1
|
Li YL, Li Y, Tu H, Evans AJ, Patel TA, Zheng H, Patel KP. Stellate Ganglia: A Key Therapeutic Target for Malignant Ventricular Arrhythmia in Heart Disease. Circ Res 2025; 136:1049-1069. [PMID: 40273204 PMCID: PMC12026290 DOI: 10.1161/circresaha.124.325384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Malignant ventricular arrhythmias (VAs), such as ventricular tachycardia and ventricular fibrillation, are the cause of approximately half a million deaths per year in the United States, which is a common lethal event in heart disease, such as hypertension, catecholaminergic polymorphic ventricular tachycardia, takotsubo cardiomyopathy, long-QT syndrome, and progressing into advanced heart failure. A common characteristic of these heart diseases, and the subsequent development of VAs, is the overactivation of the sympathetic nervous system. Current treatments for VAs in these heart diseases, such as β-adrenergic receptor blockers and cardiac sympathetic ablation, aim at inhibiting cardiac sympathetic overactivation. However, these treatments do not translate into becoming efficacious as long-term suppressors of ventricular tachycardia/ventricular fibrillation events. As a key regulatory component in the heart, cardiac postganglionic sympathetic neurons residing in the stellate ganglia (SGs) release neurotransmitters (such as norepinephrine and NPY [neuropeptide Y]) to perform their regulatory role in dictating cardiac function. Growing evidence from animal experiments and clinical studies has demonstrated that the remodeling of the SG may be intimately involved in malignant arrhythmogenesis. This identifies the SG as a key potential therapeutic target for the treatment of malignant VAs in heart disease. Therefore, this review summarizes the role of SG in ventricular arrhythmogenesis and updates the novel targeting of SG for clinical treatment of VAs in heart disease.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yu Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tapan A. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Kaushik P. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
2
|
Chen JY, Pan HC, Wu VC. Association of Angiotensin Receptor-Neprilysin Inhibitors Use and Better Cardiorenal Outcomes in Patients With Heart Failure and Acute Kidney Disease. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07698-x. [PMID: 40266448 DOI: 10.1007/s10557-025-07698-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE Angiotensin receptor-neprilysin inhibitors (ARNi) have been shown to improve cardiovascular outcomes in heart failure (HF) patients. However, their impact on HF patients with concurrent acute kidney disease (AKD) remains underexplored. This study investigated the outcomes of ARNi compared to angiotensin-converting enzyme inhibitors (ACEi) in HF patients with AKD. METHODS The study included 20,009 hospitalized HF and AKD patients who underwent dialysis during hospitalization, recovered from dialysis within 90 days after discharge, and were followed until November 30, 2022, using data from TriNetX. The study period began in July 2015, coinciding with the availability of ARNi in the market. Propensity score matching (1:1) was applied to balance ARNi and ACEi groups. Adjusted hazard ratios (aHR) with 95% confidence intervals (CI) were calculated to assess the risks of mortality, major adverse kidney events (MAKE), readmission and major adverse cardiac events (MACE). The follow-up period was conducted with a maximum duration of 5 years. RESULTS A total of 20,009 AKD patients (mean [SD] age, 59.1 [12.2] years) were enrolled, of whom 21.9% received ARNi, with a median follow-up of 2.3 years. After matching, 4391 patients (mean age, 58.6 years; male, 67.9%) were identified in both the ARNi and control groups. ARNi users exhibited a significantly lower risk of mortality (aHR, 0.32, 95% CI 0.13-0.80, p = 0.01), MAKE (aHR, 0.58, 95% CI 0.51-0.66, p < 0.01 ), and readmission (aHR, 0.61, 95% CI 0.55-0.68, p <0.01) versus controls. However, no significant difference in the risk of MACE was observed between the two groups (aHR, 0.94, 95% CI 0.82-1.09, p = 0.78). Subgroup analysis revealed ARNi users, when concomitantly treated with mineralocorticoids, diuretics, or beta-blockers had significantly lower risks of mortality, readmission, and MAKE than the control group. In addition, ARNi significantly reduced mortality and MAKE in patients with GFR 30-60 mL/min/1.73 m2, irrespective of proteinuria status. However, no significant benefit was observed in patients with GFR <30 mL/min/1.73 m2. CONCLUSIONS In HF patients with AKD, ARNi was associated with reduced all-cause mortality, MAKE, and readmission risks compared to ACEi, particularly with concurrent mineralocorticoids, diuretics, or beta-blockers. Future research is necessary to further investigate the impact of ARNi on outcomes in patients with HF and AKD.
Collapse
Affiliation(s)
- Jui-Yi Chen
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Heng-Chih Pan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- NSARF (National Taiwan University Hospital Study Group of ARF) and TAIPAI, (Taiwan Primary Aldosteronism Investigators), Taipei, Taiwan.
| |
Collapse
|
3
|
Noujaim SF, Dobrev D. Non-neuronal ventricular cardiomyocyte-located nicotinergic acetylcholine receptors cause remodelling and arrhythmias. Eur Heart J 2025; 46:1247-1249. [PMID: 39842853 PMCID: PMC11959178 DOI: 10.1093/eurheartj/ehae845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Affiliation(s)
- Sami F Noujaim
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Beura SK, Panigrahi AR, Yadav P, Kulkarni PP, Lakhanpal V, Singh B, Singh SK. Role of Thrombosis in Neurodegenerative Diseases: An Intricate Mechanism of Neurovascular Complications. Mol Neurobiol 2025; 62:4802-4836. [PMID: 39482419 DOI: 10.1007/s12035-024-04589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | | | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Bhupinder Singh
- Department of Cardiology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
5
|
Pham T, Taberner AJ, Han J. Cardiac muscle contracts more efficiently at lower contraction frequencies. Exp Physiol 2025; 110:561-573. [PMID: 39888146 PMCID: PMC11963897 DOI: 10.1113/ep092367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
This study investigated how contraction frequency impacts the mechano-energetics of cardiac muscle performing mechanical work. Left-ventricular trabeculae were isolated from rat hearts and mounted in our work-loop calorimeter to assess their function at physiological temperature (37°C) across three stimulation frequencies, 2 Hz, 3.5 Hz and 5 Hz, in a randomised sequence. Each trabecula was subjected to two experimental protocols: work-loop contractions under a range of afterloads and isometric contractions under a range of muscle lengths. Two contraction protocols allowed the partition of the various components of energy expenditure during cardiac contraction. By simultaneously measuring force-length work and heat output, mechanical efficiency was calculated over a range of afterloads to determine the peak value. Our findings revealed that force production, activation heat (energy associated with Ca2+ cycling) and cross-bridge heat were unaffected by stimulation frequency. Trabeculae produced greater work output per twitch at 2 Hz and 3.5 Hz than at 5 Hz. Positive correlations among work output, shortening extent and mechanical efficiency were detected. From these findings it was concluded that the higher work output at lower frequencies is associated with greater extent of shortening, which correlates to greater mechanical efficiency. This study highlights the mechano-energetic advantage of ventricular trabeculae in terms of increased work output and energy efficiency gained from operating at lower contraction frequencies, supporting the notion that heart rate reduction produces direct benefits on cardiac energetics.
