51
|
Rossi VA, Gruebler M, Monzo L, Galluzzo A, Beltrami M. The Different Pathways of Epicardial Adipose Tissue across the Heart Failure Phenotypes: From Pathophysiology to Therapeutic Target. Int J Mol Sci 2023; 24:6838. [PMID: 37047810 PMCID: PMC10095298 DOI: 10.3390/ijms24076838] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Epicardial adipose tissue (EAT) is an endocrine and paracrine organ constituted by a layer of adipose tissue directly located between the myocardium and visceral pericardium. Under physiological conditions, EAT exerts protective effects of brown-like fat characteristics, metabolizing excess fatty acids, and secreting anti-inflammatory and anti-fibrotic cytokines. In certain pathological conditions, EAT acquires a proatherogenic transcriptional profile resulting in increased synthesis of biologically active adipocytokines with proinflammatory properties, promoting oxidative stress, and finally causing endothelial damage. The role of EAT in heart failure (HF) has been mainly limited to HF with preserved ejection fraction (HFpEF) and related to the HFpEF obese phenotype. In HFpEF, EAT seems to acquire a proinflammatory profile and higher EAT values have been related to worse outcomes. Less data are available about the role of EAT in HF with reduced ejection fraction (HFrEF). Conversely, in HFrEF, EAT seems to play a nutritive role and lower values may correspond to the expression of a catabolic, adverse phenotype. As of now, there is evidence that the beneficial systemic cardiovascular effects of sodium-glucose cotransporter-2 receptors-inhibitors (SGLT2-i) might be partially mediated by inducing favorable modifications on EAT. As such, EAT may represent a promising target organ for the development of new drugs to improve cardiovascular prognosis. Thus, an approach based on detailed phenotyping of cardiac structural alterations and distinctive biomolecular pathways may change the current scenario, leading towards a precision medicine model with specific therapeutic targets considering different individual profiles. The aim of this review is to summarize the current knowledge about the biomolecular pathway of EAT in HF across the whole spectrum of ejection fraction, and to describe the potential of EAT as a therapeutic target in HF.
Collapse
Affiliation(s)
- Valentina A. Rossi
- University Heart Center, Department of Cardiology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Martin Gruebler
- Regional Hospital Neustadt, 2700 Wiener Neustadt, Austria
- Faculty of Medicine, Medical University of Graz, 8036 Graz, Austria
- Faculty of Medicine, Sigmund Freud University Vienna, 1020 Vienna, Austria
| | - Luca Monzo
- Centre d’Investigations Cliniques Plurithématique 1433 and Inserm U1116, Université de Lorraine, CHRU Nancy, FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), 54035 Nancy, France
| | | | - Matteo Beltrami
- Azienda USL Toscana Centro, Cardiology Unit, San Giovanni di Dio Hospital, 50143 Florence, Italy;
| |
Collapse
|
52
|
Thapa P, Upadhyay SP, Singh V, Boinpelly VC, Zhou J, Johnson DK, Gurung P, Lee ES, Sharma R, Sharma M. Chalcone: A potential scaffold for NLRP3 inflammasome inhibitors. EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY REPORTS 2023; 7:100100. [PMID: 37033416 PMCID: PMC10081147 DOI: 10.1016/j.ejmcr.2022.100100] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Overactivated NLRP3 inflammasome has been shown to associate with an increasing number of disease conditions. Activation of the NLRP3 inflammasome results in caspase-1-catalyzed formation of active pro-inflammatory cytokines (IL-1β and IL-18) resulting in pyroptosis. The multi-protein composition of the NLRP3 inflammasome and its sensitivity to several damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) make this extensively studied inflammasome an attractive target to treat chronic conditions. However, none of the known NLRP3 inhibitors has been approved for clinical use. Sulfonylurea and covalent inhibitors with electrophilic warhead (Michael acceptor) are among the prominent classes of compounds explored for their NLRP3 inhibitory effects. Chalcone, a small molecule with α, β unsaturated carbonyl group (Michael acceptor), has also been studied as a promising scaffold for the development of NLRP3 inhibitors. Low molecular weight, easy to manipulate lipophilicity and cost-effectiveness have attracted many to use chalcone scaffold for drug development. In this review, we highlight chalcone derivatives with NLRP3 inflammasome inhibitory activities. Recent developments and potential new directions summarized here will, hopefully, serve as valuable perspectives for investigators including medicinal chemists and drug discovery researchers to utilize chalcone as a scaffold for developing novel NLRP3 inflammasome inhibitors.
Collapse
Affiliation(s)
- Pritam Thapa
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
| | - Sunil P. Upadhyay
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
| | - Vikas Singh
- Division of Neurology, KCVA Medical Center, Kansas City, MO, USA
| | - Varun C. Boinpelly
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Jianping Zhou
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO, USA
| | - David K. Johnson
- Department of Computational Chemical Biology Core, Molecular Graphics and Modeling Core, University of Kansas, KS, 66047, USA
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, Iowa City, IA, 52242, USA
| | - Eung Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Ram Sharma
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
| | - Mukut Sharma
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO, USA
| |
Collapse
|
53
|
Gomez SE, Parizo J, Ermakov S, Larson J, Wallace R, Assimes T, Hlatky M, Stefanick M, Perez MV. Evaluation of the association between circulating IL-1β and other inflammatory cytokines and incident atrial fibrillation in a cohort of postmenopausal women. Am Heart J 2023; 258:157-167. [PMID: 36646198 PMCID: PMC10023332 DOI: 10.1016/j.ahj.2023.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Inflammatory cytokines play a role in atrial fibrillation (AF). Interleukin (IL)-1β, which is targeted in the treatment of ischemic heart disease, has not been well-studied in relation to AF. METHODS Postmenopausal women from the Women's Health Initiative were included. Cox proportional hazards regression models were used to evaluate the association between log-transformed baseline cytokine levels and future AF incidence. Models were adjusted for body mass index, age, race, education, hypertension, diabetes, hyperlipidemia, current smoking, and history of coronary heart disease, congestive heart failure, or peripheral artery disease. RESULTS Of 16,729 women, 3,943 developed AF over an average of 8.5 years. Racial and ethnic groups included White (77.4%), Black/African-American (16.1%), Asian (2.7%), American Indian/Alaska Native (1.0%), and Hispanic (5.5%). Baseline IL-1β log continuous levels were not significantly associated with incident AF (HR 0.86 per 1 log [pg/mL] increase, P= .24), similar to those of other inflammatory cytokines, IL-7, IL-8, IL-10, IGF-1, and TNF-α. There were significant associations between C-reactive protein (CRP) and IL-6 with incident AF. CONCLUSIONS In this large cohort of postmenopausal women, there was no significant association between IL-1β and incident AF, although downstream effectors, CRP and IL-6, were associated with incident AF.
Collapse
Affiliation(s)
- Sofia E Gomez
- Department of Medicine, Stanford, University School of Medicine, Stanford, CA
| | - Justin Parizo
- Division of Cardiovascular Medicine, Department of Medicine, Stanford, University School of Medicine, Stanford, CA
| | - Simon Ermakov
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA
| | | | - Robert Wallace
- College of Public Health, University of Iowa, Iowa City, IA
| | - Themistocles Assimes
- Division of Cardiovascular Medicine, Department of Medicine, Stanford, University School of Medicine, Stanford, CA
| | - Mark Hlatky
- Division of Cardiovascular Medicine, Department of Medicine, Stanford, University School of Medicine, Stanford, CA; Department of Health Research and Policy, Stanford University, Stanford, CA
| | - Marcia Stefanick
- Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA
| | - Marco V Perez
- Division of Cardiovascular Medicine, Department of Medicine, Stanford, University School of Medicine, Stanford, CA.
| |
Collapse
|
54
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
55
|
Vosko I, Zirlik A, Bugger H. Impact of COVID-19 on Cardiovascular Disease. Viruses 2023; 15:508. [PMID: 36851722 PMCID: PMC9962056 DOI: 10.3390/v15020508] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection with the novel severe acute respiratory distress syndrome corona virus 2 (SARS-CoV-2). Until now, more than 670 million people have suffered from COVID-19 worldwide, and roughly 7 million death cases were attributed to COVID-19. Recent evidence suggests an interplay between COVID-19 and cardiovascular disease (CVD). COVID-19 may serve as a yet underappreciated CVD risk modifier, including risk factors such as diabetes mellitus or arterial hypertension. In addition, recent data suggest that previous COVID-19 may increase the risk for many entities of CVD to an extent similarly observed for traditional cardiovascular (CV) risk factors. Furthermore, increased CVD incidence and worse clinical outcomes in individuals with preexisting CVD have been observed for myocarditis, acute coronary syndrome, heart failure (HF), thromboembolic complications, and arrhythmias. Direct and indirect mechanisms have been proposed by which COVID-19 may impact CVD and CV risk, including viral entry into CV tissue or by the induction of a massive systemic inflammatory response. In the current review, we provide an overview of the literature reporting an interaction between COVID-19 and CVD, review potential mechanisms underlying this interaction, and discuss preventive and treatment strategies and their interference with CVD that were evaluated since the onset of the COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
56
|
Dimosiari A, Patoulias D, Kitas GD, Dimitroulas T. Do Interleukin-1 and Interleukin-6 Antagonists Hold Any Place in the Treatment of Atherosclerotic Cardiovascular Disease and Related Co-Morbidities? An Overview of Available Clinical Evidence. J Clin Med 2023; 12:jcm12041302. [PMID: 36835838 PMCID: PMC9962740 DOI: 10.3390/jcm12041302] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Cardiovascular disease (CVD) constitutes a real pandemic of the 21st century. According to data from the Centers for Disease Control and Prevention, one person dies every 34 min due to some form of CVD in the United States. Apart from the extremely high morbidity and mortality accompanying CVD, the economic burden seems to be unbearable even for developed countries in the Western World. The role of inflammation in the development and progression of CVD appears to be crucial, while, various inflammatory pathways, such as the Nod-like receptor protein 3 (NLRP3) inflammasome-interleukin (IL)-1/IL-6 pathway of the innate immunity, have attracted scientific interest during the last decade, as a potential treatment target in primary and/or secondary prevention of CVD. Whereas there is a significant amount of evidence, stemming mainly from observational studies, concerning the cardiovascular safety of IL-1 and IL-6 antagonists in patients with rheumatic diseases, evidence from relevant randomized controlled trials (RCTs) is rather scarce and conflicting, especially for patients without underlying rheumatic disease. In this review, we summarize and critically present the currently available evidence, both from RCTs and observational studies, concerning the place that IL-1 and IL-6 antagonists may hold in the treatment of CVD.
