
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore.,Department of Cardiology, National University Heart Centre, Singapore.,Cardiovascular Research Institute, National University Health System, Singapore
| | - A Mark Richards
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore.,Department of Cardiology, National University Heart Centre, Singapore.,Cardiovascular Research Institute, National University Health System, Singapore.,Christchurch Heart Institute, University of Otago, New Zealand
| | - Joost P van Melle
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Carolyn S P Lam
- National Heart Centre Singapore, Singapore, Singapore.,Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
128
|
Chen L, Wang W, Peng X, Liu L, Zhang A, Li X, Ma K, Wang L. Alpha1-adrenoceptors activate NLRP3 inflammasome through downregulation of Kir2.1 in cardiac inflammation. Exp Physiol 2022; 107:589-600. [PMID: 35363405 DOI: 10.1113/ep090243] [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: 11/25/2021] [Accepted: 03/29/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? The mechanism of cardiac inflammation induced by α 1 -AR stimulation by NLRP3 inflammasome activation is unclear. What is the main finding and its importance? In the mechanism of cardiac inflammation induced by α1 -AR overreaction, Kir2.1 exerts cardioprotective and anti-inflammatory effects by inhibiting the activation of NLRP3 Inflammasome. ABSTRACT Overstimulated sympathetic nerves in cardiovascular diseases can lead to impaired cardiomyocyte function and potential heart failure, which activates not only β-AR but also α1 -AR. A previous report indicated that NLRP3 inflammasome activation is involved in cardiac inflammation induced by the α1 -AR agonist phenylephrine, but the mechanism is still unknown. Here, we aimed to study whether Kir2.1 is involved in cardiac inflammation caused by phenylephrine. The results from in vitro experiments showed that phenylephrine upregulated the expression levels of NLRP3, Caspase-1, IL-18, and IL-1β and downregulated the expression level of Kir2.1 in H9C2 cells. The Kir2.1 agonist zacopride downregulated the expression of NLRP3, Caspase-1, IL-1β and IL-18, and the Kir2.1 inhibitor ML133 upregulated the expression of these genes. To further explore the mechanism, we found that zacopride downregulated the protein expression level of p-p65 and that ML133 upregulated it. Moreover, the NF-κB signaling pathway inhibitor curcumenol reversed the expression of NLRP3 inflammasomes caused by phenylephrine in H9C2 cells. In vivo experiments, the protein expression level of Kir2.1 in the phenylephrine group was significantly decreased, and the activation of Kir2.1 by zacopride reduced cardiac inflammation. In short, Kir2.1 is related to α1 -AR overactivation, which induces cardiac inflammation, through the NF-κB signaling pathway, and activating Kir2.1 can downregulate NLRP3 inflammation and exert cardioprotective effects by zacopride. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ling Chen
- The 3rd Department of Cardiology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Wenbo Wang
- The 3rd Department of Cardiology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Xiangyang Peng
- The 3rd Department of Cardiology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Luqian Liu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Aimei Zhang
- The 3rd Department of Cardiology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| | - Li Wang
- The 3rd Department of Cardiology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence, Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832000, China
| |
Collapse
|
129
|
Sunaga A, Hikoso S, Tamaki S, Seo M, Yano M, Hayashi T, Nakagawa A, Nakagawa Y, Kurakami H, Yamada T, Kitamura T, Sato T, Oeun B, Kida H, Sotomi Y, Dohi T, Okada K, Mizuno H, Nakatani D, Yamada T, Yasumura Y, Sakata Y, Seo M, Watanabe T, Yamada T, Hayashi T, Higuchi Y, Masuda M, Asai M, Mano T, Fuji H, Masuda D, Tamaki S, Shutta R, Yamashita S, Sairyo M, Nakagawa Y, Abe H, Ueda Y, Matsumura Y, Nagai K, Yano M, Nishino M, Tanouchi J, Arita Y, Ogasawara N, Ishizu T, Ichikawa M, Takano Y, Rin E, Shinoda Y, Tachibana K, Hoshida S, Izumi M, Yamamoto H, Kato H, Nakatani K, Yasuga Y, Nishio M, Hirooka K, Yoshimura T, Yasuoka Y, Tani A, Okumoto Y, Makino Y, Onishi T, Iwakura K, Kijima Y, Kitao T, Kanai H, Fujita M, Harada K, Kumada M, Nakagawa O, Araki R, Yamada T, Nakagawa A, Yasumura Y, Sato T, Sunaga A, Oeun B, Kida H, Sotomi Y, Dohi T, Nakamoto K, Okada K, Sera F, Kioka H, Ohtani T, Takeda T, Nakatani D, Mizuno H, Hikoso S, Sakata Y. Association between prognosis and the use of angiotensin‐converting enzyme inhibitors and/or angiotensin II receptor blockers in frail patients with heart failure with preserved ejection fraction. ESC Heart Fail 2022. [PMCID: PMC9065837 DOI: 10.1002/ehf2.13873] [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] [Indexed: 12/01/2022] Open
Abstract
Aims The effectiveness of angiotensin‐converting enzyme inhibitors (ACE‐I) and angiotensin II receptor blockers (ARB) has not been demonstrated in patients with heart failure with preserved ejection fraction (HFpEF). We recently reported significant interaction between the use of ACE‐I and/or ARB (ACE‐I/ARB) and frailty on prognosis in patients with HFpEF. In the present study, we examined the association between ACE‐I/ARB and prognosis in patients with HFpEF stratified by the presence or absence of frailty. Methods and results We examined the association between the use of ACE‐I/ARB and prognosis according to the presence [Clinical Frailty Scale (CFS) ≥ 5] or absence (CFS ≤ 4) of frailty in patients with HFpEF in a post hoc analysis of registry data. Primary endpoint was the composite of all‐cause mortality and heart failure admission. Secondary endpoints were all‐cause mortality and heart failure admission. Of 1059 patients, median age was 83 years and 45% were male. Kaplan–Meier analysis showed that the risk of composite endpoint (log‐rank P = 0.001) and all‐cause death (log‐rank P = 0.005) in patients with ACE‐I/ARB was lower in those with CFS ≥ 5, but similar between patients with and without ACE‐I/ARB in patients with CFS ≤ 4 (composite endpoint: log‐rank P = 0.830; all‐cause death: log‐rank P = 0.192). In a multivariable Cox proportional hazards model, use of ACE‐I/ARB was significantly associated with lower risk of the composite endpoint [hazard ratio (HR) = 0.52, 95% confidence interval (CI) = 0.33–0.83, P = 0.005] and heart failure admission (HR = 0.45, 95% CI = 0.25–0.83, P = 0.010) in patients with CFS ≥ 5, but not in patients with CFS ≤ 4 (composite endpoint: HR = 1.41, 95% CI = 0.99–2.02, P = 0.059; heart failure admission: HR = 1.43, 95% CI = 0.94–2.18, P = 0.091). The association between ACE‐I or ARB and prognosis did not significantly differ by CFS (CFS ≤ 4: log‐rank P = 0.562; CFS ≥ 5: log‐rank P = 0.100, for with ACE‐I vs. ARB, respectively). Adjusted HRs for CFS 1–4 were higher than 1.0 but were <1.0 at CFS 5. Conclusions In patients with HFpEF, use of ACE‐I/ARB was associated with better prognosis in patients with frailty as assessed with the CFS, but not in those without frailty.