Collapse
Affiliation(s)
- Toan Pham
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
| | - Andrew J. Taberner
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
- Department of Engineering ScienceUniversity of AucklandAucklandNew Zealand
| | - June‐Chiew Han
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
6
|
Matsushita K, Harada K, Jimba T, Kohno T, Nakano H, Shindo A, Takei M, Kohsaka S, Yoshino H, Yamamoto T, Nagao K, Takayama M. Circadian variation in patients with acute heart failure with preserved ejection fraction. Minerva Cardiol Angiol 2025; 73:174-183. [PMID: 40135324 DOI: 10.23736/s2724-5683.24.06665-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
BACKGROUND The circadian system influences the pathophysiology of many cardiovascular diseases; however, circadian variations in patients with heart failure with preserved ejection fraction (HFpEF) are unknown. Thus, this study aimed to compare the clinical characteristics and risk factors for in-hospital mortality between patients with daytime- versus nighttime-onset HFpEF. METHODS This multicenter retrospective study included 3875 consecutive patients with acute HFpEF. Daytime and nighttime periods were defined as 6:00-17:59 and 18:00-5:59, respectively. Potential prognostic factors for in-hospital mortality were selected using univariable analyses. Those with P values of <0.10 were used in multivariable logistic regression analyses with forward selection (likelihood ratios) to identify significant prognostic factors. RESULTS The incidence of daytime-onset HFpEF was significantly lower but the in-hospital mortality was significantly higher than that of nighttime-onset HFpEF. Independent prognostic factors for in-hospital mortality in patients with daytime-onset HFpEF were age (odds ratio [OR], 1.057) and systolic blood pressure (OR: 0.979). In contrast, age (OR: 1.067), coexisting atrial fibrillation/flutter (OR: 2.023), systolic blood pressure (OR: 0.989), estimated glomerular filtration rate (OR: 0.971), treatment with diuretics (OR: 0.282), and treatment with beta-blockers (OR: 0.514) were independent prognostic factors in patients with nighttime-onset HFpEF. CONCLUSIONS The incidence of acute HFpEF exhibits circadian variations, and onset-related differences in clinical characteristics and prognostic factors for in-hospital mortality were identified. These findings may provide new insights for future research and guide individualized patient management strategies.
Collapse
Affiliation(s)
- Kenichi Matsushita
- Tokyo CCU Network Scientific Committee, Tokyo, Japan -
- Department of Cardiology, Saitama Medical University International Medical Center, Saitama, Japan -
| | | | | | - Takashi Kohno
- Tokyo CCU Network Scientific Committee, Tokyo, Japan
| | - Hiroki Nakano
- Tokyo CCU Network Scientific Committee, Tokyo, Japan
| | - Akito Shindo
- Tokyo CCU Network Scientific Committee, Tokyo, Japan
| | - Makoto Takei
- Tokyo CCU Network Scientific Committee, Tokyo, Japan
| | - Shun Kohsaka
- Tokyo CCU Network Scientific Committee, Tokyo, Japan
| | | | | | - Ken Nagao
- Tokyo CCU Network Scientific Committee, Tokyo, Japan
| | | |
Collapse
|
7
|
Peng JQ, Zhou MM, Xu L, Wang X, Tang YH, Liu Y. Targeted M-Channel Activation in the Left Stellate Ganglion Protects Against Ischemia-Induced Ventricular Arrhythmias in Canines. J Am Heart Assoc 2025; 14:e039059. [PMID: 40094190 DOI: 10.1161/jaha.124.039059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Acute myocardial ischemia (AMI)-triggered ventricular arrhythmias are closely linked to maladaptive sympathetic hyperactivity mediated via the left stellate ganglion (LSG). Although M-type potassium channels regulate neuronal excitability and hold therapeutic potential for neurological disorders, their role in intrinsic LSG neurons during ischemia remains unexplored. We investigated whether pharmacological M-channel activation in the LSG mitigates sympathetic overdrive and arrhythmogenesis in AMI. METHODS AND RESULTS Twenty-four beagles underwent LSG microinjection of either vehicle (n=12) or retigabine (M-channel activator, 50 μM; n=12) 30 minutes before AMI induction. We assessed (1) neural parameters (LSG electrophysiology, plasma norepinephrine levels, and c-fos+/tyrosine hydroxylase+ neuron expression); (2) cardiac electrophysiological parameters (beat-to-beat repolarization variability, spatial dispersion of effective refractory period and action potential duration, ventricular fibrillation threshold, and spontaneous ventricular arrhythmias incidence); and (3) autonomic and hemodynamic measures (heart rate variability and blood pressure). Retigabine pretreatment significantly suppressed ischemia-induced LSG hyperactivity and reduced sympathetic activation markers compared with controls. Treated animals exhibited attenuated repolarization variability and reduced electrophysiological heterogeneity in ischemic myocardium. The retigabine group demonstrated a higher ventricular fibrillation threshold (26.67±2.61 versus 12.33±1.76 voltage (V), P=0.0008) and a lower incidence of ventricular arrhythmias during AMI, with only negligible effects on baseline cardiac repolarization duration or LSG function before ischemia induction. CONCLUSIONS Targeted activation of LSG M-channels with retigabine stabilizes ischemia-induced sympathetic hyperactivity, promotes cardiac autonomic balance, preserves repolarization homogeneity, and ultimately mitigates arrhythmic susceptibility. These findings highlight ganglionic M-channel modulation as a translatable strategy to suppress neurogenic arrhythmogenesis in AMI.