Collapse
Affiliation(s)
- Athina Dimosiari
- Second Department of Internal Medicine, European Interbalkan Medical Center, 57001 Thessaloniki, Greece
| | - Dimitrios Patoulias
- Second Department of Internal Medicine, European Interbalkan Medical Center, 57001 Thessaloniki, Greece
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, General Hospital Hippokration, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Correspondence:
| | - George D. Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, General Hospital Hippokration, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
57
|
Pugliese NR, Pellicori P, Filidei F, De Biase N, Maffia P, Guzik TJ, Masi S, Taddei S, Cleland JGF. Inflammatory pathways in heart failure with preserved left ventricular ejection fraction: implications for future interventions. Cardiovasc Res 2023; 118:3536-3555. [PMID: 36004819 PMCID: PMC9897694 DOI: 10.1093/cvr/cvac133] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
Many patients with symptoms and signs of heart failure have a left ventricular ejection fraction ≥50%, termed heart failure with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous syndrome mainly affecting older people who have many other cardiac and non-cardiac conditions that often cast doubt on the origin of symptoms, such as breathlessness, or signs, such as peripheral oedema, rendering them neither sensitive nor specific to the diagnosis of HFpEF. Currently, management of HFpEF is mainly directed at controlling symptoms and treating comorbid conditions such as hypertension, atrial fibrillation, anaemia, and coronary artery disease. HFpEF is also characterized by a persistent increase in inflammatory biomarkers. Inflammation may be a key driver of the development and progression of HFpEF and many of its associated comorbidities. Detailed characterization of specific inflammatory pathways may provide insights into the pathophysiology of HFpEF and guide its future management. There is growing interest in novel therapies specifically designed to target deregulated inflammation in many therapeutic areas, including cardiovascular disease. However, large-scale clinical trials investigating the effectiveness of anti-inflammatory treatments in HFpEF are still lacking. In this manuscript, we review the role of inflammation in HFpEF and the possible implications for future trials.
Collapse
Affiliation(s)
| | - Pierpaolo Pellicori
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| | - Francesco Filidei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples 80138, Italy
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Internal and Agricultural Medicine, Jagiellonian University, Collegium Medicum, Krakow 31-008, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - John G F Cleland
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
58
|
Filipp M, Thorp EB. Revisiting IL-1β: Linking Metainflammation to Diastolic Dysfunction. JACC Basic Transl Sci 2023; 8:186-188. [PMID: 36908665 PMCID: PMC9998468 DOI: 10.1016/j.jacbts.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Affiliation(s)
- Mallory Filipp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
59
|
Fir(e)ing the Rhythm. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.12.004] [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: 02/17/2023]
|
60
|
Liu H, Huang Y, Zhao Y, Kang GJ, Feng F, Wang X, Liu M, Shi G, Revelo X, Bernlohr D, Dudley SC. Inflammatory Macrophage Interleukin-1β Mediates High-Fat Diet-Induced Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2023; 8:174-185. [PMID: 36908663 PMCID: PMC9998610 DOI: 10.1016/j.jacbts.2022.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/02/2022]
Abstract
Diabetes mellitus (DM) is a main risk factor for diastolic dysfunction (DD) and heart failure with preserved ejection fraction. High-fat diet (HFD) mice presented with diabetes mellitus, DD, higher cardiac interleukin (IL)-1β levels, and proinflammatory cardiac macrophage accumulation. DD was significantly ameliorated by suppressing IL-1β signaling or depleting macrophages. Mice with macrophages unable to adopt a proinflammatory phenotype were low in cardiac IL-1β levels and were resistant to HFD-induced DD. IL-1β enhanced mitochondrial reactive oxygen species (mitoROS) in cardiomyocytes, and scavenging mitoROS improved HFD-induced DD. In conclusion, macrophage-mediated inflammation contributed to HFD-associated DD through IL-1β and mitoROS production.
Collapse
Key Words
- CCR2, C-C motif chemokine receptor 2
- CM, cardiomyocyte
- DD, diastolic dysfunction
- DM, diabetes mellitus
- EF, ejection fraction
- FABP4, fatty acid binding protein 4
- HF, heart failure
- HFD, high-fat diet
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- IL, interleukin
- IL-1β
- IL1RA, interleukin 1 receptor antagonist
- KO, knockout
- MCP, monocyte chemoattractant protein
- MyBP-C, myosin binding protein C
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- Timd4, T cell immunoglobulin and mucin domain containing 4
- WT, wild-type
- diabetes
- diastolic dysfunction
- inflammation
- macrophage
- mitoROS, mitochondrial reactive oxygen species
- mitochondria
Collapse
Affiliation(s)
- Hong Liu
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yimao Huang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yang Zhao
- Division of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu, China
| | - Gyeoung-Jin Kang
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Feng Feng
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xiaodan Wang
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Man Liu
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guangbin Shi
- Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Xavier Revelo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - David Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Samuel C. Dudley
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
61
|
Evans MA, Walsh K. Clonal hematopoiesis, somatic mosaicism, and age-associated disease. Physiol Rev 2023; 103:649-716. [PMID: 36049115 PMCID: PMC9639777 DOI: 10.1152/physrev.00004.2022] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 07/19/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
Somatic mosaicism, the occurrence of multiple genetically distinct cell clones within the same tissue, is an evitable consequence of human aging. The hematopoietic system is no exception to this, where studies have revealed the presence of expanded blood cell clones carrying mutations in preleukemic driver genes and/or genetic alterations in chromosomes. This phenomenon is referred to as clonal hematopoiesis and is remarkably prevalent in elderly individuals. While clonal hematopoiesis represents an early step toward a hematological malignancy, most individuals will never develop blood cancer. Somewhat unexpectedly, epidemiological studies have found that clonal hematopoiesis is associated with an increase in the risk of all-cause mortality and age-related disease, particularly in the cardiovascular system. Studies using murine models of clonal hematopoiesis have begun to shed light on this relationship, suggesting that driver mutations in mature blood cells can causally contribute to aging and disease by augmenting inflammatory processes. Here we provide an up-to-date review of clonal hematopoiesis within the context of somatic mosaicism and aging and describe recent epidemiological studies that have reported associations with age-related disease. We will also discuss the experimental studies that have provided important mechanistic insight into how driver mutations promote age-related disease and how this knowledge could be leveraged to treat individuals with clonal hematopoiesis.
Collapse
Affiliation(s)
- Megan A Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
62
|
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
|
63
|
Mahfooz K, Rana A, Palagati K, Suvarna AK, Perryman C, Gaddipati SP, Adhnon A, Andani R, Vasavada A. Anakinra in Heart Failure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Med Sci (Basel) 2022; 11:4. [PMID: 36649041 PMCID: PMC9844326 DOI: 10.3390/medsci11010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Background: Heart failure (HF) has become increasingly difficult to manage given its increasing incidence. Despite the availability of novel treatment target relieving inhibition and congestions for neurohormonal activation, heart failure is one of leading health conditions associated with high hospitalization and readmission rates, resulting in poor quality of life. In light of this, this article serves to demonstrate the effect of anakinra as one of the treatment paradigms for HF to explore the need for advanced novel interventions. Methods: We conducted a search in five electronic databases, including Embase, MEDLINE, Cochrane, Scopus, and PubMed, for RCTs (randomized controlled trials) evaluating the effects of anakinra against placebo in HF. Meta-analysis was performed using RevMan version 5.4. Results: Eight RCTs were obtained and included for analysis in this study. The results demonstrate that anakinra significantly reduces the levels of CRP (C-reactive protein), with significant difference between anakinra- and placebo-treated groups. Analyses also show that CRP failed to cause an improvement in peak oxygen consumption and ventilatory efficiency. Additionally, the treatment-related adverse events were insignificant. Some considerable limitations are that the same set of researchers were involved in most of the studies; hence, more independent studies need to be encouraged. Conclusion: Anakinra was associated with a reduction in CRP levels, indicating some anti-inflammatory effects but no effect on function, exercise capacity, and adverse effects.
Collapse
Affiliation(s)
- Kamran Mahfooz
- Department of Internal Medicine, Lincoln Medical Center, New York, NY 10451, USA
| | - Aditya Rana
- Department of Internal Medicine, Armed Forces Medical College, Pune 411040, India
| | - Keerthi Palagati
- Department of Internal Medicine, Government Medical College, Ananthapur 515001, India
| | | | - Christian Perryman
- Department of Internal Medicine, Saint James School of Medicine, Cane Hall Road, Arnos Vale VC0280, Saint Vincent and the Grenadines
| | - Sai Pranathi Gaddipati
- Department of Internal Medicine, Mallareddy Medical College for Women, Hyderabad 500055, India
| | - Arshiya Adhnon
- Department of Internal Medicine, Dubai Medical College, Al Muhaisanah 1, Al Mizhar, Dubai P.O. Box 20170, United Arab Emirates
| | - Rupesh Andani
- Department of Internal Medicine, MP Shah Medical College, Jamnagar 361008, India
| | - Advait Vasavada
- Department of Internal Medicine, MP Shah Medical College, Jamnagar 361008, India
| |
Collapse
|
64
|
Heart Failure in Menopause: Treatment and New Approaches. Int J Mol Sci 2022; 23:ijms232315140. [PMID: 36499467 PMCID: PMC9735523 DOI: 10.3390/ijms232315140] [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: 10/25/2022] [Revised: 11/15/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Aging is an important risk factor for the development of heart failure (HF) and half of patients with HF have preserved ejection fraction (HFpEF) which is more common in elderly women. In general, sex differences that lead to discrepancies in risk factors and to the development of cardiovascular disease (CVD) have been attributed to the reduced level of circulating estrogen during menopause. Estrogen receptors adaptively modulate fibrotic, apoptotic, inflammatory processes and calcium homeostasis, factors that are directly involved in the HFpEF. Therefore, during menopause, estrogen depletion reduces the cardioprotection. Preclinical menopause models demonstrated that several signaling pathways and organ systems are closely involved in the development of HFpEF, including dysregulation of the renin-angiotensin system (RAS), chronic inflammatory process and alteration in the sympathetic nervous system. Thus, this review explores thealterations observed in the condition of HFpEF induced by menopause and the therapeutic targets with potential to interfere with the disease progress.