Collapse
Affiliation(s)
- Akihiro Sunaga
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Shungo Hikoso
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Shunsuke Tamaki
- Department of Cardiology Rinku General Medical Center Osaka Japan
| | - Masahiro Seo
- Division of Cardiology Osaka General Medical Center Osaka Japan
| | | | | | - Akito Nakagawa
- Division of Cardiology Amagasaki Chuo Hospital Amagasaki Japan
- Department of Medical Informatics Osaka University Graduate School of Medicine Suita Japan
| | - Yusuke Nakagawa
- Division of Cardiology Kawanishi City Hospital Kawanishi Japan
| | - Hiroyuki Kurakami
- Department of Medical Innovation Osaka University Hospital Suita Japan
| | - Tomomi Yamada
- Department of Medical Innovation Osaka University Hospital Suita Japan
| | - Tetsuhisa Kitamura
- Department of Social and Environmental Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Taiki Sato
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Bolrathanak Oeun
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Hirota Kida
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Yohei Sotomi
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Tomoharu Dohi
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Katsuki Okada
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
- Department of Transformative System for Medical Information Osaka University Graduate School of Medicine Suita Japan
| | - Hiroya Mizuno
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Daisaku Nakatani
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Takahisa Yamada
- Division of Cardiology Osaka General Medical Center Osaka Japan
| | - Yoshio Yasumura
- Division of Cardiology Amagasaki Chuo Hospital Amagasaki Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Dyck JRB, Sossalla S, Hamdani N, Coronel R, Weber NC, Light PE, Zuurbier CJ. Cardiac mechanisms of the beneficial effects of SGLT2 inhibitors in heart failure: Evidence for potential off-target effects. J Mol Cell Cardiol 2022; 167:17-31. [PMID: 35331696 DOI: 10.1016/j.yjmcc.2022.03.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
Abstract
Sodium glucose cotransporter 2 inhibitors (SGLT2i) constitute a promising drug treatment for heart failure patients with either preserved or reduced ejection fraction. Whereas SGLT2i were originally developed to target SGLT2 in the kidney to facilitate glucosuria in diabetic patients, it is becoming increasingly clear that these drugs also have important effects outside of the kidney. In this review we summarize the literature on cardiac effects of SGLT2i, focussing on pro-inflammatory and oxidative stress processes, ion transport mechanisms controlling sodium and calcium homeostasis and metabolic/mitochondrial pathways. These mechanisms are particularly important as disturbances in these pathways result in endothelial dysfunction, diastolic dysfunction, cardiac stiffness, and cardiac arrhythmias that together contribute to heart failure. We review the findings that support the concept that SGLT2i directly and beneficially interfere with inflammation, oxidative stress, ionic homeostasis, and metabolism within the cardiac cell. However, given the very low levels of SGLT2 in cardiac cells, the evidence suggests that SGLT2-independent effects of this class of drugs likely occurs via off-target effects in the myocardium. Thus, while there is still much to be understood about the various factors which determine how SGLT2i affect cardiac cells, much of the research clearly demonstrates that direct cardiac effects of these SGLT2i exist, albeit mediated via SGLT2-independent pathways, and these pathways may play a role in explaining the beneficial effects of SGLT2 inhibitors in heart failure.
Collapse
Affiliation(s)
- Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany; Klinik für Kardiologie und Pneumologie, Georg-August-Universität Goettingen, DZHK (German Centre for Cardiovascular Research), Robert-Koch Str. 40, D-37075 Goettingen, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital Ruhr University Bochum, Bochum, Germany
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Amsterdam, the Netherlands
| | - Nina C Weber
- Department of Anesthesiology - L.E.I.C.A, Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Amsterdam, the Netherlands
| | - Peter E Light
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Coert J Zuurbier
- Department of Anesthesiology - L.E.I.C.A, Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Amsterdam, the Netherlands.
| |
Collapse
|
131
|
Fu B, Wei X, Lin Y, Chen J, Yu D. Pathophysiologic Basis and Diagnostic Approaches for Ischemia With Non-obstructive Coronary Arteries: A Literature Review. Front Cardiovasc Med 2022; 9:731059. [PMID: 35369287 PMCID: PMC8968033 DOI: 10.3389/fcvm.2022.731059] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 01/31/2022] [Indexed: 02/05/2023] Open
Abstract
Ischemia with non-obstructive coronary arteries (INOCA) has gained increasing attention due to its high prevalence, atypical clinical presentations, difficult diagnostic procedures, and poor prognosis. There are two endotypes of INOCA-one is coronary microvascular dysfunction and the other is vasospastic angina. Diagnosis of INOCA lies in evaluating coronary flow reserve, microcirculatory resistance, and vasoreactivity, which is usually obtained via invasive coronary interventional techniques. Non-invasive diagnostic approaches such as echocardiography, single-photon emission computed tomography, cardiac positron emission tomography, and cardiac magnetic resonance imaging are also valuable for assessing coronary blood flow. Some new techniques (e.g., continuous thermodilution and angiography-derived quantitative flow reserve) have been investigated to assist the diagnosis of INOCA. In this review, we aimed to discuss the pathophysiologic basis and contemporary and novel diagnostic approaches for INOCA, to construct a better understanding of INOCA evaluation.