Collapse
Affiliation(s)
- Jin-Qiang Peng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Ming-Min Zhou
- Department of Cardiology Sir Run Run Shaw Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Liao Xu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Xi Wang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Yan-Hong Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Yu Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| |
Collapse
|
8
|
Ali KA, Kerrigan DLG, Berkman JM. Influence of Primary Neurologic Disease on Cardiovascular Health in Females. Circ Res 2025; 136:618-627. [PMID: 40080534 DOI: 10.1161/circresaha.124.325545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Abstract
Neurocardiology is an interdisciplinary field that examines the complex interactions between the nervous and the cardiovascular systems, exploring how neurological processes, such as autonomic nervous system regulation and brain-heart communication impact heart function and contribute to cardiovascular health and disease. Although much of the focus on cardiovascular health has centered on traditional risk factors, the influence of the nervous system, especially in females, is increasingly recognized as a key determinant of cardiovascular outcomes. This article reviews existing literature on the neurological mechanisms that impact cardiovascular function in females. Specifically, we analyze how primary neurological disorders including cerebrovascular disease, headache disorders, and multiple sclerosis have specific downstream effects on cardiac function. By understanding the complex relationship between neurological and cardiovascular health, this review highlights the need for sex-specific approaches to prevention, diagnosis, and treatment of cardiovascular disease in females, ultimately encouraging the discovery of more effective care strategies and improving health outcomes.
Collapse
Affiliation(s)
- Khadija Awais Ali
- Department of Neurology, Stroke Divison, Vanderbilt University Medical Center, Nashville, TN
| | - Deborah L G Kerrigan
- Department of Neurology, Stroke Divison, Vanderbilt University Medical Center, Nashville, TN
| | - Jillian Molli Berkman
- Department of Neurology, Stroke Divison, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
9
|
Su H, Liu M, Wang S, Tian B, Hu H, Ma LK, Pan J. Co-administration of isoprenaline and phenylephrine induced a new HFrEF mouse model through activation of both SNS and RAAS. Front Cardiovasc Med 2025; 12:1531509. [PMID: 40129768 PMCID: PMC11931062 DOI: 10.3389/fcvm.2025.1531509] [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: 11/20/2024] [Accepted: 02/03/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction The pathogenesis of human heart failure is diverse, and a large number of animal models have emerged to better understand the development of heart failure in humans. Among them, there are several methods of induction in mouse heart failure models, each with its advantages and disadvantages. The use of drug induced heart failure models has greatly facilitated basic research and reduced the disadvantages of time-consuming and labor-intensive surgical modeling. Methods In our experiments, we used a combination of isoprenaline (ISO) and phenylephrine (PE) for modeling; we aimed to evaluate whether it is superior to conventional drug-induced models, especially those induced by isoprenaline alone. The ISO and PE were administered for 2 weeks by subcutaneous implantation with a micro-osmolar pump, and the mice were monitored dynamically for cardiac ultrasound and blood pressure. Results RNA sequencing of myocardial tissues after execution of mice further clarified that hypertrophy, fibrosis genes, Sympathetic nervous system (SNS), and Renin-angiotensin-aldosterone system (RAAS) pathways were upregulated. Discussion Therefore, we conclude that the ISO/PE-induced mouse heart failure model can activate both the SNS and RAAS, through the activation of both α-adrenergic receptor (α-AR) and β-adrenergic receptor (β-AR), which is more consistent with the development of human heart failure than the ISO-induced model and is expected to be a unique and representative heart failure modeling method.
Collapse
Affiliation(s)
- Huimin Su
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming Liu
- Department of Cardiology, The First Hospital of Xinjiang Medical University, Urumqi, China
| | - Siteng Wang
- Department of Cardiology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Beiduo Tian
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hao Hu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Li-Kun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianyuan Pan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
10
|
Ajijola OA, Aksu T, Arora R, Biaggioni I, Chen PS, De Ferrari G, Dusi V, Fudim M, Goldberger JJ, Green AL, Herring N, Khalsa SS, Kumar R, Lakatta E, Mehra R, Meyer C, Po S, Stavrakis S, Somers VK, Tan AY, Valderrabano M, Shivkumar K. Clinical neurocardiology: defining the value of neuroscience-based cardiovascular therapeutics - 2024 update. J Physiol 2025; 603:1781-1839. [PMID: 40056025 DOI: 10.1113/jp284741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 01/28/2025] [Indexed: 04/01/2025] Open
Abstract
The intricate role of the autonomic nervous system (ANS) in regulating cardiac physiology has long been recognized. Aberrant function of the ANS is central to the pathophysiology of cardiovascular diseases. It stands to reason, therefore, that neuroscience-based cardiovascular therapeutics hold great promise in the treatment of cardiovascular diseases in humans. A decade after the inaugural edition, this White Paper reviews the current state of understanding of human cardiac neuroanatomy, neurophysiology and pathophysiology in specific disease conditions, autonomic testing, risk stratification, and neuromodulatory strategies to mitigate the progression of cardiovascular diseases.