Collapse
|
65
|
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
|
66
|
Tao WX, Qian GY, Li HD, Su F, Wang Z. Body mass index and outcomes of patients with cardiogenic shock: A systematic review and meta-analysis. World J Clin Cases 2022; 10:10956-10966. [PMID: 36338207 PMCID: PMC9631130 DOI: 10.12998/wjcc.v10.i30.10956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/09/2022] [Accepted: 09/09/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cardiogenic shock continues to be a highly morbid complication that affects around 7%-10% of patients with acute myocardial infarction or heart failure. Similarly, obesity has become a worldwide epidemic.
AIM To analyze the impact of higher body mass index (BMI) on outcomes of patients with cardiogenic shock.
METHODS A systematic and comprehensive search was undertaken on the electronic databases of PubMed, Embase, ScienceDirect, CENTRAL, and Google Scholar for all types of studies comparing mortality outcomes of patients with cardiogenic shock based on BMI. All studies defined overweight or obese patients based on the World Health Organization BMI criteria. The data were then extracted and assessed on the basis of the Reference Citation Analysis (https://www.referencecitationanalysis.com/).
RESULTS Five studies were included. On pooled analysis of multivariable-adjusted ratios, we noted a statistically significantly reduced risk of mortality in overweight/ obese vs normal patients (three studies; odds ratio [OR] = 0.92, 95% confidence interval [CI]: 0.85-0.98, I2 = 85%). On meta-analysis, we noted that crude mortality rates did not significantly differ between overweight/obese and normal patients after cardiogenic shock (OR = 0.95, 95%CI: 0.79-1.15, I2 = 99%). The results were not stable on sensitivity analysis and were associated with substantial heterogeneity.
CONCLUSION Current evidence on the association between overweight/obesity and mortality after cardiogenic shock is scarce and conflicting. The obesity paradox might exist in patients with cardiogenic shock but could be confounded by the use of mechanical circulatory support. There is a need for further studies to clarify this relationship.
Collapse
Affiliation(s)
- Wen-Xia Tao
- Department of Cardiovascular Medicine, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou 313000, Zhejiang Province, China
| | - Guo-Ying Qian
- Department of Cardiovascular Medicine, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou 313000, Zhejiang Province, China
| | - Hong-Dan Li
- Department of Cardiovascular Medicine, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou 313000, Zhejiang Province, China
| | - Feng Su
- Department of Cardiovascular Medicine, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou 313000, Zhejiang Province, China
| | - Zhou Wang
- Department of Cardiovascular Medicine, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou 313000, Zhejiang Province, China
| |
Collapse
|
67
|
Ferlito A, Campochiaro C, Tomelleri A, Dagna L, De Luca G. Primary heart involvement in systemic sclerosis, from conventional to innovative targeted therapeutic strategies. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2022; 7:179-188. [PMID: 36211207 PMCID: PMC9537702 DOI: 10.1177/23971983221083772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/08/2022] [Indexed: 10/03/2023]
Abstract
Primary heart involvement is frequent in systemic sclerosis, even though often sub-clinical, and includes cardiac abnormalities that are predominantly attributable to systemic sclerosis rather than other causes and/or complications. A timely diagnosis is crucial to promptly start the appropriate therapy and to prevent the potential life-threatening early and late complications. There is little evidence on how to best manage systemic sclerosis-primary heart involvement as no specific treatment recommendations for heart disease are available, and a shared treatment approach is still lacking. The objective of this review is to summarize the state of the art of current literature and the overall management strategies and therapeutic approaches for systemic sclerosis-primary heart involvement. Novel insights into pathogenic mechanisms of systemic sclerosis-primary heart involvement are presented to facilitate the comprehension of therapeutic targets and novel treatment strategies.
Collapse
Affiliation(s)
| | - Corrado Campochiaro
- Vita-Salute San Raffaele University,
Milan, Italy
- Unit of Immunology, Rheumatology,
Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alessandro Tomelleri
- Vita-Salute San Raffaele University,
Milan, Italy
- Unit of Immunology, Rheumatology,
Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Lorenzo Dagna
- Vita-Salute San Raffaele University,
Milan, Italy
- Unit of Immunology, Rheumatology,
Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giacomo De Luca
- Vita-Salute San Raffaele University,
Milan, Italy
- Unit of Immunology, Rheumatology,
Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
68
|
Zheng Y, Xu L, Dong N, Li F. NLRP3 inflammasome: The rising star in cardiovascular diseases. Front Cardiovasc Med 2022; 9:927061. [PMID: 36204568 PMCID: PMC9530053 DOI: 10.3389/fcvm.2022.927061] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the prevalent cause of mortality around the world. Activation of inflammasome contributes to the pathological progression of cardiovascular diseases, including atherosclerosis, abdominal aortic aneurysm, myocardial infarction, dilated cardiomyopathy, diabetic cardiomyopathy, heart failure, and calcific aortic valve disease. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome plays a critical role in the innate immune response, requiring priming and activation signals to provoke the inflammation. Evidence shows that NLRP3 inflammasome not only boosts the cleavage and release of IL-1 family cytokines, but also leads to a distinct cell programmed death: pyroptosis. The significance of NLRP3 inflammasome in the CVDs-related inflammation has been extensively explored. In this review, we summarized current understandings of the function of NLRP3 inflammasome in CVDs and discussed possible therapeutic options targeting the NLRP3 inflammasome.
Collapse
|
69
|
Elkholey K, Niewiadomska M, Morris L, Whyte S, Houser J, Humphrey MB, Stavrakis S. Transcutaneous Vagus Nerve Stimulation Ameliorates the Phenotype of Heart Failure With Preserved Ejection Fraction Through Its Anti-Inflammatory Effects. Circ Heart Fail 2022; 15:e009288. [PMID: 35862007 PMCID: PMC9388556 DOI: 10.1161/circheartfailure.122.009288] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND A systemic proinflammatory state plays a central role in the development of heart failure with preserved ejection fraction (HFpEF). Low-level transcutaneous vagus nerve stimulation (LLTS) suppresses inflammation in animals and humans, mediated by an α7nAchR (alpha7 nicotinic acetylcholine receptor)-dependent pathway. We examined the effects of LLTS on cardiac function, inflammation, and fibrosis in the presence of α7nAchR pharmacological blockade in a rat model of HFpEF. METHODS Dahl salt-sensitive rats at 7 weeks of age were treated with high-salt diet for 6 weeks to induce HFpEF, followed by 4 weeks of (1) LLTS, (2) LLTS with the α7nAchR blocker methyllycaconitine, (3) sham, and (4) olmesartan. Blood pressure, cardiac function by echocardiography, heart rate variability, and serum cytokines were measured at 13 and 17 weeks of age. Cardiac fibrosis, inflammatory cell infiltration, and gene expression were determined at 17 weeks. RESULTS LLTS attenuated the increase in blood pressure; improved cardiac function; decreased inflammatory cytokines, macrophage infiltration, and fibrosis; and improved survival compared with other groups. Methyllycaconitine attenuated these effects, whereas olmesartan did not improve cardiac function or fibrosis despite maintaining similar blood pressure as LLTS. Heart rate variability was similarly improved in the LLTS and LLTS plus methyllycaconitine groups but remained low in the other groups. LLTS reversed the dysregulated inflammatory signaling pathways in HFpEF hearts. CONCLUSIONS Neuromodulation with LLTS improved cardiac function in a rat model of HFpEF through its anti-inflammatory and antifibrotic effects. These results provide the basis for further clinical trials in humans.
Collapse
Affiliation(s)
- Khaled Elkholey
- Cardiovascular Section, Department of Medicine (K.E., M.N., L.M., S.W., S.S.), University of Oklahoma Health Science Center, Oklahoma City
| | - Monika Niewiadomska
- Cardiovascular Section, Department of Medicine (K.E., M.N., L.M., S.W., S.S.), University of Oklahoma Health Science Center, Oklahoma City
| | - Lynsie Morris
- Cardiovascular Section, Department of Medicine (K.E., M.N., L.M., S.W., S.S.), University of Oklahoma Health Science Center, Oklahoma City
| | - Seabrook Whyte
- Cardiovascular Section, Department of Medicine (K.E., M.N., L.M., S.W., S.S.), University of Oklahoma Health Science Center, Oklahoma City
| | - Jeremy Houser
- Rheumatology Section, Department of Medicine (J.H., M.B.H.), University of Oklahoma Health Science Center, Oklahoma City
| | - Mary Beth Humphrey
- Rheumatology Section, Department of Medicine (J.H., M.B.H.), University of Oklahoma Health Science Center, Oklahoma City
| | - Stavros Stavrakis
- Cardiovascular Section, Department of Medicine (K.E., M.N., L.M., S.W., S.S.), University of Oklahoma Health Science Center, Oklahoma City
| |
Collapse
|
70
|
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
|
71
|
Shen S, Duan J, Hu J, Qi Y, Kang L, Wang K, Chen J, Wu X, Xu B, Gu R. Colchicine alleviates inflammation and improves diastolic dysfunction in heart failure rats with preserved ejection fraction. Eur J Pharmacol 2022; 929:175126. [PMID: 35779623 DOI: 10.1016/j.ejphar.2022.175126] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/09/2022] [Accepted: 06/24/2022] [Indexed: 01/23/2023]
Abstract
PURPOSE Several studies have reported that colchicine attenuates cardiac inflammation and improves cardiac function in myocardial infarction and atrial fibrillation. However, no study has investigated its effect on heart failure with preserved ejection fraction (HFpEF). Hence, this study aimed to assess its efficacy in a high salt diet (HSD)-induced HFpEF rat model. METHODS A rat hypertension-induced HFpEF model was created by treating Dahl/SS salt-sensitive rats with an HSD for 6 weeks. Colchicine was given via gavage daily as treatment. Cardiac function and inflammation were assessed using echocardiography, histology, and ELISA. Furthermore, the expression levels of NLRP3 and NF-κB signaling pathways were examined. RESULTS Treatment with colchicine increased survival and attenuated cardiac dysfunction, as indicated by decreased echocardiographic E/A ratio and longer exercise endurance along with reduced ventricular fibrosis and remodeling in HSD-induced Dahl rats. The treatment also reduced cardiac oxidative stress and inflammatory cell infiltration, as inferred from lower mRNA expressions of TNFα and CCL2 as well as protein expressions of NLRP3 and NF-κB pathways. CONCLUSION The findings signify that colchicine plays a crucial role in alleviating systemic inflammation and NLRP3 inflammation activation as well as in attenuating cardiac dysfunction and fibrosis in HSD-induced HFpEF model. Colchicine, therefore, holds therapeutic potential for further clinical applications.