Collapse
Affiliation(s)
- Bingqi Fu
- Shantou University Medical College, Shantou, China
- Division of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xuebiao Wei
- Division of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Division of Geriatric Intensive Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yingwen Lin
- Shantou University Medical College, Shantou, China
- Division of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiyan Chen
- Division of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Danqing Yu
- Division of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
132
|
Abstract
Obesity has reached epidemic proportions and is a major contributor to insulin resistance (IR) and type 2 diabetes (T2D). Importantly, IR and T2D substantially increase the risk of cardiovascular (CV) disease. Although there are successful approaches to maintain glycemic control, there continue to be increased CV morbidity and mortality associated with metabolic disease. Therefore, there is an urgent need to understand the cellular and molecular processes that underlie cardiometabolic changes that occur during obesity so that optimal medical therapies can be designed to attenuate or prevent the sequelae of this disease. The vascular endothelium is in constant contact with the circulating milieu; thus, it is not surprising that obesity-driven elevations in lipids, glucose, and proinflammatory mediators induce endothelial dysfunction, vascular inflammation, and vascular remodeling in all segments of the vasculature. As cardiometabolic disease progresses, so do pathological changes in the entire vascular network, which can feed forward to exacerbate disease progression. Recent cellular and molecular data have implicated the vasculature as an initiating and instigating factor in the development of several cardiometabolic diseases. This Review discusses these findings in the context of atherosclerosis, IR and T2D, and heart failure with preserved ejection fraction. In addition, novel strategies to therapeutically target the vasculature to lessen cardiometabolic disease burden are introduced.
Collapse
|
133
|
Feng G, Bajpai G, Ma P, Koenig A, Bredemeyer A, Lokshina I, Lai L, Förster I, Leuschner F, Kreisel D, Lavine KJ. CCL17 Aggravates Myocardial Injury by Suppressing Recruitment of Regulatory T Cells. Circulation 2022; 145:765-782. [PMID: 35113652 PMCID: PMC8957788 DOI: 10.1161/circulationaha.121.055888] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Recent studies have established that CCR2 (C-C chemokine receptor type 2) marks proinflammatory subsets of monocytes, macrophages, and dendritic cells that contribute to adverse left ventricle (LV) remodeling and heart failure progression. Elucidation of the effector mechanisms that mediate adverse effects of CCR2+ monocytes, macrophages, and dendritic cells will yield important insights into therapeutic strategies to suppress myocardial inflammation. METHODS We used mouse models of reperfused myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation to investigate CCL17 (C-C chemokine ligand 17). We used Ccl17 knockout mice, flow cytometry, RNA sequencing, biochemical assays, cell trafficking studies, and in vivo cell depletion to identify the cell types that generate CCL17, define signaling pathways that controlled its expression, delineate the functional importance of CCL17 in adverse LV remodeling and heart failure progression, and determine the mechanistic basis by which CCL17 exerts its effects. RESULTS We demonstrated that CCL17 is expressed in CCR2+ macrophages and cluster of differentiation 11b+ conventional dendritic cells after myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation. We clarified the transcriptional signature of CCL17+ macrophages and dendritic cells and identified granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling as a key regulator of CCL17 expression through cooperative activation of STAT5 (signal transducer and activator of transcription 5) and canonical NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling. Ccl17 deletion resulted in reduced LV remodeling, decreased myocardial fibrosis and cardiomyocyte hypertrophy, and improved LV systolic function after myocardial infarction and angiotensin II and phenylephrine infusion. We observed increased abundance of regulatory T cells (Tregs) in the myocardium of injured Ccl17 knockout mice. CCL17 inhibited Treg recruitment through biased activation of CCR4. CCL17 activated Gq signaling and CCL22 (C-C chemokine ligand 22) activated both Gq and ARRB (β-arrestin) signaling downstream of CCR4. CCL17 competitively inhibited CCL22 stimulated ARRB signaling and Treg migration. We provide evidence that Tregs mediated the protective effects of Ccl17 deletion on myocardial inflammation and adverse LV remodeling. CONCLUSIONS These findings identify CCL17 as a proinflammatory mediator of CCR2+ macrophages and dendritic cells and suggest that inhibition of CCL17 may serve as an effective strategy to promote Treg recruitment and suppress myocardial inflammation.
Collapse
Affiliation(s)
- Guoshuai Feng
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Geetika Bajpai
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Pan Ma
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Andrew Koenig
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Inessa Lokshina
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Lulu Lai
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | | | - Florian Leuschner
- LIMES Institute, University of Bonn, Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany (F.L.)
| | - Daniel Kreisel
- Department of Surgery, Washington University, Saint Louis, Missouri, USA (D.K.)
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA (D.K., K.L.)
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA (D.K., K.L.)
- Department of Developmental Biology, Washington University, Saint Louis, Missouri, USA (K.L.)
| |
Collapse
|
134
|
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: 28] [Impact Index Per Article: 9.3] [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
|
135
|
Rocca A, van Heeswijk RB, Richiardi J, Meyer P, Hullin R. The Cardiomyocyte in Heart Failure with Preserved Ejection Fraction-Victim of Its Environment? Cells 2022; 11:867. [PMID: 35269489 PMCID: PMC8909081 DOI: 10.3390/cells11050867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/01/2022] [Indexed: 12/07/2022] Open
Abstract
Heart failure (HF) with preserved left ventricular ejection fraction (HFpEF) is becoming the predominant form of HF. However, medical therapy that improves cardiovascular outcome in HF patients with almost normal and normal systolic left ventricular function, but diastolic dysfunction is missing. The cause of this unmet need is incomplete understanding of HFpEF pathophysiology, the heterogeneity of the patient population, and poor matching of therapeutic mechanisms and primary pathophysiological processes. Recently, animal models improved understanding of the pathophysiological role of highly prevalent and often concomitantly presenting comorbidity in HFpEF patients. Evidence from these animal models provide first insight into cellular pathophysiology not considered so far in HFpEF disease, promising that improved understanding may provide new therapeutical targets. This review merges observation from animal models and human HFpEF disease with the intention to converge cardiomyocytes pathophysiological aspects and clinical knowledge.