Collapse
Affiliation(s)
- Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tolga Aksu
- Division of Cardiology, Yeditepe University Hospital, Istanbul, Türkiye
| | - Rishi Arora
- Division of Cardiology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Italo Biaggioni
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peng-Sheng Chen
- Department of Cardiology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Gaetano De Ferrari
- Department of Medical Sciences, University of Turin, Italy and Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Torino, Italy
| | - Veronica Dusi
- Department of Medical Sciences, University of Turin, Italy and Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Torino, Italy
| | - Marat Fudim
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey J Goldberger
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander L Green
- Department of Clinical Neurosciences, John Radcliffe Hospital, and Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sahib S Khalsa
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rajesh Kumar
- Department of Neurobiology and the Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Edward Lakatta
- National Institute of Aging, National Institutes of Health, Bethesda, MD, USA
| | - Reena Mehra
- Division of Pulmonary Medicine, University of Washington, Seattle, WA, USA
| | - Christian Meyer
- Klinik für Kardiologie, Angiologie, Intensivmedizin, cNEP Research Consortium EVK, Düsseldorf, Germany
- Heart Rhythm Institute, Overland Park, KS, USA
| | - Sunny Po
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stavros Stavrakis
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Virend K Somers
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Alex Y Tan
- Division of Cardiology, Richmond Veterans Affairs Hospital, Richmond, VA, USA
| | - Miguel Valderrabano
- Methodist DeBakey Heart and Vascular Center and Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
11
|
Habecker BA, Bers DM, Birren SJ, Chang R, Herring N, Kay MW, Li D, Mendelowitz D, Mongillo M, Montgomery JM, Ripplinger CM, Tampakakis E, Winbo A, Zaglia T, Zeltner N, Paterson DJ. Molecular and cellular neurocardiology in heart disease. J Physiol 2025; 603:1689-1728. [PMID: 38778747 PMCID: PMC11582088 DOI: 10.1113/jp284739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
This paper updates and builds on a previous White Paper in this journal that some of us contributed to concerning the molecular and cellular basis of cardiac neurobiology of heart disease. Here we focus on recent findings that underpin cardiac autonomic development, novel intracellular pathways and neuroplasticity. Throughout we highlight unanswered questions and areas of controversy. Whilst some neurochemical pathways are already demonstrating prognostic viability in patients with heart failure, we also discuss the opportunity to better understand sympathetic impairment by using patient specific stem cells that provides pathophysiological contextualization to study 'disease in a dish'. Novel imaging techniques and spatial transcriptomics are also facilitating a road map for target discovery of molecular pathways that may form a therapeutic opportunity to treat cardiac dysautonomia.
Collapse
Affiliation(s)
- Beth A Habecker
- Department of Chemical Physiology & Biochemistry, Department of Medicine Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Susan J Birren
- Department of Biology, Volen Center for Complex Systems, Brandeis University, Waltham, MA, USA
| | - Rui Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Johanna M Montgomery
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | | | - Annika Winbo
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Nadja Zeltner
- Departments of Biochemistry and Molecular Biology, Cell Biology, and Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Vrabec T, Bender S, Chan S, Cha S, Haridas S, Hanna P, Ajijola OA, Shivkumar K, Smith C, Ardell JL. Bioelectronic block of stellate ganglia mitigates pacing-induced heterogeneous release of catecholamine and neuropeptide Y in the infarcted pig heart. J Physiol 2025; 603:2071-2088. [PMID: 39557601 PMCID: PMC11955864 DOI: 10.1113/jp286924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/23/2024] [Indexed: 11/20/2024] Open
Abstract
The sympathetic nervous system modulates cardiac contractile and electrophysiological function and contributes to adverse remodelling following myocardial infarction (MI). Axonal modulation therapy (AMT), directed at the sympathetic chain, blocks efferent sympathetic outflow to the heart and is a strategy to transiently and controllably mitigate chronic MI-associated sympatho-excitation. In porcine models, we evaluated scalable AMT, directed at the paravertebral chain, in blocking reflex-mediated pacing-induced sympatho-excitation post-MI. The level of sympatho-excitation was assessed by dynamic interstitial measurement of noradrenaline (NA) and neuropeptide Y (NPY). In anaesthetized normal (n = 5) and age-matched pigs 6 weeks post-MI induction (n = 10), we electrically stimulated the right sympathetic chain and determined levels of direct current block applied at the T1-T2 level sufficient to reduce the evoked changes in heart rate and/or contractility by 25-75%. Reflex-mediated neural release of NA and NPY into the interstitial space during programmed pacing (PP) was assessed using fast-scanning cyclic voltammetry and capacitive immunoprobes. Normal animals demonstrated homogeneous NA and NPY release profiles during PP. In contrast, for MI animals PP evoked differential NA and NPY release in remote and MI border zones of the left ventricle. Right-sided AMT mitigated NA and NPY pacing-induced release in the remote left ventricle with a positive correlation to increasing AMT levels. Pacing-induced NA and NPY release in the MI border zone was not mitigated by AMT. Differential effects of AMT on NA and NPY may underlie the anti-arrhythmic effects of partial stellate ganglion block in the setting of chronic MI. KEY POINTS: Programmed cardiac pacing evokes homogeneous noradrenaline (NA) and neuropeptide Y (NPY) release in equivalent areas (e.g. medial and lateral aspects) of the normal left ventricle. Programmed cardiac pacing evokes differential NA and NPY release in remote and border zones of the infarcted left ventricle. Axonal modulation therapy (AMT), using a graded direct current block applied to the stellate ganglia, can proportionally modulate cardiac sympathetic reflexes. Unilateral AMT mitigates NA and NPY release in remote left ventricular tissue, with release negatively correlated to increasing AMT levels. Heterogeneities in NA and NPY between the border and remote tissues are reduced by progressive AMT.
Collapse
Affiliation(s)
- Tina Vrabec
- Department of Physical Medicine & RehabilitationMetroHealth Medical CenterClevelandOHUSA
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
| | - Shane Bender
- Department of Physical Medicine & RehabilitationMetroHealth Medical CenterClevelandOHUSA
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
| | - Shyue‐An Chan
- Department of Physiology and BiophysicsCase Western Reserve UniversityClevelandOHUSA
| | - Steven Cha
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Sahil Haridas
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Peter Hanna
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Olujimi A. Ajijola
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Kalyanam Shivkumar
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Corey Smith
- Department of Physiology and BiophysicsCase Western Reserve UniversityClevelandOHUSA
| | - Jeffrey L. Ardell
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| |
Collapse
|
13
|
Zou F, Levine H, Mohanty S, Natale A, Di Biase L. Atrial Fibrillation-Induced Cardiomyopathy. Card Electrophysiol Clin 2025; 17:13-18. [PMID: 39893034 DOI: 10.1016/j.ccep.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Atrial fibrillation (AF) is one of the most prevalent cardiac arrhythmias in the world. Patients with AF also suffer from heart failure (HF). The relationship between AF and HF is often considered bidirectional and both share very similar risk factors. The mechanism of AF-induced cardiomyopathy lies in 3 distinct components: tachycardia-related cardiac dysfunction, heart rhythm irregularity, and AF-induced atrial myopathy. These components are mediated by calcium mishandling, neurohormonal activation, oxidative stress, myocardial supply-demand mismatch, and irreversible fibrosis and remodeling. Managing AF-induced cardiomyopathy should focus on early rhythm control to mitigate the development of irreversible remodeling and atrial myopathy.