Collapse
Affiliation(s)
- Song Shen
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Junfeng Duan
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Jiaxin Hu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Yu Qi
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Lina Kang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Kun Wang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Jianzhou Chen
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Xiang Wu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China
| | - Biao Xu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China.
| | - Rong Gu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, PR China.
| |
Collapse
|
72
|
Bayes-Genis A, Cediel G, Domingo M, Codina P, Santiago E, Lupón J. Biomarkers in Heart Failure with Preserved Ejection Fraction. Card Fail Rev 2022; 8:e20. [PMID: 35815256 PMCID: PMC9253965 DOI: 10.15420/cfr.2021.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous disorder developing from multiple aetiologies with overlapping pathophysiological mechanisms. HFpEF diagnosis may be challenging, as neither cardiac imaging nor physical examination are sensitive in this situation. Here, we review biomarkers of HFpEF, of which the best supported are related to myocardial stretch and injury, including natriuretic peptides and cardiac troponins. An overview of biomarkers of inflammation, extracellular matrix derangements and fibrosis, senescence, vascular dysfunction, anaemia/iron deficiency and obesity is also provided. Finally, novel biomarkers from -omics technologies, including plasma metabolites and circulating microRNAs, are outlined briefly. A cardiac-centred approach to HFpEF diagnosis using natriuretic peptides seems reasonable at present in clinical practice. A holistic approach including biomarkers that provide information on the non-cardiac components of the HFpEF syndrome may enrich our understanding of the disease and may be useful in classifying HFpEF phenotypes or endotypes that may guide patient selection in HFpEF trials.
Collapse
Affiliation(s)
- Antoni Bayes-Genis
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; Department of Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Germán Cediel
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Domingo
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Pau Codina
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Evelyn Santiago
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Lupón
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
73
|
Del Buono MG, Damonte JI, Chiabrando JG, Markley R, Turlington J, Trankle CR, Kang L, Biondi-Zoccai G, Van Tassell BW, Abbate A. Effect of IL-1 Blockade With Anakinra on Heart Failure Outcomes in Patients With Anterior Versus Nonanterior ST Elevation Myocardial Infarction. J Cardiovasc Pharmacol 2022; 79:774-780. [PMID: 35170493 PMCID: PMC9177574 DOI: 10.1097/fjc.0000000000001240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/22/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Patients with ST elevation myocardial infarction (STEMI) are at risk of future heart failure (HF), particularly those with anterior STEMI. Interleukin-1 (IL-1) is a key mediator of the inflammatory response, and its blockade has emerged as a potential therapeutic strategy to prevent HF events. The aim of this analysis was to explore the effects of anakinra, an IL-1 receptor antagonist, on HF outcomes based on anterior versus nonanterior location STEMI and to explore whether this effect is mediated through the amelioration of left ventricular systolic function and cardiac remodeling. We pooled data from 3 early phase randomized clinical trials. The primary end point was a composite of all-cause death and new-onset HF at 1-year follow-up. The left anterior descending coronary artery as culprit vessel was used to identify anterior STEMI. We included 139 patients, 47 (34%) with anterior STEMI and 92 (66%) with nonanterior STEMI. Anakinra significantly reduced the combined end point of death or new-onset HF in patients with anterior STEMI [4 (13%) vs. 7 (42%), log-rank P value = 0.049] and in patients with nonanterior STEMI [3 (6%) vs. 9 (24%), log-rank P value = 0.014]. We found no significant differences comparing anakinra versus placebo in interval changes in left ventricular ejection fraction and volumes in anterior and nonanterior STEMI. In conclusion, anakinra is associated with a reduction of HF events in patients with STEMI, irrespective of anterior or nonanterior location, or of changes in left ventricular ejection fraction or cardiac remodeling.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Juan Ignacio Damonte
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Juan Guido Chiabrando
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Roshanak Markley
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jeremy Turlington
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Cory R. Trankle
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Benjamin W. Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
74
|
Majmundar M, Kansara T, Park H, Ibarra G, Marta Lenik J, Shah P, Kumar A, Doshi R, Zala H, Chaudhari S, Kalra A. Absolute lymphocyte count as a predictor of mortality and readmission in heart failure hospitalization. IJC HEART & VASCULATURE 2022; 39:100981. [PMID: 35281758 PMCID: PMC8904225 DOI: 10.1016/j.ijcha.2022.100981] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/15/2022]
Abstract
Lymphopenia (<1500/mm3) was associated with 82% higher mortality in heart failure patients irrespective of ejection fraction. Lymphopenia was a good predictor of all-cause readmission in heart failure patients with reduced ejection fraction. Due to cost-effectiveness, easy availability, and ability to predict outcomes in the short-term and medium-term, lymphopenia can be a valuable tool in the mortality, readmission prediction model of heart failure.
Background There is renewed interest in pursuing frugal and readily available laboratory markers to predict mortality and readmission in heart failure. We aim to determine the relationship between absolute lymphocyte count (ALC) and clinical outcomes in patients with heart failure hospitalization. Methods This was a retrospective cohort study of patients with heart failure. Patients were divided into two groups based on ALC, less than or equal to 1500 cells/mm3 and > 1500 cells/ mm3. The primary outcome was all-cause mortality. We did subgroup analysis based on ejection fraction and studied the association between ALC categories and clinical outcomes. Both ALC groups are matched by propensity score, outcomes were analyzed by Cox regression, and estimates are presented in hazard ratios (HR) and 95% confidence intervals (CI). Results We included 1029 patients in the pre-matched cohort and 766 patients in the propensity-score matched cohort. The median age was 64 years (IQR, 54–75), and 60.78% were male. In the matched cohort, ALC less than or equal to 1500 cells/mm3 had a higher risk of mortality compared with ALC > 1500 cells/mm3 (HR 1.51, 95% CI: 1.17–1.95; P = 0.002). These results were reproducible in subgroups of heart failure. When ALC was divided into four groups based on their levels, the lowest group of ALC had the highest risk of mortality. Conclusions In patients with heart failure and both subgroups, ALC less than or equal to 1500 cells/mm3 had a higher risk of mortality. Patients in lower groups of the ALC categories had a higher risk of mortality.
Collapse
Affiliation(s)
- Monil Majmundar
- Department of Cardiology, Maimonides Medical Center, Brooklyn, NY, USA
| | - Tikal Kansara
- Department of Internal Medicine, Cleveland Clinic Union Hospital, OH, USA
| | - Hansang Park
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital, NY, USA
| | - Gabriel Ibarra
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital, NY, USA
| | - Joanna Marta Lenik
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital, NY, USA
| | - Palak Shah
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital, NY, USA
| | - Ashish Kumar
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, USA
| | - Rajkumar Doshi
- Department of Cardiology, St. Joseph's Medical Center, NJ, USA
| | - Harshvardhan Zala
- Department of Clinical Research, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Shobhana Chaudhari
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital, NY, USA
| | - Ankur Kalra
- Division of Cardiovascular Medicine, Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
75
|
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
|
76
|
Prausmüller S, Spinka G, Stasek S, Arfsten H, Bartko PE, Goliasch G, Hülsmann M, Pavo N. Neutrophil Activation/Maturation Markers in Chronic Heart Failure with Reduced Ejection Fraction. Diagnostics (Basel) 2022; 12:444. [PMID: 35204534 PMCID: PMC8871325 DOI: 10.3390/diagnostics12020444] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Neutrophils are critically involved in the immune response. Inflammatory stimuli alter the expression status of their surface molecule toolset, while inflammation-stimulated granulopoiesis might also influence their maturation status. Data on neutrophil status in heart failure with reduced ejection fraction (HFrEF) are scarce. The present study aims to evaluate the role of neutrophil CD11b, CD66b and CD64 expression in HFrEF. METHODS A total of 135 HFrEF patients and 43 controls were recruited. Mean fluorescence intensity of the activation/maturation markers CD11b, CD66b and CD64 was measured on neutrophils by flow cytometry. CD10 (neprilysin) expression was simultaneously determined. RESULTS Neutrophil CD64 expression was higher in HFrEF compared with controls, while CD11b/CD66b levels were similar. Neutrophil CD11b and CD66b showed a significant direct correlation to neutrophil CD10 expression (rs = 0.573, p < 0.001 and rs = 0.184, p = 0.033). Neutrophil CD11b and CD66b correlated inversely with heart failure severity reflected by NT-proBNP and NYHA class (NT-proBNP: rs = -0.243, p = 0.005 and rs = -0.250, p = 0.004; NYHA class: p = 0.032 and p = 0.055), whereas no association for CD64 could be found. Outcome analysis did not reveal a significant association between the expression of CD11b, CD66b and CD64 and all-cause mortality (p = ns). CONCLUSIONS The results underline the potential role of neutrophils in HFrEF disease pathophysiology and risk stratification and should stimulate further research, characterizing subpopulations of neutrophils and searching for key molecules involved in the downward spiral of inflammation and heart failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Martin Hülsmann
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (G.S.); (S.S.); (H.A.); (P.E.B.); (G.G.); (N.P.)
| | | |
Collapse
|
77
|
Park E, Griffin J, Bathon JM. Myocardial Dysfunction and Heart Failure in Rheumatoid Arthritis. Arthritis Rheumatol 2022; 74:184-199. [PMID: 34523821 PMCID: PMC8795468 DOI: 10.1002/art.41979] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/24/2021] [Accepted: 09/09/2021] [Indexed: 02/03/2023]
Abstract
Rheumatoid arthritis (RA) patients have almost twice the risk of heart failure (HF) as individuals without RA, even with adjustment for the presence of ischemic heart disease. Moreover, RA patients remain at a 2-fold higher risk of mortality from HF compared to non-RA patients. These observations suggest that RA-specific inflammatory pathways are significant contributors to this increased risk of HF. Herein we summarize the epidemiology of HF in RA patients, the differences in myocardial structure or function between RA patients and non-RA patients without clinical signs of HF, and data on the role of systemic and local inflammation in RA HF pathophysiology. We also discuss the impact of subduing inflammation through the use of RA disease-modifying therapies on HF and myocardial structure and function, emphasizing gaps in the literature and areas needing further research.