Collapse
Affiliation(s)
- Angela Rocca
- Department of Cardiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| | - Ruud B. van Heeswijk
- Department of Diagnostic and Interventional Radiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (R.B.v.H.); (J.R.)
| | - Jonas Richiardi
- Department of Diagnostic and Interventional Radiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (R.B.v.H.); (J.R.)
| | - Philippe Meyer
- Cardiology Service, Department of Medical Specialties, Faculty of Science, Geneva University Hospital, University of Geneva, 1205 Geneva, Switzerland;
| | - Roger Hullin
- Department of Cardiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| |
Collapse
|
136
|
Nona P, Russell C. Cardio-Rheumatology: Prevention of Cardiovascular Disease in Inflammatory Disorders. Med Clin North Am 2022; 106:349-363. [PMID: 35227435 DOI: 10.1016/j.mcna.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inflammation plays a well-established role in the development and progression of atherosclerosis. Individuals exposed to chronic inflammation are at an increased risk of developing cardiovascular disease, including coronary artery disease and heart failure, independent of associated traditional risk factors. Traditional risk assessment tools and calculators underestimate the true cardiac risk in this population. In addition to this, there is a lack of awareness on the association between inflammation and cardiovascular disease. These factors lead to undertreatment in terms of preventive cardiac care in patients with chronic inflammatory disease.
Collapse
Affiliation(s)
- Paul Nona
- Department of Internal Medicine, Division of Cardiology, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - Cori Russell
- Department of Internal Medicine, Division of Cardiology, 2799 West Grand Boulevard, Detroit, MI 48202, USA.
| |
Collapse
|
137
|
Schiattarella GG, Alcaide P, Condorelli G, Gillette TG, Heymans S, Jones EAV, Kallikourdis M, Lichtman A, Marelli-Berg F, Shah S, Thorp EB, Hill JA. Immunometabolic Mechanisms of Heart Failure with Preserved Ejection Fraction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:211-222. [PMID: 35755006 PMCID: PMC9229992 DOI: 10.1038/s44161-022-00032-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing in prevalence worldwide, already accounting for at least half of all heart failure (HF). As most patients with HFpEF are obese with metabolic syndrome, metabolic stress has been implicated in syndrome pathogenesis. Recently, compelling evidence for bidirectional crosstalk between metabolic stress and chronic inflammation has emerged, and alterations in systemic and cardiac immune responses are held to participate in HFpEF pathophysiology. Indeed, based on both preclinical and clinical evidence, comorbidity-driven systemic inflammation, coupled with metabolic stress, have been implicated together in HFpEF pathogenesis. As metabolic alterations impact immune function(s) in HFpEF, major changes in immune cell metabolism are also recognized in HFpEF and in HFpEF-predisposing conditions. Both arms of immunity - innate and adaptive - are implicated in the cardiomyocyte response in HFpEF. Indeed, we submit that crosstalk among adipose tissue, the immune system, and the heart represents a critical component of HFpEF pathobiology. Here, we review recent evidence in support of immunometabolic mechanisms as drivers of HFpEF pathogenesis, discuss pivotal biological mechanisms underlying the syndrome, and highlight questions requiring additional inquiry.
Collapse
Affiliation(s)
- Gabriele G. Schiattarella
- Center for Cardiovascular Research (CCR), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Gianluigi Condorelli
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Cardio Center, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Elizabeth A. V. Jones
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marinos Kallikourdis
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Adaptive Immunity Lab, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Andrew Lichtman
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sanjiv Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Edward B. Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
138
|
Weerts J, Mourmans SGJ, Barandiarán Aizpurua A, Schroen BLM, Knackstedt C, Eringa E, Houben AJHM, van Empel VPM. The Role of Systemic Microvascular Dysfunction in Heart Failure with Preserved Ejection Fraction. Biomolecules 2022; 12:biom12020278. [PMID: 35204779 PMCID: PMC8961612 DOI: 10.3390/biom12020278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a condition with increasing incidence, leading to a health care problem of epidemic proportions for which no curative treatments exist. Consequently, an urge exists to better understand the pathophysiology of HFpEF. Accumulating evidence suggests a key pathophysiological role for coronary microvascular dysfunction (MVD), with an underlying mechanism of low-grade pro-inflammatory state caused by systemic comorbidities. The systemic entity of comorbidities and inflammation in HFpEF imply that patients develop HFpEF due to systemic mechanisms causing coronary MVD, or systemic MVD. The absence or presence of peripheral MVD in HFpEF would reflect HFpEF being predominantly a cardiac or a systemic disease. Here, we will review the current state of the art of cardiac and systemic microvascular dysfunction in HFpEF (Graphical Abstract), resulting in future perspectives on new diagnostic modalities and therapeutic strategies.