Collapse
Affiliation(s)
- Fengwei Zou
- Department of Medicine/Cardiology, Montefiore-Einstein Center for Heart & Vascular Care, Bronx, NY 10467, USA
| | - Hannah Levine
- Department of Medicine/Cardiology, Montefiore-Einstein Center for Heart & Vascular Care, Bronx, NY 10467, USA
| | - Sanghamitra Mohanty
- Department of Clinical Cardiac Electrophysiology, Texas Cardiac Arrhythmia Institute at St. David's Medical Center, Austin, TX 78705, USA
| | - Andrea Natale
- Department of Clinical Cardiac Electrophysiology, Texas Cardiac Arrhythmia Institute at St. David's Medical Center, Austin, TX 78705, USA
| | - Luigi Di Biase
- Department of Medicine/Cardiology, Montefiore-Einstein Center for Heart & Vascular Care, Bronx, NY 10467, USA; Systems Head of Electrophysiology, Director of Arrhythmias at Montefiore Health System, Bronx, NY 10467, USA.
| |
Collapse
|
14
|
Gillman S, Zucker IH, Wang H. Heartificial intelligence: smart solutions for CHF: An A(I)MT approach. J Physiol 2025; 603:1909-1910. [PMID: 39726247 PMCID: PMC11955859 DOI: 10.1113/jp287953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Samuel Gillman
- Department of Genetics Cell Biology and AnatomyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Irving H. Zucker
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Han‐Jun Wang
- Department of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| |
Collapse
|
15
|
Zohora FT, Nazari MA, Sinusas AJ. Novel F-18-labeled Tracers of Sympathetic Function for Improved Risk Stratification and Clinical Outcomes. Curr Cardiol Rep 2025; 27:61. [PMID: 40009333 DOI: 10.1007/s11886-025-02197-9] [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] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE OF REVIEW This review summarizes the role of the novel 18F-labeled positron emission tomography (PET) sympathetic radiotracers for risk stratification in patients with ischemic heart disease. PET tracers have demonstrated prognostic value by characterizing myocardial sympathetic nerve density and by extension the extent of myocardial sympathetic denervation. The unique features of these PET radiotracers are discussed in relation to clinical application. RECENT FINDINGS Absolute quantification of sympathetic denervation has been possible with 18F-labeled PET tracers which outperform low ejection fraction (<35%) in predicting sudden cardiac death (SCD) and allow for more optimal risk stratification in patients with heart failure. This underscores their utility in selecting patients for preventable strategies with implantable cardioverter defibrillators (ICD). Appropriate candidate selection for ICD placement is a major priority as at present 80% of patients that die from SCD do not receive an ICD (potentially preventable mortality) while only 1 in 8 patients with an ICD receive a life-saving shock. Furthermore, 1 in 3 patients with ICDs receive inappropriate shocks. Thus, there is a pressing need to more appropriately select and exclude patients who will and will not benefit from ICD placement, respectively, as both suffer poor outcomes. Despite the clear prognostic benefit offered by prior PET sympathetic radiotracers in imaging myocardial sympathetic denervation, their short half-lives necessitated costly onsite cyclotron synthesis obviating their pragmatic clinical use. 18F-labeled radiotracers have a longer half-life allowing centralized synthesis and transport to their point of use. As such, 18F-labeled sympathetic radiotracers define an innovation and may offer a more affordable and clinically practical approach for evaluation of risk in patients with cardiovascular disease. 18F-labeled sympathetic radiotracers are currently available for evaluation and risk stratification of patients with ischemic heart disease and heart failure. These radiotracers may offer a more practical approach for selection of ICD placement and consequent prevention of SCD; a major, yet unmet need, in heart failure patients and those that suffer SCD at large. However, further development and clinical testing of these 18F-labeled sympathetic radiotracers is required.
Collapse
Affiliation(s)
- Fatema Tuj Zohora
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Matthew A Nazari
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Yale Translational Research Imaging Center, Yale University School of Medicine, DANA3, P.O. Box 208017, New Haven, CT, 06520-8017, USA.
| |
Collapse
|
16
|
Zisis M, Chondrogianni ME, Androutsakos T, Rantos I, Oikonomou E, Chatzigeorgiou A, Kassi E. Linking Cardiovascular Disease and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): The Role of Cardiometabolic Drugs in MASLD Treatment. Biomolecules 2025; 15:324. [PMID: 40149860 PMCID: PMC11940321 DOI: 10.3390/biom15030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
The link between cardiovascular disease (CVD) and metabolic dysfunction-associated steatotic liver disease (MASLD) is well-established at both the epidemiological and pathophysiological levels. Among the common pathophysiological mechanisms involved in the development and progression of both diseases, oxidative stress and inflammation, insulin resistance, lipid metabolism deterioration, hepatokines, and gut dysbiosis along with genetic factors have been recognized to play a pivotal role. Pharmacologic interventions with drugs targeting common modifiable cardiometabolic risk factors, such as T2DM, dyslipidemia, and hypertension, are a reasonable strategy to prevent CVD development and progression of MASLD. Recently, a novel drug for metabolic dysfunction-associated steatohepatitis (MASH), resmetirom, has shown positive effects regarding CVD risk, opening new opportunities for the therapeutic approach of MASLD and CVD. This review provides current knowledge on the epidemiologic association of MASLD to CVD morbidity and mortality and enlightens the possible underlying pathophysiologic mechanisms linking MASLD with CVD. The role of cardiometabolic drugs such as anti-hypertensive drugs, hypolipidemic agents, glucose-lowering medications, acetylsalicylic acid, and the thyroid hormone receptor-beta agonist in the progression of MASLD is also discussed. Metformin failed to prove beneficial effects in MASLD progression. Studies on the administration of thiazolinediones in MASLD suggest effectiveness in improving steatosis, steatohepatitis, and fibrosis, while newer categories of glucose-lowering agents such as GLP-1Ra and SGLT-2i are currently being tested for their efficacy across the whole spectrum of MASLD. Statins alone or in combination with ezetimibe have yielded promising results. The conduction of long-duration, large, high-quality, randomized-controlled trials aiming to assess by biopsy the efficacy of cardiometabolic drugs to reverse MASLD progression is of great importance.