Collapse
Affiliation(s)
- Elizabeth Park
- Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, New York
| | - Jan Griffin
- Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, New York
| | - Joan M Bathon
- Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, New York
| |
Collapse
|
78
|
Boulmpou A, Theodorakopoulou MP, Alexandrou ME, Boutou AK, Papadopoulos CE, Pella E, Sarafidis P, Vassilikos V. Meta-analysis addressing the impact of cardiovascular-acting medication on peak oxygen uptake of patients with HFpEF. Heart Fail Rev 2022; 27:609-623. [DOI: 10.1007/s10741-021-10207-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 11/28/2022]
|
79
|
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: 38] [Impact Index Per Article: 12.7] [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
|
80
|
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
|
81
|
Olsen MB, Gregersen I, Sandanger Ø, Yang K, Sokolova M, Halvorsen BE, Gullestad L, Broch K, Aukrust P, Louwe MC. Targeting the Inflammasome in Cardiovascular Disease. JACC Basic Transl Sci 2022; 7:84-98. [PMID: 35128212 PMCID: PMC8807732 DOI: 10.1016/j.jacbts.2021.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 01/10/2023]
Abstract
The pathogenesis of cardiovascular disease (CVD) is complex and multifactorial, and inflammation plays a central role. Inflammasomes are multimeric protein complexes that are activated in a 2-step manner in response to infection or tissue damage. Upon activation the proinflammatory cytokines, interleukins-1β and -18 are released. In the last decade, the evidence that inflammasome activation plays an important role in CVD development became stronger. We discuss the role of different inflammasomes in the pathogenesis of CVD, focusing on atherosclerosis and heart failure. This review also provides an overview of existing experimental studies and clinical trials on inflammasome inhibition as a therapeutic target in these disorders.
Collapse
Key Words
- ACS, acute coronary syndrome
- AIM2, absent in melanoma 2
- ASC, apoptosis associated speck-like protein
- ATP, adenosine triphosphate
- CAD, coronary artery disease
- CRP, C-reactive protein
- CVD, cardiovascular disease
- DAMP, damage associated molecular pattern
- GSDMD, gasdermin-D
- GSDMD-NT, gasdermin-D N-terminal
- HF, heart failure
- HFpEF, HF with preserved ejection fraction
- HFrEF, HF with reduced ejection fraction
- IL, interleukin
- IL-1
- LDL, low-density lipoprotein
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MI, myocardial infarction
- NF-κB, nuclear factor κB
- NLR, NOD-like receptor
- NLRP3
- NLRP3, NOD-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- PRR, pattern recognition receptor
- STEMI, ST-elevation myocardial infarction
- TLR, toll-like receptor
- atherosclerosis
- cardiovascular disease
- heart failure
- inflammasome
Collapse
Affiliation(s)
- Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Øystein Sandanger
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Dermatology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Marina Sokolova
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Bente E. Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mieke C. Louwe
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
82
|
Toldo S, Mezzaroma E, Buckley LF, Potere N, Di Nisio M, Biondi-Zoccai G, Van Tassell BW, Abbate A. Targeting the NLRP3 inflammasome in cardiovascular diseases. Pharmacol Ther 2021; 236:108053. [PMID: 34906598 PMCID: PMC9187780 DOI: 10.1016/j.pharmthera.2021.108053] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/21/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023]
Abstract
The NACHT, leucine-rich repeat (LRR), and pyrin domain (PYD)-containing protein 3 (NLRP3) inflammasome is an intracellular sensing protein complex that plays a major role in innate immunity. Following tissue injury, activation of the NLRP3 inflammasome results in cytokine production, primarily interleukin(IL)-1β and IL-18, and, eventually, inflammatory cell death - pyroptosis. While a balanced inflammatory response favors damage resolution and tissue healing, excessive NLRP3 activation causes detrimental effects. A key involvement of the NLRP3 inflammasome has been reported across a wide range of cardiovascular diseases (CVDs). Several pharmacological agents selectively targeting the NLRP3 inflammasome system have been developed and tested in animals and early phase human studies with overall promising results. While the NLRP3 inhibitors are in clinical development, multiple randomized trials have demonstrated the safety and efficacy of IL-1 blockade in atherothrombosis, heart failure and recurrent pericarditis. Furthermore, the non-selective NLRP3 inhibitor colchicine has been recently shown to significantly reduce cardiovascular events in patients with chronic coronary disease. In this review, we will outline the mechanisms driving NLRP3 assembly and activation, and discuss the pathogenetic role of the NLRP3 inflammasome in CVDs, providing an overview of the current and future therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Stefano Toldo
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Eleonora Mezzaroma
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Pharmacotherapy and Outcome Studies, Virginia Commonwealth University, Richmond, VA, USA
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicola Potere
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marcello Di Nisio
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Pharmacotherapy and Outcome Studies, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
83
|
de Alencar AKN, Wang H, de Oliveira GMM, Sun X, Zapata-Sudo G, Groban L. Crossroads between Estrogen Loss, Obesity, and Heart Failure with Preserved Ejection Fraction. Arq Bras Cardiol 2021; 117:1191-1201. [PMID: 34644788 PMCID: PMC8757160 DOI: 10.36660/abc.20200855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/16/2020] [Accepted: 01/27/2021] [Indexed: 11/24/2022] Open
Abstract
The prevalence of obesity and heart failure with preserved ejection fraction (HFpEF) increases significantly in postmenopausal women. Although obesity is a risk factor for left ventricular diastolic dysfunction (LVDD), the mechanisms that link the cessation of ovarian hormone production, and particularly estrogens, to the development of obesity, LVDD, and HFpEF in aging females are unclear. Clinical, and epidemiologic studies show that postmenopausal women with abdominal obesity (defined by waist circumference) are at greater risk for developing HFpEF than men or women without abdominal obesity. The study presents a review of clinical data that support a mechanistic link between estrogen loss plus obesity and left ventricular remodeling with LVDD. It also seeks to discuss potential cell and molecular mechanisms for estrogen-mediated protection against adverse adipocyte cell types, tissue depots, function, and metabolism that may contribute to LVDD and HFpEF.
Collapse
Affiliation(s)
| | - Hao Wang
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
- Wake Forest School of MedicineWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Internal Medicine-Section of Molecular Medicine, Winston-Salem, North Carolina - Estados Unidos da América
| | - Gláucia Maria Moraes de Oliveira
- Universidade Federal do Rio de JaneiroDepartamento de Clínica MédicaFaculdade de MedicinaRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Departamento de Clínica Médica, Faculdade de Medicina, Rio de Janeiro, RJ - Brasil
| | - Xuming Sun
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
| | - Gisele Zapata-Sudo
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Instituto de Ciências Biomédicas, Rio de Janeiro, RJ - Brasil
- Universidade Federal do Rio de JaneiroInstituto de Cardiologia Edson SaadFaculdade de MedicinaRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Instituto de Cardiologia Edson Saad, Faculdade de Medicina, Rio de Janeiro, RJ - Brasil
| | - Leanne Groban
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
- Wake Forest School of MedicineWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Internal Medicine-Section of Molecular Medicine, Winston-Salem, North Carolina - Estados Unidos da América
| |
Collapse
|
84
|
Wu J, Dong E, Zhang Y, Xiao H. The Role of the Inflammasome in Heart Failure. Front Physiol 2021; 12:709703. [PMID: 34776995 PMCID: PMC8581560 DOI: 10.3389/fphys.2021.709703] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation promotes the development of heart failure (HF). The inflammasome is a multimeric protein complex that plays an essential role in the innate immune response by triggering the cleavage and activation of the proinflammatory cytokines interleukins (IL)-1β and IL-18. Blocking IL-1β with the monoclonal antibody canakinumab reduced hospitalizations and mortality in HF patients, suggesting that the inflammasome is involved in HF pathogenesis. The inflammasome is activated under various pathologic conditions that contribute to the progression of HF, including pressure overload, acute or chronic overactivation of the sympathetic system, myocardial infarction, and diabetic cardiomyopathy. Inflammasome activation is responsible for cardiac hypertrophy, fibrosis, and pyroptosis. Besides inflammatory cells, the inflammasome in other cardiac cells initiates local inflammation through intercellular communication. Some inflammasome inhibitors are currently being investigated in clinical trials in patients with HF. The current evidence suggests that the inflammasome is a critical mediator of cardiac inflammation during HF and a promising therapeutic target. The present review summarizes the recent advances in both basic and clinical research on the role of the inflammasome in HF.
Collapse
Affiliation(s)
- Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| |
Collapse
|
85
|
Korotaeva AA, Samoilova EV, Mindzaev DR, Nasonova SN, Zhirov IV, Tereschenko SN. Pro-inflammatory cytokines in chronic cardiac failure: state of problem. TERAPEVT ARKH 2021; 93:1389-1394. [DOI: 10.26442/00403660.2021.11.201170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 01/09/2023]
Abstract
Systemic inflammation is characterized by the induction of pro-inflammatory cytokines, the increased level of which in the blood of patients with chronic heart failure (CHF) correlates with unfavorable clinical outcomes. However, it is unclear whether pro-inflammatory cytokines are the cause or the consequence of the disease progression. CHF with preserved ejection fraction and CHF with reduced ejection fraction demonstrate different inflammatory features, which suggests different degrees of pro-inflammatory pathway activation. The review deals with participation of pro-inflammatory cytokines in pathophysiological processes of CHF development, emphasizing the role of interleukin-6 activation and the effects of accompanying diseases on the course of systemic inflammation. The search for new approaches to prevention and therapy of CHF remains actual. The review presents the results of clinical trials of targeted anti-cytokine therapy which have revealed difficulties in controlling inflammation under the conditions of CHF. Identification of specific pro-inflammatory pathways in CHF pathogenesis will allow one to control inflammatory cascades, thus providing a prospective therapeutic strategy.