Collapse
Affiliation(s)
- Jerremy Weerts
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
- Correspondence: ; Tel.: +31-43-387-7097
| | - Sanne G. J. Mourmans
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Arantxa Barandiarán Aizpurua
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Blanche L. M. Schroen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Christian Knackstedt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Etto Eringa
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6211 LK Maastricht, The Netherlands;
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Alfons J. H. M. Houben
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Vanessa P. M. van Empel
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| |
Collapse
|
139
|
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: 0.3] [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
|
140
|
Dysregulated Epicardial Adipose Tissue as a Risk Factor and Potential Therapeutic Target of Heart Failure with Preserved Ejection Fraction in Diabetes. Biomolecules 2022; 12:biom12020176. [PMID: 35204677 PMCID: PMC8961672 DOI: 10.3390/biom12020176] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular (CV) disease and heart failure (HF) are the leading cause of mortality in type 2 diabetes (T2DM), a metabolic disease which represents a fast-growing health challenge worldwide. Specifically, T2DM induces a cluster of systemic metabolic and non-metabolic signaling which may promote myocardium derangements such as inflammation, fibrosis, and myocyte stiffness, which represent the hallmarks of heart failure with preserved ejection fraction (HFpEF). On the other hand, several observational studies have reported that patients with T2DM have an abnormally enlarged and biologically transformed epicardial adipose tissue (EAT) compared with non-diabetic controls. This expanded EAT not only causes a mechanical constriction of the diastolic filling but is also a source of pro-inflammatory mediators capable of causing inflammation, microcirculatory dysfunction and fibrosis of the underlying myocardium, thus impairing the relaxability of the left ventricle and increasing its filling pressure. In addition to representing a potential CV risk factor, emerging evidence shows that EAT may guide the therapeutic decision in diabetic patients as drugs such as metformin, glucagon-like peptide‑1 (GLP-1) receptor agonists and sodium-glucose cotransporter 2 inhibitors (SGLT2-Is), have been associated with attenuation of EAT enlargement.
Collapse
|
141
|
Stavrakis S, Elkholey K, Morris L, Niewiadomska M, Asad ZUA, Humphrey MB. Neuromodulation of Inflammation to Treat Heart Failure With Preserved Ejection Fraction: A Pilot Randomized Clinical Trial. J Am Heart Assoc 2022; 11:e023582. [PMID: 35023349 PMCID: PMC9238491 DOI: 10.1161/jaha.121.023582] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background A systemic proinflammatory state plays a central role in the development of heart failure with preserved ejection fraction. Low‐level transcutaneous vagus nerve stimulation suppresses inflammation in humans. We conducted a sham‐controlled, double‐blind, randomized clinical trial to examine the effect of chronic low‐level transcutaneous vagus nerve stimulation on cardiac function, exercise capacity, and inflammation in patients with heart failure with preserved ejection fraction. Methods and Results Patients with heart failure with preserved ejection fraction and at least 2 additional comorbidities (obesity, diabetes, hypertension, or age ≥65 years) were randomized to either active (tragus) or sham (earlobe) low‐level transcutaneous vagus nerve stimulation (20 Hz, 1 mA below discomfort threshold), for 1 hour daily for 3 months. Echocardiography, 6‐minute walk test, quality of life, and serum cytokines were assessed at baseline and 3 months. Fifty‐two patients (mean age 70.4±9.2 years; 70% female) were included (active, n=26; sham, n=26). Baseline characteristics were balanced between the 2 arms. Adherence to the protocol of daily stimulation was >90% in both arms (P>0.05). While the early mitral inflow Doppler velocity to the early diastolic mitral annulus velocity ratio did not differ between groups, global longitudinal strain and tumor necrosis factor‐α levels at 3 months were significantly improved in the active compared with the sham arm (−18.6%±2.5% versus −16.0%±2.4%, P=0.002; 8.9±2.8 pg/mL versus 11.3±2.9 pg/mL, P=0.007, respectively). The reduction in tumor necrosis factor‐α levels correlated with global longitudinal strain improvement (r=−0.73, P=0.001). Quality of life was better in the active arm. No device‐related side effects were observed. Conclusions Neuromodulation with low‐level transcutaneous vagus nerve stimulation over 3 months resulted in a significant improvement in global longitudinal strain, inflammatory cytokines, and quality of life in patients with heart failure with preserved ejection fraction. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT03327649.
Collapse
Affiliation(s)
| | - Khaled Elkholey
- University of Oklahoma Health Science Center Oklahoma City OK
| | - Lynsie Morris
- University of Oklahoma Health Science Center Oklahoma City OK
| | | | | | | |
Collapse
|
142
|
Zhang Y, Van Laer AO, Baicu CF, Neff LS, Hoffman S, Katz MR, Zeigler SM, Zile MR, Bradshaw AD. Phenotypic characterization of primary cardiac fibroblasts from patients with HFpEF. PLoS One 2022; 17:e0262479. [PMID: 35015787 PMCID: PMC8752005 DOI: 10.1371/journal.pone.0262479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/24/2021] [Indexed: 01/08/2023] Open
Abstract
Heart failure is a leading cause of hospitalizations and mortality worldwide. Heart failure with a preserved ejection fraction (HFpEF) represents a significant clinical challenge due to the lack of available treatment modalities for patients diagnosed with HFpEF. One symptom of HFpEF is impaired diastolic function that is associated with increases in left ventricular stiffness. Increases in myocardial fibrillar collagen content is one factor contributing to increases in myocardial stiffness. Cardiac fibroblasts are the primary cell type that produce fibrillar collagen in the heart. However, relatively little is known regarding phenotypic changes in cardiac fibroblasts in HFpEF myocardium. In the current study, cardiac fibroblasts were established from left ventricular epicardial biopsies obtained from patients undergoing cardiovascular interventions and divided into three categories: Referent control, hypertension without a heart failure designation (HTN (-) HFpEF), and hypertension with heart failure (HTN (+) HFpEF). Biopsies were evaluated for cardiac myocyte cross-sectional area (CSA) and collagen volume fraction. Primary fibroblast cultures were assessed for differences in proliferation and protein expression of collagen I, Membrane Type 1-Matrix Metalloproteinase (MT1-MMP), and α smooth muscle actin (αSMA). Biopsies from HTN (-) HFpEF and HTN (+) HFpEF exhibited increases in myocyte CSA over referent control although only HTN (+) HFpEF exhibited significant increases in fibrillar collagen content. No significant changes in proliferation or αSMA was detected in HTN (-) HFpEF or HTN (+) HFpEF cultures versus referent control. Significant increases in production of collagen I was detected in HF (-) HFpEF fibroblasts, whereas significant decreases in MT1-MMP levels were measured in HTN (+) HFpEF cells. We conclude that epicardial biopsies provide a viable source for primary fibroblast cultures and that phenotypic differences are demonstrated by HTN (-) HFpEF and HTN (+) HFpEF cells versus referent control.