Collapse
Affiliation(s)
- Marios Zisis
- Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Greece; (M.Z.); (I.R.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theodoros Androutsakos
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Ilias Rantos
- Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Greece; (M.Z.); (I.R.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, University of Athens Medical School, 11527 Athens, Greece;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
17
|
Shen S, Tian B, Zhang H, Wang YC, Li T, Cao Y. Heart Failure and Gut Microbiota: What Is Cause and Effect? RESEARCH (WASHINGTON, D.C.) 2025; 8:0610. [PMID: 39981296 PMCID: PMC11839986 DOI: 10.34133/research.0610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Emerging evidence highlights the central role of gut microbiota in maintaining physiological homeostasis within the host. Disruptions in gut microbiota can destabilize systemic metabolism and inflammation, driving the onset and progression of cardiometabolic diseases. In heart failure (HF), intestinal dysfunction may induce the release of endotoxins and metabolites, leading to dysbiosis and exacerbating HF through the gut-heart axis. Understanding the relationship between gut microbiota and HF offers critical insights into disease mechanisms and therapeutic opportunities. Current research highlights promising potential to improve patient outcomes by restoring microbiota balance. In this review, we summarize the current studies in understanding the gut microbiota-HF connection and discuss avenues for future investigation.
Collapse
Affiliation(s)
- Shichun Shen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Beiduo Tian
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Haizhu Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yu-Chen Wang
- Department of Medicine, Division of Cardiology, Department of Microbiology, Immunology and Molecular Genetics, and Department of Human Genetics,
University of California, Los Angeles, CA, USA
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics,
West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yang Cao
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
18
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
19
|
Zhao Y, Yang M, Liu Y, Wan Z, Chen M, He Q, Liao Y, Shuai P, Shi J, Guo S. Pathogenesis of cardiovascular diseases: effects of mitochondrial CF6 on endothelial cell function. Mol Cell Biochem 2025; 480:841-853. [PMID: 38985252 DOI: 10.1007/s11010-024-05065-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
Cardiovascular disease (CVD) stands as a predominant global cause of morbidity and mortality, necessitating effective and cost-efficient therapies for cardiovascular risk reduction. Mitochondrial coupling factor 6 (CF6), identified as a novel proatherogenic peptide, emerges as a significant risk factor in endothelial dysfunction development, correlating with CVD severity. CF6 expression can be heightened by CVD risk factors like mechanical force, hypoxia, or high glucose stimuli through the NF-κB pathway. Many studies have explored the CF6-CVD relationship, revealing elevated plasma CF6 levels in essential hypertension, atherosclerotic cardiovascular disease (ASCVD), stroke, and preeclampsia patients. CF6 acts as a vasoactive and proatherogenic peptide in CVD, inducing intracellular acidosis in vascular endothelial cells, inhibiting nitric oxide (NO) and prostacyclin generation, increasing blood pressure, and producing proatherogenic molecules, significantly contributing to CVD development. CF6 induces an imbalance in endothelium-dependent factors, including NO, prostacyclin, and asymmetric dimethylarginine (ADMA), promoting vasoconstriction, vascular remodeling, thrombosis, and insulin resistance, possibly via C-src Ca2+ and PRMT-1/DDAH-2-ADMA-NO pathways. This review offers a comprehensive exploration of CF6 in the context of CVD, providing mechanistic insights into its role in processes impacting CVD, with a focus on CF6 functions, intracellular signaling, and regulatory mechanisms in vascular endothelial cells.
Collapse
Affiliation(s)
- Yingying Zhao
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming Yang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Youren Liu
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhengwei Wan
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mengchun Chen
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiumei He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Liao
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Shuai
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Shujin Guo
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
20
|
Matsushita H, Saku K, Nishikawa T, Unoki T, Yokota S, Sato K, Morita H, Yoshida Y, Fukumitsu M, Uemura K, Kawada T, Kikuchi A, Yamaura K. Impact of right ventricular and pulmonary vascular characteristics on Impella hemodynamic support in biventricular heart failure: A simulation study. J Cardiol 2025; 85:100-107. [PMID: 39097144 DOI: 10.1016/j.jjcc.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Impella (Abiomed, Danvers, MA, USA) is a percutaneous ventricular assist device commonly used in cardiogenic shock, providing robust hemodynamic support, improving the systemic circulation, and relieving pulmonary congestion. Maintaining adequate left ventricular (LV) filling is essential for optimal hemodynamic support by Impella. This study aimed to investigate the impact of pulmonary vascular resistance (PVR) and right ventricular (RV) function on Impella-supported hemodynamics in severe biventricular failure using cardiovascular simulation. METHODS We used Simulink® (Mathworks, Inc., Natick, MA, USA) for the simulation, incorporating pump performance of Impella CP determined using a mock circulatory loop. Both systemic and pulmonary circulation were modeled using a 5-element resistance-capacitance network. The four cardiac chambers were represented by time-varying elastance with unidirectional valves. In the scenario of severe LV dysfunction (LV end-systolic elastance set at a low level of 0.4 mmHg/mL), we compared the changes in right (RAP) and left atrial pressures (LAP), total systemic flow, and pressure-volume loop relationship at varying degrees of RV function, PVR, and Impella flow rate. RESULTS The simulation results showed that under low PVR conditions, an increase in Impella flow rate slightly reduced RAP and LAP and increased total systemic flow, regardless of RV function. Under moderate RV dysfunction and high PVR conditions, an increase in Impella flow rate elevated RAP and excessively reduced LAP to induce LV suction, which limited the increase in total systemic flow. CONCLUSIONS PVR is the primary determinant of stable and effective Impella hemodynamic support in patients with severe biventricular failure.