Collapse
|
86
|
Wenzl FA, Ambrosini S, Mohammed SA, Kraler S, Lüscher TF, Costantino S, Paneni F. Inflammation in Metabolic Cardiomyopathy. Front Cardiovasc Med 2021; 8:742178. [PMID: 34671656 PMCID: PMC8520939 DOI: 10.3389/fcvm.2021.742178] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Overlapping pandemics of lifestyle-related diseases pose a substantial threat to cardiovascular health. Apart from coronary artery disease, metabolic disturbances linked to obesity, insulin resistance and diabetes directly compromise myocardial structure and function through independent and shared mechanisms heavily involving inflammatory signals. Accumulating evidence indicates that metabolic dysregulation causes systemic inflammation, which in turn aggravates cardiovascular disease. Indeed, elevated systemic levels of pro-inflammatory cytokines and metabolic substrates induce an inflammatory state in different cardiac cells and lead to subcellular alterations thereby promoting maladaptive myocardial remodeling. At the cellular level, inflammation-induced oxidative stress, mitochondrial dysfunction, impaired calcium handling, and lipotoxicity contribute to cardiomyocyte hypertrophy and dysfunction, extracellular matrix accumulation and microvascular disease. In cardiometabolic patients, myocardial inflammation is maintained by innate immune cell activation mediated by pattern recognition receptors such as Toll-like receptor 4 (TLR4) and downstream activation of the NLRP3 inflammasome and NF-κB-dependent pathways. Chronic low-grade inflammation progressively alters metabolic processes in the heart, leading to a metabolic cardiomyopathy (MC) phenotype and eventually to heart failure with preserved ejection fraction (HFpEF). In accordance with preclinical data, observational studies consistently showed increased inflammatory markers and cardiometabolic features in patients with HFpEF. Future treatment approaches of MC may target inflammatory mediators as they are closely intertwined with cardiac nutrient metabolism. Here, we review current evidence on inflammatory processes involved in the development of MC and provide an overview of nutrient and cytokine-driven pro-inflammatory effects stratified by cell type.
Collapse
Affiliation(s)
- Florian A Wenzl
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
87
|
Reina-Couto M, Pereira-Terra P, Quelhas-Santos J, Silva-Pereira C, Albino-Teixeira A, Sousa T. Inflammation in Human Heart Failure: Major Mediators and Therapeutic Targets. Front Physiol 2021; 12:746494. [PMID: 34707513 PMCID: PMC8543018 DOI: 10.3389/fphys.2021.746494] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/20/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammation has been recognized as a major pathophysiological contributor to the entire spectrum of human heart failure (HF), including HF with reduced ejection fraction, HF with preserved ejection fraction, acute HF and cardiogenic shock. Nevertheless, the results of several trials attempting anti-inflammatory strategies in HF patients have not been consistent or motivating and the clinical implementation of anti-inflammatory treatments for HF still requires larger and longer trials, as well as novel and/or more specific drugs. The present work reviews the different inflammatory mechanisms contributing to each type of HF, the major inflammatory mediators involved, namely tumor necrosis factor alpha, the interleukins 1, 6, 8, 10, 18, and 33, C-reactive protein and the enzymes myeloperoxidase and inducible nitric oxide synthase, and their effects on heart function. Furthermore, several trials targeting these mediators or involving other anti-inflammatory treatments in human HF are also described and analyzed. Future therapeutic advances will likely involve tailored anti-inflammatory treatments according to the patient's inflammatory profile, as well as the development of resolution pharmacology aimed at stimulating resolution of inflammation pathways in HF.
Collapse
Affiliation(s)
- Marta Reina-Couto
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
- Departamento de Medicina Intensiva, Centro Hospitalar e Universitário São João, Porto, Portugal
| | - Patrícia Pereira-Terra
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Janete Quelhas-Santos
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Carolina Silva-Pereira
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| | - António Albino-Teixeira
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| | - Teresa Sousa
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| |
Collapse
|
88
|
Elsanhoury A, Nelki V, Kelle S, Van Linthout S, Tschöpe C. Epicardial Fat Expansion in Diabetic and Obese Patients With Heart Failure and Preserved Ejection Fraction-A Specific HFpEF Phenotype. Front Cardiovasc Med 2021; 8:720690. [PMID: 34604353 PMCID: PMC8484763 DOI: 10.3389/fcvm.2021.720690] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with diverse etiologies and pathophysiological factors. Obesity and type 2 diabetes mellitus (T2DM), conditions that coexist frequently, induce a cluster of metabolic and non-metabolic signaling derangements which are in favor to induce inflammation, fibrosis, myocyte stiffness, all hallmarks of HFpEF. In contrast to other HFpEF risk factors, obesity and T2DM are often associated with the generation of enlarged epicardial adipose tissue (EAT). EAT acts as an endocrine tissue that may exacerbate myocardial inflammation and fibrosis via various paracrine and vasocrine signals. In addition, an abnormally large EAT poses mechanical stress on the heart via pericardial restrain. HFpEF patients with enlarged EAT may belong to a unique phenotype that can benefit from specific EAT-targeted interventions, including life-style modifications and pharmacologically via statins and fat modifying anti-diabetics drugs; like metformin, sodium-glucose cotransporter 2 inhibitors, or glucagon-like peptide-1 receptor agonists, respectively.
Collapse
Affiliation(s)
- Ahmed Elsanhoury
- Berlin Institute of Health at Charite (BIH), Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Vivian Nelki
- Department of Cardiology, Campus Virchow Klinikum (CVK), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Kelle
- Department of Internal Medicine/Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health at Charite (BIH), Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health at Charite (BIH), Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Department of Cardiology, Campus Virchow Klinikum (CVK), Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
89
|
Pandey A, Shah SJ, Butler J, Kellogg DL, Lewis GD, Forman DE, Mentz RJ, Borlaug BA, Simon MA, Chirinos JA, Fielding RA, Volpi E, Molina AJA, Haykowsky MJ, Sam F, Goodpaster BH, Bertoni AG, Justice JN, White JP, Ding J, Hummel SL, LeBrasseur NK, Taffet GE, Pipinos II, Kitzman D. Exercise Intolerance in Older Adults With Heart Failure With Preserved Ejection Fraction: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 78:1166-1187. [PMID: 34503685 PMCID: PMC8525886 DOI: 10.1016/j.jacc.2021.07.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/16/2022]
Abstract
Exercise intolerance (EI) is the primary manifestation of chronic heart failure with preserved ejection fraction (HFpEF), the most common form of heart failure among older individuals. The recent recognition that HFpEF is likely a systemic, multiorgan disorder that shares characteristics with other common, difficult-to-treat, aging-related disorders suggests that novel insights may be gained from combining knowledge and concepts from aging and cardiovascular disease disciplines. This state-of-the-art review is based on the outcomes of a National Institute of Aging-sponsored working group meeting on aging and EI in HFpEF. We discuss aging-related and extracardiac contributors to EI in HFpEF and provide the rationale for a transdisciplinary, "gero-centric" approach to advance our understanding of EI in HFpEF and identify promising new therapeutic targets. We also provide a framework for prioritizing future research, including developing a uniform, comprehensive approach to phenotypic characterization of HFpEF, elucidating key geroscience targets for treatment, and conducting proof-of-concept trials to modify these targets.
Collapse
Affiliation(s)
- Ambarish Pandey
- University of Texas Southwestern Medical Center, Dallas, Texas, USA. https://twitter.com/ambarish4786
| | - Sanjiv J Shah
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Dean L Kellogg
- University of Texas Health Science Center and GRECC, South Texas Veterans Affairs Health System, San Antonio, Texas, USA
| | | | - Daniel E Forman
- University of Pittsburgh and VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Robert J Mentz
- Duke Clinical Research Center, Durham, North Carolina, USA
| | | | - Marc A Simon
- University of Pittsburgh and VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | | | | | - Elena Volpi
- University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | | | | | - Flora Sam
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Bret H Goodpaster
- Advent Health Translational Research Institute, Orlando, Florida, USA
| | - Alain G Bertoni
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jamie N Justice
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Jingzhone Ding
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Scott L Hummel
- University of Michigan and the VA Ann Arbor Health System, Ann Arbor, Michigan, USA
| | | | | | | | - Dalane Kitzman
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
90
|
Martí-Carvajal AJ, De Sanctis JB, Dayer M, Martí-Amarista CE, Alegría E, Monge Martín D, Abd El Aziz M, Correa-Pérez A, Nicola S, Parise Vasco JM. Interleukin-receptor antagonist and tumor necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Hippokratia 2021. [DOI: 10.1002/14651858.cd014741] [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]
Affiliation(s)
- Arturo J Martí-Carvajal
- Facultad de Ciencias de la Salud Eugenio Espejo (Centro Cochrane Ecuador); Universidad UTE; Quito Ecuador
- Facultad de Medicina (Centro Cochrane Madrid); Universidad Francisco de Vitoria; Madrid Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia; Universidad de Carabobo; Valencia Venezuela
| | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine; Palacky University Olomouc, Faculty of Medicine and Dentistry; Olomouc Czech Republic
| | - Mark Dayer
- Department of Cardiology; Somerset NHS Foundation Trust; Taunton UK
| | | | - Eduardo Alegría
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
| | | | - Mohamed Abd El Aziz
- Internal medicine; Texas Tech University Health Sciences Center El PasoPaul L. Foster School of Medicine; El Paso, Texas USA
| | - Andrea Correa-Pérez
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
- Clinical Biostatistics Unit; Hospital Universitario Ramón y Cajal (IRYCIS); Madrid Spain
| | - Susana Nicola
- Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC); Universidad UTE; Quito Ecuador
| | - Juan Marcos Parise Vasco
- Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC); Universidad UTE; Quito Ecuador
| |
Collapse
|
91
|
Mesquita T, Lin Y, Ibrahim A. Chronic low-grade inflammation in heart failure with preserved ejection fraction. Aging Cell 2021; 20:e13453. [PMID: 34382743 PMCID: PMC8441359 DOI: 10.1111/acel.13453] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently the predominant form of HF with a dramatic increase in risk with age. Low-grade inflammation, as occurs with aging (termed "inflammaging"), is a common feature of HFpEF pathology. Suppression of proinflammatory pathways has been associated with attenuated HFpEF disease severity and better outcomes. From this perspective, inflammasome signaling plays a central role in mediating chronic inflammation and cardiovascular disease progression. However, the causal link between the inflammasome-immune signaling axis on the age-dependent progression of HFpEF remains conjectural. In this review, we summarize the current understanding of the role of inflammatory pathways in age-dependent cardiac function decline. We will also evaluate recent advances and evidence regarding the inflammatory pathway in the pathophysiology of HFpEF, with special attention to inflammasome signaling.