Collapse
Affiliation(s)
- Yuhua Zhang
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - An O. Van Laer
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Catalin F. Baicu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Lily S. Neff
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Marc R. Katz
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Sanford M. Zeigler
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Michael R. Zile
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| | - Amy D. Bradshaw
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| |
Collapse
|
143
|
Fopiano KA, Jalnapurkar S, Davila AC, Arora V, Bagi Z. Coronary Microvascular Dysfunction and Heart Failure with Preserved Ejection Fraction - implications for Chronic Inflammatory Mechanisms. Curr Cardiol Rev 2022; 18:e310821195986. [PMID: 34488616 PMCID: PMC9413735 DOI: 10.2174/1573403x17666210831144651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/01/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
Coronary Microvascular Dysfunction (CMD) is now considered one of the key underlying pathologies responsible for the development of both acute and chronic cardiac complications. It has been long recognized that CMD contributes to coronary no-reflow, which occurs as an acute complication during percutaneous coronary interventions. More recently, CMD was proposed to play a mechanistic role in the development of left ventricle diastolic dysfunction in heart failure with preserved ejection fraction (HFpEF). Emerging evidence indicates that a chronic low-grade pro-inflammatory activation predisposes patients to both acute and chronic cardiovascular complications raising the possibility that pro-inflammatory mediators serve as a mechanistic link in HFpEF. Few recent studies have evaluated the role of the hyaluronan-CD44 axis in inflammation-related cardiovascular pathologies, thus warranting further investigations. This review article summarizes current evidence for the role of CMD in the development of HFpEF, focusing on molecular mediators of chronic proinflammatory as well as oxidative stress mechanisms and possible therapeutic approaches to consider for treatment and prevention.
Collapse
Affiliation(s)
- Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Sawan Jalnapurkar
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University Augusta, GA 30912, USA
| | - Alec C Davila
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Vishal Arora
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University Augusta, GA 30912, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
144
|
Soetkamp D, Gallet R, Parker SJ, Holewinski R, Venkatraman V, Peck K, Goldhaber JI, Marbán E, Van Eyk JE. Myofilament Phosphorylation in Stem Cell Treated Diastolic Heart Failure. Circ Res 2021; 129:1125-1140. [PMID: 34641704 DOI: 10.1161/circresaha.119.316311] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
RATIONALE Phosphorylation of sarcomeric proteins has been implicated in heart failure with preserved ejection fraction (HFpEF); such changes may contribute to diastolic dysfunction by altering contractility, cardiac stiffness, Ca2+-sensitivity, and mechanosensing. Treatment with cardiosphere-derived cells (CDCs) restores normal diastolic function, attenuates fibrosis and inflammation, and improves survival in a rat HFpEF model. OBJECTIVE Phosphorylation changes that underlie HFpEF and those reversed by CDC therapy, with a focus on the sarcomeric subproteome were analyzed. METHODS AND RESULTS Dahl salt-sensitive rats fed a high-salt diet, with echocardiographically verified diastolic dysfunction, were randomly assigned to either intracoronary CDCs or placebo. Dahl salt-sensitive rats receiving low salt diet served as controls. Protein and phosphorylated Ser, Thr, and Tyr residues from left ventricular tissue were quantified by mass spectrometry. HFpEF hearts exhibited extensive hyperphosphorylation with 98% of the 529 significantly changed phospho-sites increased compared with control. Of those, 39% were located within the sarcomeric subproteome, with a large group of proteins located or associated with the Z-disk. CDC treatment partially reverted the hyperphosphorylation, with 85% of the significantly altered 76 residues hypophosphorylated. Bioinformatic upstream analysis of the differentially phosphorylated protein residues revealed PKC as the dominant putative regulatory kinase. PKC isoform analysis indicated increases in PKC α, β, and δ concentration, whereas CDC treatment led to a reversion of PKCβ. Use of PKC isoform specific inhibition and overexpression of various PKC isoforms strongly suggests that PKCβ is the dominant kinase involved in hyperphosphorylation in HFpEF and is altered with CDC treatment. CONCLUSIONS Increased protein phosphorylation at the Z-disk is associated with diastolic dysfunction, with PKC isoforms driving most quantified phosphorylation changes. Because CDCs reverse the key abnormalities in HFpEF and selectively reverse PKCβ upregulation, PKCβ merits being classified as a potential therapeutic target in HFpEF, a disease notoriously refractory to medical intervention.