Collapse
Affiliation(s)
- Hiroki Matsushita
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Keita Saku
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.
| | - Takuya Nishikawa
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Takashi Unoki
- Department of Cardiology and Intensive Care Unit, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Shohei Yokota
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Kei Sato
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Hidetaka Morita
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Yuki Yoshida
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Masafumi Fukumitsu
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Kazunori Uemura
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan; NTTR-NCVC Bio Digital Twin Center, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Toru Kawada
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Atsushi Kikuchi
- Department of Cardiology, Osaka General Medical Center, Suita, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
21
|
Dasari TW, Nagai M, Ewbank H, Chakraborty P, Po SS. Heart rate variability metrics and myocardial recovery in heart failure with reduced ejection fraction. Clin Auton Res 2025; 35:115-124. [PMID: 39312106 DOI: 10.1007/s10286-024-01064-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/02/2024] [Indexed: 03/27/2025]
Abstract
PURPOSE Autonomic dysregulation is observed in heart failure (HF) with reduced ejection fraction (HFrEF). Abnormal heart rate variability (HRV), a measure of such dysregulation, is associated with poor prognosis in HFrEF. It is unknown if novel HRV metrics normalize in the patients with recovered ejection fraction (HFrecEF) compared to persistent HFrEF. The aim of this study was to investigate novel HRV indexes in persistent HFrEF in comparison to HFrecEF METHODS: A standard 10-min electrocardiography measurement was performed in patients categorized in four groups: persistent HFrEF (n = 40), HFrecEF (n = 41), stage A HF (n = 73) and healthy controls (n = 40). RESULTS All HRV indexes were significantly different between the four groups. Specifically, novel metrics, such as higher parasympathetic nervous system (PNS) index and lower sympathetic nervous system (SNS) index, were observed in the HFrecEF group compared to the persistent HFrEF group. In multiple logistic regression analysis, higher PNS index (odds ratio [OR] 2.02, 95% confidence interval [CI] 1.17-3.49; p = 0.01) and lower SNS index (OR 0.68, 95% CI 0.52-0.87; p = 0.002) were associated with HFrecEF. Receiver operating characteristic analysis showed that the SNS index had the highest area under the curve (AUC), followed by the PNS index and mean heart rate for the HF phenotype regarding EF recovery (AUC = 0.71, 0.69 and 0.69, respectively). CONCLUSION Myocardial functional recovery in HFrEF is associated with improved parasympathetic activity and reduced sympathetic activity, as reflected in the PNS and SNS indexes. These novel metrics can be potentially used to aid in identifying recovered versus non-recovered phenotypes in patients with HFrEF.
Collapse
Affiliation(s)
- Tarun W Dasari
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Science Center, Andrews Academic Tower, Suite 5400, Oklahoma City, OK, 73104, USA.
| | - Michiaki Nagai
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Science Center, Andrews Academic Tower, Suite 5400, Oklahoma City, OK, 73104, USA.
| | - Hallum Ewbank
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Science Center, Andrews Academic Tower, Suite 5400, Oklahoma City, OK, 73104, USA
| | - Praloy Chakraborty
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Science Center, Andrews Academic Tower, Suite 5400, Oklahoma City, OK, 73104, USA
| | - Sunny S Po
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Science Center, Andrews Academic Tower, Suite 5400, Oklahoma City, OK, 73104, USA
| |
Collapse
|
22
|
Chang JWH, Ramchandra R. The sympathetic nervous system in heart failure with preserved ejection fraction. Heart Fail Rev 2025; 30:209-218. [PMID: 39438394 DOI: 10.1007/s10741-024-10456-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
The sympathetic nervous system (SNS) is a major mediator of cardiovascular physiology during exercise in healthy people. However, its role in heart failure with preserved ejection fraction (HFpEF), where exercise intolerance is a cardinal symptom, has remained relatively unexplored. The present review summarizes and critically explores the currently limited data on SNS changes in HFpEF patients with a particular emphasis on caveats of the data and the implications for its subsequent interpretation. While direct measurements of SNS activity in HFpEF patients is scarce, modest increases in resting levels of muscle sympathetic nerve activity are apparent, although this may be due to the co-morbidities associated with the syndrome rather than HFpEF per se. In addition, despite some evidence for dysfunctional sympathetic signaling in the heart, there is no clear evidence for elevated cardiac sympathetic nerve activity. The lack of a compelling prognostic benefit with use of β-blockers in HFpEF patients also suggests a lack of sympathetic hyperactivity to the heart. Similarly, while renal and splanchnic denervation studies have been performed in HFpEF patients, there is no concrete evidence that the sympathetic nerves innervating these organs exhibit heightened activity. Taken together, the totality of data suggests limited evidence for elevated sympathetic nerve activity in HFpEF and that any SNS perturbations that do occur are not universal to all HFpEF patients. Finally, how the SNS responds during exertion in HFpEF patients remains unknown and requires urgent investigation.
Collapse
Affiliation(s)
- Joshua W-H Chang
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Rohit Ramchandra
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
23
|
Ryu DG, Yu F, Yoon KT, Liu H, Lee SS. The Cardiomyocyte in Cirrhosis: Pathogenic Mechanisms Underlying Cirrhotic Cardiomyopathy. Rev Cardiovasc Med 2024; 25:457. [PMID: 39742234 PMCID: PMC11683693 DOI: 10.31083/j.rcm2512457] [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: 07/11/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 01/03/2025] Open
Abstract
Cirrhotic cardiomyopathy is defined as systolic and diastolic dysfunction in patients with cirrhosis, in the absence of any primary heart disease. These changes are mainly due to the malfunction or abnormalities of cardiomyocytes. Similar to non-cirrhotic heart failure, cardiomyocytes in cirrhotic cardiomyopathy demonstrate a variety of abnormalities: from the cell membrane to the cytosol and nucleus. At the cell membrane level, biophysical plasma membrane fluidity, and membrane-bound receptors such as the beta-adrenergic, muscarinic and cannabinoid receptors are abnormal either functionally or structurally. Other changes include ion channels such as L-type calcium channels, potassium channels, and sodium transporters. In the cytosol, calcium release and uptake processes are dysfunctional and the myofilaments such as myosin heavy chain and titin, are either functionally abnormal or have structural alterations. Like the fibrotic liver, the heart in cirrhosis also shows fibrotic changes such as a collagen isoform switch from more compliant collagen III to stiffer collagen I which also impacts diastolic function. Other abnormalities include the secondary messenger cyclic adenosine monophosphate, cyclic guanosine monophosphate, and their downstream effectors such as protein kinase A and G-proteins. Finally, other changes such as excessive apoptosis of cardiomyocytes also play a critical role in the pathogenesis of cirrhotic cardiomyopathy. The present review aims to summarize these changes and review their critical role in the pathogenesis of cirrhotic cardiomyopathy.