Collapse
Affiliation(s)
- Thassio Mesquita
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| | - Yen‐Nien Lin
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
- Division of Cardiovascular MedicineDepartment of MedicineChina Medical University and HospitalTaichungTaiwan
| | - Ahmed Ibrahim
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| |
Collapse
|
92
|
Rosalia L, Ozturk C, Shoar S, Fan Y, Malone G, Cheema FH, Conway C, Byrne RA, Duffy GP, Malone A, Roche ET, Hameed A. Device-Based Solutions to Improve Cardiac Physiology and Hemodynamics in Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2021; 6:772-795. [PMID: 34754993 PMCID: PMC8559325 DOI: 10.1016/j.jacbts.2021.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022]
Abstract
Characterized by a rapidly increasing prevalence, elevated mortality and rehospitalization rates, and inadequacy of pharmaceutical therapies, heart failure with preserved ejection fraction (HFpEF) has motivated the widespread development of device-based solutions. HFpEF is a multifactorial disease of various etiologies and phenotypes, distinguished by diminished ventricular compliance, diastolic dysfunction, and symptoms of heart failure despite a normal ejection performance; these symptoms include pulmonary hypertension, limited cardiac reserve, autonomic imbalance, and exercise intolerance. Several types of atrial shunts, left ventricular expanders, stimulation-based therapies, and mechanical circulatory support devices are currently under development aiming to target one or more of these symptoms by addressing the associated mechanical or hemodynamic hallmarks. Although the majority of these solutions have shown promising results in clinical or preclinical studies, no device-based therapy has yet been approved for the treatment of patients with HFpEF. The purpose of this review is to discuss the rationale behind each of these devices and the findings from the initial testing phases, as well as the limitations and challenges associated with their clinical translation.
Collapse
Key Words
- BAT, baroreceptor activation therapy
- CCM, cardiac contractility modulation
- CRT, cardiac resynchronization therapy
- HF, heart failure
- HFmEF, heart failure with mid-range ejection fraction
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IASD, Interatrial Shunt Device
- LAAD, left atrial assist device
- LAP, left atrial pressure
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MCS, mechanical circulatory support
- NYHA, New York Heart Association
- PCWP, pulmonary capillary wedge pressure
- QoL, quality of life
- TAA, transapical approach
- atrial shunt devices
- electrostimulation
- heart failure devices
- heart failure with preserved ejection fraction
- left ventricular expanders
- mechanical circulatory support
- neuromodulation
Collapse
Affiliation(s)
- Luca Rosalia
- Health Sciences and Technology Program, Harvard–Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Caglar Ozturk
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Grainne Malone
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Faisal H. Cheema
- HCA Healthcare, Houston, Texas, USA
- University of Houston, College of Medicine, Houston, Texas, USA
| | - Claire Conway
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Robert A. Byrne
- Department of Cardiology, Mater Private Hospital, Dublin, Ireland
- Cardiovascular Research, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Garry P. Duffy
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, Galway, Ireland
- Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
- Advanced Materials for Biomedical Engineering and Regenerative Medicine, Trinity College Dublin, and National University of Ireland Galway, Galway, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Andrew Malone
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ellen T. Roche
- Health Sciences and Technology Program, Harvard–Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
93
|
Lin Y, Fu S, Yao Y, Li Y, Zhao Y, Luo L. Heart failure with preserved ejection fraction based on aging and comorbidities. J Transl Med 2021; 19:291. [PMID: 34229717 PMCID: PMC8259336 DOI: 10.1186/s12967-021-02935-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/10/2021] [Indexed: 12/25/2022] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a leading cause of hospitalizations and mortality when diagnosed at the age of ≥ 65 years. HFpEF represents multifactorial and multisystemic syndrome and has different pathophysiology and phenotypes. Its diagnosis is difficult to be established based on left ventricular ejection fraction and may benefit from individually tailored approaches, underlying age-related changes and frequent comorbidities. Compared with the rapid development in the treatment of heart failure with reduced ejection fraction, HFpEF presents a great challenge and needs to be addressed considering the failure of HF drugs to improve its outcomes. Further extensive studies on the relationships between HFpEF, aging, and comorbidities in carefully phenotyped HFpEF subgroups may help understand the biology, diagnosis, and treatment of HFpEF. The current review summarized the diagnostic and therapeutic development of HFpEF based on the complex relationships between aging, comorbidities, and HFpEF.
Collapse
Affiliation(s)
- Ying Lin
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 572013, China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 572013, China.
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| | - Yao Yao
- Centre for the Study of Ageing and Human Development and Geriatrics Division, Medical School of Duke University, Durham, NC, 27708, USA
- Centre for Healthy Ageing and Development Studies, National School of Development, Peking University, Beijing, 100871, China
| | - Yulong Li
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yali Zhao
- Central Laboratory, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 572013, China.
| | - Leiming Luo
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| |
Collapse
|
94
|
Liu Z, Liu H, Deng Q, Sun C, He W, Zheng W, Tang R, Li W, Xie Q. Association Between Dietary Inflammatory Index and Heart Failure: Results From NHANES (1999-2018). Front Cardiovasc Med 2021; 8:702489. [PMID: 34307508 PMCID: PMC8292138 DOI: 10.3389/fcvm.2021.702489] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: To explore the relationship between dietary inflammatory index (DII) and heart failure (HF) in participants with cardiovascular and cerebrovascular diseases. Methods: NHANES (1998–2018) data were collected and used to assess the association of HF with DII. Twenty-four-hour dietary consumptions were used to calculate the scores of DII. Demographic characteristics and physical and laboratory examinations were collected for the comparison between HF and non-HF groups. Logistic regression analysis and random forest analysis were performed to calculate the odds rate and determine the potential beneficial dietary components in HF. Results: A total of 19,067 cardiac-cerebral vascular disease participants were categorized as HF (n = 1,382; 7.25%) and non-HF (n = 17,685; 92.75%) groups. Heart failure participants had higher levels of DII score compared with those in the non-HF group (0.239 ± 1.702 vs. −0.145 ± 1.704, p < 0.001). Compared with individuals with T1 (DII: −3.884 to −0.570) of DII, those in T3 (DII: 1.019 to 4.598) had a higher level of total cholesterol (4.49 ± 1.16 vs. 4.75 ± 1.28 mmol/L, p < 0.01), globulin (29.92 ± 5.37 vs. 31.29 ± 5.84 g/L, p < 0.001), and pulse rate (69.90 ± 12.22 vs. 72.22 ± 12.77, p < 0.001) and lower levels of albumin (40.76 ± 3.52 vs. 39.86 ± 3.83 g/L, p < 0.001), hemoglobin (13.76 ± 1.65 vs. 13.46 ± 1.77 g/dl, p < 0.05), and hematocrit (40.83 ± 4.69 vs. 40.17 ± 5.01%, p < 0.05). The odds rates of HF for DII from the logistic regression were 1.140, 1.158, and 1.110 in models 1, 2, and 3, respectively. In addition, from the results of random forest analysis, dietary magnesium, fiber, and beta carotene may be essential in HF. Conclusion: Dietary inflammatory index was positively associated with HF in US adults, and dietary intervention might be a promising method in the therapy of HF.
Collapse
Affiliation(s)
- Zuheng Liu
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Haiyue Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Qinsheng Deng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Changqing Sun
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wangwei He
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wuyang Zheng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Rong Tang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Weihua Li
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qiang Xie
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
95
|
Abstract
Neurohormones and inflammatory mediators have effects in both the heart and the peripheral vasculature. In patients with heart failure (HF), neurohormonal activation and increased levels of inflammatory mediators promote ventricular remodeling and development of HF, as well as vascular dysfunction and arterial stiffness. These processes may lead to a vicious cycle, whereby arterial stiffness perpetuates further ventricular remodeling leading to exacerbation of symptoms. Although significant advances have been made in the treatment of HF, currently available treatment strategies slow, but do not halt, this cycle. The current treatment for HF patients involves the inhibition of neurohormonal activation, which can reduce morbidity and mortality related to this condition. Beyond benefits associated with neurohormonal blockade, other strategies have focused on inhibition of inflammatory pathways implicated in the pathogenesis of HF. Unfortunately, attempts to target inflammation have not yet been successful to improve prognosis of HF. Further work is required to interrupt key maladaptive mechanisms involved in disease progression.
Collapse
|
96
|
Insight into the Pro-inflammatory and Profibrotic Role of Macrophage in Heart Failure With Preserved Ejection Fraction. J Cardiovasc Pharmacol 2021; 76:276-285. [PMID: 32501838 DOI: 10.1097/fjc.0000000000000858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prevalence of heart failure (HF) with preserved ejection fraction (HFpEF) is higher than that of HF with reduced/midrange ejection fraction (HFrEF/HFmrEF). However, no evidence-based guidelines for managing HFpEF have been generated. The current body of knowledge indicates that fibrosis and inflammation are important components of the cardiac remodeling process in HFpEF. In addition, macrophages potentially play an important role in pro-inflammatory and profibrotic processes in HFpEF patients, whereas HFpEF comorbidities could be a driving force for systemic microvascular inflammation and endothelial dysfunction. Under such circumstances, macrophages reportedly contribute to inflammation and fibrosis through 3 phases namely, inflammation, repair, and resolution. Signal transduction pathway-targeted therapies using animal experiments have generated important discoveries and breakthroughs for understanding the underlying mechanisms of HFpEF. However, only a handful of studies have reported promising results using human trials. Further investigations are therefore needed to elucidate the exact mechanisms underlying HFpEF and immune-pathogenesis of cardiac fibrosis.