Collapse
Affiliation(s)
- Daniel Soetkamp
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Romain Gallet
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Sarah J Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | - Kiel Peck
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | |
Collapse
|
145
|
Mengozzi A, Pugliese NR, Chiriacò M, Masi S, Virdis A, Taddei S. Microvascular Ageing Links Metabolic Disease to Age-Related Disorders: The Role of Oxidative Stress and Inflammation in Promoting Microvascular Dysfunction. J Cardiovasc Pharmacol 2021; 78:S78-S87. [PMID: 34840260 DOI: 10.1097/fjc.0000000000001109] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/26/2021] [Indexed: 01/09/2023]
Abstract
ABSTRACT Longer life span and increased prevalence of chronic, noncommunicable, inflammatory diseases fuel cardiovascular mortality. The microcirculation is central in the cross talk between ageing, inflammation, cardiovascular, and metabolic diseases. Microvascular dysfunction, characterized by alteration in the microvascular endothelial function and wall structure, is described in an increasing number of chronic age-associated diseases, suggesting that it might be a marker of ageing superior to chronological age. The aim of this review is to thoroughly explore the connections between microvascular dysfunction, ageing, and metabolic disorders by detailing the major role played by inflammation and oxidative stress in their evolution. Older age, hypertension, nutrient abundance, and hyperglycemia concur in the induction of a persistent low-grade inflammatory response, defined as meta-inflammation or inflammageing. This increases the local generation of reactive oxygen species that further impairs endothelial function and amplifies the local inflammatory response. Mitochondrial dysfunction is a hallmark of many age-related diseases. The alterations of mitochondrial function promote irreversible modification in microvascular structure. The interest in the hypothesis of chronic inflammation at the center of the ageing process lies in its therapeutic implications. Inhibition of specific inflammatory pathways has been shown to lower the risk of many age-related diseases, including cardiovascular disease. However, the whole architecture of the inflammatory response underpinning the ageing process and its impact on the burden of age-related diseases remain to be fully elucidated. Additional studies are needed to unravel the connection between these biological pathways and to address their therapeutic power in terms of cardiovascular prevention.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy ; and
| | | | - Martina Chiriacò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
146
|
Nemska S, Gassmann M, Bang ML, Frossard N, Tavakoli R. Antagonizing the CX3CR1 Receptor Markedly Reduces Development of Cardiac Hypertrophy After Transverse Aortic Constriction in Mice. J Cardiovasc Pharmacol 2021; 78:792-801. [PMID: 34882111 DOI: 10.1097/fjc.0000000000001130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 08/01/2021] [Indexed: 12/12/2022]
Abstract
ABSTRACT Left-ventricular hypertrophy, characterized by cardiomyocyte hypertrophy, interstitial cell proliferation, and immune cell infiltration, is a high risk factor for heart failure and death. Chemokines interacting with G protein-coupled chemokine receptors probably play a role in left-ventricular hypertrophy development by promoting recruitment of activated leukocytes and modulating left-ventricular remodeling. Using the minimally invasive model of transverse aortic constriction in mice, we demonstrated that a variety of chemokine and chemokine receptor messenger Ribonucleic Acid are overexpressed in the early and late phase of hypertrophy progression. Among the chemokine receptors, Cx3cr1 and Ccr2 were most strongly overexpressed and were significantly upregulated at 3, 7, and 14 days after transverse aortic constriction. Ligands of CX3CR1 (Cx3cl1) and CCR2 (Ccl2, Ccl7, Ccl12) were significantly overexpressed in the left ventricle at the early stages after mechanical pressure overload. Pharmacological inhibition of CX3CR1 signaling using the antagonist AZD8797 led to a significant reduction of hypertrophy, whereas inhibition of CCR2 with the RS504393 antagonist did not show any effect. Furthermore, AZD8797 treatment reduced the expression of the hypertrophic marker genes Nppa and Nppb as well as the profibrotic genes Tgfb1 and Col1a1 at 14 days after transverse aortic constriction. These findings strongly suggest the involvement of the CX3CR1/CX3CL1 pathway in the pathogenesis of left-ventricular hypertrophy.
Collapse
MESH Headings
- Animals
- Aorta/physiopathology
- Aorta/surgery
- Atrial Natriuretic Factor/genetics
- Atrial Natriuretic Factor/metabolism
- CX3C Chemokine Receptor 1/antagonists & inhibitors
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Chemokine CX3CL1/genetics
- Chemokine CX3CL1/metabolism
- Collagen Type I, alpha 1 Chain/genetics
- Collagen Type I, alpha 1 Chain/metabolism
- Constriction
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Male
- Mice, Inbred C57BL
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Natriuretic Peptide, Brain/genetics
- Natriuretic Peptide, Brain/metabolism
- Pyrimidines/pharmacology
- Signal Transduction
- Thiazoles/pharmacology
- Time Factors
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Simona Nemska
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Marie-Louise Bang
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; and
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan Unit, Milan, Italy
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Reza Tavakoli
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
147
|
Cohen AJ, Teramoto K, Claggett B, Buckley L, Solomon S, Ballantyne C, Selvin E, Shah AM. Mid- to Late-Life Inflammation and Risk of Cardiac Dysfunction, HFpEF and HFrEF in Late Life. J Card Fail 2021; 27:1382-1392. [PMID: 34314823 PMCID: PMC8823406 DOI: 10.1016/j.cardfail.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Epidemiologic data supporting the association of accumulated inflammation from mid- to late life with late-life risk of cardiac dysfunction and heart failure (HF) is limited. METHODS AND RESULTS Among 4011 participants in the Atherosclerosis Risk in Communities study who were free of prevalent cardiovascular disease at study Visit 5, accumulated inflammation was defined as time-averaged high-sensitivity c-reactive protein (hsCRP) over 3 visits spanning 1990 to 2013. Associations with left ventricular (LV) function at Visit 5 and with incident adjudicated HF post Visit 5 were assessed using linear and Cox regression, adjusting for demographics and comorbidities. Higher accumulated hsCRP was associated with greater LV mass index, lower e', higher E/e', and higher adjusting for demographics (all P ≤0.01), but only with higher pulmonary artery systolic pressure after adjustment for comorbidities (P = 0.024). At 5.3 ± 1.2 year follow-up, higher accumulated hsCRP was associated with greater risk of incident HF (HR 1.31 [95% CI 1.18-1.47], P < 0.001), HFrEF (1.26 [1.05-1.52], P = 0.01), and HFpEF (1.30 [1.11-1.53], P = 0.001) in demographic-adjusted models, but not after adjustment for comorbidities (all P > 0.10). Only Visit 5 hsCRP remained associated with incident HF (1.12 [1.02-1.24], P = 0.02) after full adjustment. CONCLUSIONS Greater accumulated inflammation is associated with worse LV function and heightened HF risk in late-life. These relationships are attenuated after adjusting for HF risk factors.