Collapse
|
97
|
Gavini MP, Mahmood A, Belenchia AM, Beauparlant P, Kumar SA, Ardhanari S, DeMarco VG, Pulakat L. Suppression of Inflammatory Cardiac Cytokine Network in Rats with Untreated Obesity and Pre-Diabetes by AT2 Receptor Agonist NP-6A4. Front Pharmacol 2021; 12:693167. [PMID: 34220518 PMCID: PMC8253363 DOI: 10.3389/fphar.2021.693167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity affects over 42% of the United States population and exacerbates heart disease, the leading cause of death in men and women. Obesity also increases pro-inflammatory cytokines that cause chronic tissue damage to vital organs. The standard-of-care does not sufficiently attenuate these inflammatory sequelae. Angiotensin II receptor AT2R is an anti-inflammatory and cardiovascular protective molecule; however, AT2R agonists are not used in the clinic to treat heart disease. NP-6A4 is a new AT2R peptide agonist with an FDA orphan drug designation for pediatric cardiomyopathy. NP-6A4 increases AT2R expression (mRNA and protein) and nitric oxide generation in human cardiovascular cells. AT2R-antagonist PD123319 and AT2RSiRNA suppress NP-6A4-effects indicating that NP-6A4 acts through AT2R. To determine whether NP-6A4 would mitigate cardiac damage from chronic inflammation induced by untreated obesity, we investigated the effects of 2-weeks NP-6A4 treatment (1.8 mg/kg delivered subcutaneously) on cardiac pathology of male Zucker obese (ZO) rats that display obesity, pre-diabetes and cardiac dysfunction. NP-6A4 attenuated cardiac diastolic and systolic dysfunction, cardiac fibrosis and cardiomyocyte hypertrophy, but increased myocardial capillary density. NP-6A4 treatment suppressed tubulointerstitial injury marker urinary β-NAG, and liver injury marker alkaline phosphatase in serum. These protective effects of NP-6A4 occurred in the presence of obesity, hyperinsulinemia, hyperglycemia, and hyperlipidemia, and without modulating blood pressure. NP-6A4 increased expression of AT2R (consistent with human cells) and cardioprotective erythropoietin (EPO) and Notch1 in ZO rat heart, but suppressed nineteen inflammatory cytokines. Cardiac miRNA profiling and in silico analysis showed that NP-6A4 activated a unique miRNA network that may regulate expression of AT2R, EPO, Notch1 and inflammatory cytokines, and mitigate cardiac pathology. Seventeen pro-inflammatory and pro-fibrotic cytokines that increase during lethal cytokine storms caused by infections such as COVID-19 were among the cytokines suppressed by NP-6A4 treatment in ZO rat heart. Thus, NP-6A4 activates a novel anti-inflammatory network comprised of 21 proteins in the heart that was not reported previously. Since NP-6A4's unique mode of action suppresses pro-inflammatory cytokine network and attenuates myocardial damage, it can be an ideal adjuvant drug with other anti-glycemic, anti-hypertensive, standard-of-care drugs to protect the heart tissues from pro-inflammatory and pro-fibrotic cytokine attack induced by obesity.
Collapse
Affiliation(s)
| | - Abuzar Mahmood
- Dalton Cardiovascular Research Center, Columbia, MO, United States.,Department of Medicine, Boston, MA, United States.,Harry S. Truman Memorial VA Hospital, Columbia, MO, United States
| | - Anthony M Belenchia
- Dalton Cardiovascular Research Center, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Paige Beauparlant
- Dalton Cardiovascular Research Center, Columbia, MO, United States.,Department of Medicine, Boston, MA, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | | | | | - Vincent G DeMarco
- Dalton Cardiovascular Research Center, Columbia, MO, United States.,Department of Medicine, Boston, MA, United States.,Harry S. Truman Memorial VA Hospital, Columbia, MO, United States
| | - Lakshmi Pulakat
- Dalton Cardiovascular Research Center, Columbia, MO, United States.,Department of Medicine, Boston, MA, United States.,Harry S. Truman Memorial VA Hospital, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States.,Tufts Medical Center and Department of Medicine, Molecular Cardiology Research Institute, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
98
|
Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2021; 18:400-423. [PMID: 33432192 PMCID: PMC8574228 DOI: 10.1038/s41569-020-00480-6] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 01/30/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) affects half of all patients with heart failure worldwide, is increasing in prevalence, confers substantial morbidity and mortality, and has very few effective treatments. HFpEF is arguably the greatest unmet medical need in cardiovascular disease. Although HFpEF was initially considered to be a haemodynamic disorder characterized by hypertension, cardiac hypertrophy and diastolic dysfunction, the pandemics of obesity and diabetes mellitus have modified the HFpEF syndrome, which is now recognized to be a multisystem disorder involving the heart, lungs, kidneys, skeletal muscle, adipose tissue, vascular system, and immune and inflammatory signalling. This multiorgan involvement makes HFpEF difficult to model in experimental animals because the condition is not simply cardiac hypertrophy and hypertension with abnormal myocardial relaxation. However, new animal models involving both haemodynamic and metabolic disease, and increasing efforts to examine human pathophysiology, are revealing new signalling pathways and potential therapeutic targets. In this Review, we discuss the cellular and molecular pathobiology of HFpEF, with the major focus being on mechanisms relevant to the heart, because most research has focused on this organ. We also highlight the involvement of other important organ systems, including the lungs, kidneys and skeletal muscle, efforts to characterize patients with the use of systemic biomarkers, and ongoing therapeutic efforts. Our objective is to provide a roadmap of the signalling pathways and mechanisms of HFpEF that are being characterized and which might lead to more patient-specific therapies and improved clinical outcomes.
Collapse
Affiliation(s)
- Sumita Mishra
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A. Kass
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,
| |
Collapse
|
99
|
Paulus WJ, Zile MR. From Systemic Inflammation to Myocardial Fibrosis: The Heart Failure With Preserved Ejection Fraction Paradigm Revisited. Circ Res 2021; 128:1451-1467. [PMID: 33983831 PMCID: PMC8351796 DOI: 10.1161/circresaha.121.318159] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In accordance with the comorbidity-inflammation paradigm, comorbidities and especially metabolic comorbidities are presumed to drive development and severity of heart failure with preserved ejection fraction through a cascade of events ranging from systemic inflammation to myocardial fibrosis. Recently, novel experimental and clinical evidence emerged, which strengthens the validity of the inflammatory/profibrotic paradigm. This evidence consists among others of (1) myocardial infiltration by immunocompetent cells not only because of an obesity-induced metabolic load but also because of an arterial hypertension-induced hemodynamic load. The latter is sensed by components of the extracellular matrix like basal laminin, which also interact with cardiomyocyte titin; (2) expression in cardiomyocytes of inducible nitric oxide synthase because of circulating proinflammatory cytokines. This results in myocardial accumulation of degraded proteins because of a failing unfolded protein response; (3) definition by machine learning algorithms of phenogroups of patients with heart failure with preserved ejection fraction with a distinct inflammatory/profibrotic signature; (4) direct coupling in mediation analysis between comorbidities, inflammatory biomarkers, and deranged myocardial structure/function with endothelial expression of adhesion molecules already apparent in early preclinical heart failure with preserved ejection fraction (HF stage A, B). This new evidence paves the road for future heart failure with preserved ejection fraction treatments such as biologicals directed against inflammatory cytokines, stimulation of protein ubiquitylation with phosphodiesterase 1 inhibitors, correction of titin stiffness through natriuretic peptide-particulate guanylyl cyclase-PDE9 (phosphodiesterase 9) signaling and molecular/cellular regulatory mechanisms that control myocardial fibrosis.
Collapse
Affiliation(s)
- Walter J Paulus
- Amsterdam University Medical Centers, The Netherlands (W.J.P.)
| | - Michael R Zile
- RHJ Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston (M.R.Z.)
| |
Collapse
|
100
|
Fragoulis GE, Soulaidopoulos S, Sfikakis PP, Dimitroulas T, D Kitas G. Effect of Biologics on Cardiovascular Inflammation: Mechanistic Insights and Risk Reduction. J Inflamm Res 2021; 14:1915-1931. [PMID: 34017189 PMCID: PMC8131071 DOI: 10.2147/jir.s282691] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/14/2021] [Indexed: 12/17/2022] Open
Abstract
It is increasingly recognized that atherosclerosis and consequently cardiovascular disease (CVD) are closely linked with inflammatory processes. The latter is in the center of the pathogenic mechanism underlying autoimmune rheumatic diseases (ARD). It follows then, that optimal control of inflammation in ARDs may lead to a decrease of the accompanied CVD risk. Major trials (eg, CANTOS, CIRT), aimed at examining the possible benefits of immunomodulatory treatments in CVD, demonstrated conflicting results. On the other hand, substantial evidence is accumulating about the possible beneficial effects of biologic disease modifying antirheumatic drugs (bDMARDs) in patients with ARDs, particularly those with rheumatoid arthritis (RA). It seems that bDMARDs (some more than others) alter the lipid profile in RA patients but do not adversely affect, in most cases, the TC/HDL ratio. Favorable effects are noted for arterial stiffness and endothelial function. This is reflected in the lower risk for CVD events, seen in observational studies of RA patients treated with bDMARDs. It should be stressed that more data exist for the TNF-inhibitors than for other bDMARDs, such as tocilizumab, abatacept and rituximab. As regards the spondyloarthropathies (SpA), data are less robust. For TNF-inhibitors, effects appear to be on par with those seen in RA but no conclusions can be drawn for newer biologic drugs used in SpA (eg, IL-17 blockers). Finally, there is accumulating evidence for a beneficial effect of immunosuppressive treatment in cardiac inflammation and function in several ARDs. Introduction of newer therapeutic options in clinical practice seem to have a positive impact on CVD in the setting of ARD. This is probably due to better control of inflammation, but direct improvement in vascular pathology is also a valid hypothesis. Most data are derived from observational studies and, therefore, randomized controlled trials are needed to assess the possible favorable effect of bDMARDs on CVD outcomes.
Collapse
Affiliation(s)
- George E Fragoulis
- Rheumatology Unit, Joint Rheumatology Program, Medical School, First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, 115 27, Greece
| | - Stergios Soulaidopoulos
- First Department of Cardiology, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, 115 27, Greece
| | - Petros P Sfikakis
- Rheumatology Unit, Joint Rheumatology Program, Medical School, First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, 115 27, Greece
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, Hippokration Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 546 41, Greece
| | - George D Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, DY1 2HQ, UK.,Arthritis Research UK Epidemiology Unit, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|