Collapse
Affiliation(s)
- Aaron J Cohen
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | - Kanako Teramoto
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Division of Cardiology, St. Marianna University School of Medicine Hospital, Kanagawa, Japan
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Leo Buckley
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Scott Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amil M Shah
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
148
|
Kobusiak-Prokopowicz M, Kaaz K, Marciniak D, Karolko B, Mysiak A. Relationships between Circulating Matrix Metalloproteinases, Tissue Inhibitor TIMP-2, and Renal Function in Patients with Myocarditis. Kidney Blood Press Res 2021; 46:749-757. [PMID: 34801997 DOI: 10.1159/000519594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 09/11/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Under physiological conditions, the myocardial extracellular matrix (ECM) is maintained by matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs). However, changes in the balance between MMPs and TIMPs can lead to pathological remodeling of the ECM, which contributes to cardiovascular and kidney diseases. The aim of our study was to assess levels of MMPs and TIMP-2 in patients with myocarditis and their relationship to renal function. MATERIALS AND METHODS Forty five patients with myocarditis who underwent CMR were included, comprising 11 with concurrent chronic kidney disease (CKD). Blood samples were obtained to assess serum levels of MMP-2, MMP-3, MMP-9, and TIMP-2. RESULTS Serum MMP-2, MMP-3, and TIMP-2 levels negatively correlated with the ejection fraction in patients with myocarditis, while MMP-3 levels correlated with longitudinal deformation (p < 0.05). Serum MMP-2, MMP-3, and TIMP-2 levels also negatively correlated with renal function, as assessed by the estimated glomerular filtration rate (eGFR) (p < 0.05). Patients with myocarditis and concurrent CKD had higher levels of MMP-2 and TIMP-2 than those without kidney damage. CONCLUSIONS (1) We demonstrated that MMP-2, MMP-3, and TIMP-2 concentrations were related to left-ventricular ejection fraction, and MMP-3 levels correlated with longitudinal deformation, indicating MMPs play an important role in the post-inflammatory remodeling of the myocardium. (2) A negative correlation between the eGFR and MMP-2, MMP-3, and TIMP-2 and a positive correlation between creatinine and MMP-3 levels indicate the role of MMPs and TIMP-2 in renal dysfunction.
Collapse
Affiliation(s)
| | - Konrad Kaaz
- Department of Cardiology, Wroclaw Medical University, Wroclaw, Poland
| | - Dominik Marciniak
- Department of Drugs Form Technology, Wroclaw Medical University, Wroclaw, Poland
| | - Bożena Karolko
- Department of Cardiology, Wroclaw Medical University, Wroclaw, Poland
| | - Andrzej Mysiak
- Department of Cardiology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
149
|
Priksz D, Lampe N, Kovacs A, Herwig M, Bombicz M, Varga B, Wilisicz T, Szilvassy J, Posa A, Kiss R, Gesztelyi R, Raduly A, Szekeres R, Sieme M, Papp Z, Toth A, Hamdani N, Szilvassy Z, Juhasz B. Nicotinic-acid derivative BGP-15 improves diastolic function in a rabbit model of atherosclerotic cardiomyopathy. Br J Pharmacol 2021; 179:2240-2258. [PMID: 34811751 DOI: 10.1111/bph.15749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Small molecule BGP-15 has been reported to alleviate signs of heart failure and improve muscle function in murine models. Here, we investigated the acute and chronic effects of BGP-15 in a rabbit model of atherosclerotic cardiomyopathy. EXPERIMENTAL APPROACH Rabbits were maintained on standard chow (Control) or atherogenic diet (HC) for 16 weeks. BGP-15 was administered intravenously (once) or orally (for 16 weeks), to assess acute and chronic effects. Cardiac function was evaluated by echocardiography, endothelium-dependent vasorelaxation was assessed, and key molecules of the protein kinase G (PKG) axis were examined by ELISA and Western blot. Passive force generation was investigated in skinned cardiomyocytes. KEY RESULTS Both acute and chronic BGP-15 treatment improved the diastolic performance of the diseased heart, however, vasorelaxation and serum lipid markers were unaffected. Myocardial cGMP levels were elevated in the BGP-15-treated group, along with preserved PKG activity and increased phospholamban Ser16-phosphorylation. PDE5 expression decreased in the BGP-15-treated group, and the substance inhibited PDE1 enzyme. Cardiomyocyte passive tension reduced in BGP-15-treated rabbits, the ratio of titin N2BA/N2B isoforms increased, and PKG-dependent N2B-titin phosphorylation elevated in the BGP-15-treated group. CONCLUSIONS AND IMPLICATIONS Here we report that BGP-15-treatment improves diastolic function, reduces cardiomyocyte stiffness, and restores titin compliance in a rabbit model of atherosclerotic cardiomyopathy by increasing the activity of the cGMP-PKG axis. As BGP-15 is proven to be safe, it may have clinical value in the treatment of diastolic dysfunction.
Collapse
Affiliation(s)
- Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Nora Lampe
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Arpad Kovacs
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany
| | - Melissa Herwig
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Balazs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Tician Wilisicz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Szilvassy
- Department of Otorhinolaryngology and Head-Neck Surgery, University of Debrecen, Debrecen, Hungary
| | - Aniko Posa
- Department of Physiology, Anatomy and Neuroscience, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Arnold Raduly
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Reka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Marcel Sieme
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany
| | - Zoltan Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Zoltan Szilvassy
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Bela Juhasz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
150
|
Cytokine-Mediated Alterations of Human Cardiac Fibroblast's Secretome. Int J Mol Sci 2021; 22:ijms222212262. [PMID: 34830141 PMCID: PMC8617966 DOI: 10.3390/ijms222212262] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 11/17/2022] Open
Abstract
Fibroblasts contribute to approximately 20% of the non-cardiomyocytic cells in the heart. They play important roles in the myocardial adaption to stretch, inflammation, and other pathophysiological conditions. Fibroblasts are a major source of extracellular matrix (ECM) proteins whose production is regulated by cytokines, such as TNF-α or TGF-β. The resulting myocardial fibrosis is a hallmark of pathological remodeling in dilated cardiomyopathy (DCM). Therefore, in the present study, the secretome and corresponding transcriptome of human cardiac fibroblasts from patients with DCM was investigated under normal conditions and after TNF-α or TGF-β stimulation. Secreted proteins were quantified via mass spectrometry and expression of genes coding for secreted proteins was analyzed via Affymetrix Transcriptome Profiling. Thus, we provide comprehensive proteome and transcriptome data on the human cardiac fibroblast’s secretome. In the secretome of quiescent fibroblasts, 58% of the protein amount belonged to the ECM fraction. Interestingly, cytokines were responsible for 5% of the total protein amount in the secretome and up to 10% in the corresponding transcriptome. Furthermore, cytokine gene expression and secretion were upregulated upon TNF-α stimulation, while collagen secretion levels were elevated after TGF-β treatment. These results suggest that myocardial fibroblasts contribute to pro-fibrotic and to inflammatory processes in response to extracellular stimuli.
Collapse
|