Collapse
Affiliation(s)
- Dae Gon Ryu
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
- Division of Gastroenterology, Yangsan Hospital, Pusan National University Faculty of Medicine, 50612 Pusan, Republic of Korea
| | - Fengxue Yu
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
- Telemedicine Center, Second Hospital of Hebei Medical University, 050004 Shijiazhuang, Hebei, China
| | - Ki Tae Yoon
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
- Division of Gastroenterology, Yangsan Hospital, Pusan National University Faculty of Medicine, 50612 Pusan, Republic of Korea
| | - Hongqun Liu
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| | - Samuel S. Lee
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
24
|
Lanspa MJ, Khan A, Lyons PG, Gong MN, Naqvi AA, Dugar S, Duggal A, Johnson NJ, Schoeneck JH, Smith L, Bose S, Shapiro NI, Shvilkina T, Groat D, Jacobs JR, Olsen TD, Cannavina S, Knox DB, Hirshberg EL, Self WH, Brown SM. Crystalloid Liberal or Vasopressors Early Resuscitation in Sepsis-Study of Treatment's Echocardiographic Mechanisms (CLOVERS-STEM). Crit Care Explor 2024; 6:e1182. [PMID: 39652431 PMCID: PMC11631020 DOI: 10.1097/cce.0000000000001182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
IMPORTANCE Receipt of fluid and vasopressors, common treatments in septic shock, may affect cardiac function. OBJECTIVES We sought to determine whether a liberal or restrictive fluid resuscitation strategy was associated with changes in cardiac function. DESIGN We prospectively studied a subset of patients enrolled in the Crystalloid Liberal or Vasopressors Early Resuscitation in Sepsis (CLOVERS) trial, performing echocardiography at baseline and at 24 hours after randomization. Among patients who had an echocardiogram performed at 24 hours, we measured left ventricular global longitudinal strain (LV GLS) and right ventricular free-wall longitudinal strain (RVFWLS). We performed linear regressions with dependent variables of LV GLS, change in LV GLS (ΔLV GLS), and RVFWLS using treatment assignment as an independent variable. We adjusted for ratio of early diastolic mitral inflow velocity to early diastolic mitral annulus velocity, mean arterial pressure, and history of congestive heart failure and myocardial infarction. SETTING Emergency department and ICUs. PATIENTS Adults with sepsis enrolled in the CLOVERS trial. MAIN OUTCOMES AND MEASURES We enrolled 180 patients. Our analytic cohort comprised 131 patients with an echocardiogram performed at 24 hours. We observed no differences between treatment arms with respect to demographic, clinical, or echocardiographic data at baseline. We observed no association between restrictive fluid assignment and LV GLS (coefficient, 1.22; p = 0.23), ΔLV GLS (-1.97; p = 0.27), or RVFWLS (2.33; p = 0.19). CONCLUSIONS AND RELEVANCE In a subset of patients enrolled in CLOVERS, we observed no association between receipt of fluid and vasopressors and short-term changes in cardiac function. Decreased enrollment may limit inferences.
Collapse
Affiliation(s)
- Michael J. Lanspa
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| | - Akram Khan
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Sciences University, Portland, OR
| | - Patrick G. Lyons
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Sciences University, Portland, OR
| | - Michelle N. Gong
- Division of Critical Care Medicine, Montefiore Medical Center, New York, NY
| | - Ali A. Naqvi
- Division of Critical Care Medicine, Montefiore Medical Center, New York, NY
| | - Siddharth Dugar
- Department of Critical Care, Cleveland Clinic, Cleveland, OH
| | - Abhijit Duggal
- Department of Critical Care, Cleveland Clinic, Cleveland, OH
| | | | - Jacob H. Schoeneck
- Department of Emergency Medicine, Wake Forest Baptist, Winston-Salem, NC
| | - Lane Smith
- Department of Emergency Medicine, Wake Forest Baptist, Winston-Salem, NC
| | - Somnath Bose
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Nathan I. Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Tatyana Shvilkina
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Danielle Groat
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| | - Jason R. Jacobs
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| | - Troy D. Olsen
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| | - Steven Cannavina
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| | - Daniel B. Knox
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| | | | - Wesley H. Self
- Department of Emergency Medicine, Vanderbilt University, Nashville, TN
| | - Samuel M. Brown
- Intermountain, Critical Care Echocardiography Service, Salt Lake City, UT
| |
Collapse
|
25
|
Braga PGS, Vieira JDS, Gurgel ARB, Brum PC. β-blockers and statins: exploring the potential off-label applications in breast, colorectal, prostate, and lung cancers. Front Pharmacol 2024; 15:1423502. [PMID: 39605917 PMCID: PMC11598443 DOI: 10.3389/fphar.2024.1423502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
Despite advances in cancer treatment, current cancer incidence and prevalence still demand multimodal treatments to enhance survival and clinical outcomes. Drugs used in cardiology, such as β-blockers and statins have gained attention for their potential roles in oncology. This review focused on their possible complementary use in solid tumors, including breast, colorectal, lung, and prostate cancers. The involvement of the autonomic nervous system in promoting tumor growth can be disrupted by β-blockers, potentially hindering cancer progression. Statins, known for their pleiotropic effects, may also inhibit cancer growth by reducing cholesterol availability, a key factor in cell proliferation. We will provide an update on the impact of these therapies on cancer treatment and surveillance, discuss the underlying mechanisms, and explore their effects on the heart, contributing to the growing field of cardio-oncology.
Collapse
Affiliation(s)
- Pedro Gabriel Senger Braga
- Laboratory of Cellular and Molecular Exercise Physiology, School of Physical Education and Sport of University of São Paulo, São Paulo, Brazil
- Clinica Pro-Coracao, São Paulo, Brazil
| | - Janaína da Silva Vieira
- Laboratory of Cellular and Molecular Exercise Physiology, School of Physical Education and Sport of University of São Paulo, São Paulo, Brazil
- Molecular Oncology Center, Sírio-Libanês Hospital, São Paulo, São Paulo, Brazil
| | - Aline Rachel Bezerra Gurgel
- Laboratory of Cellular and Molecular Exercise Physiology, School of Physical Education and Sport of University of São Paulo, São Paulo, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences of University of Sao Paulo, São Paulo, Brazil
| | - Patricia Chakur Brum
- Laboratory of Cellular and Molecular Exercise Physiology, School of Physical Education and Sport of University of São Paulo, São Paulo, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences of University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Dimitriadis K, Iliakis P, Pyrpyris N, Tatakis F, Fragkoulis C, Mantziaris V, Plaitis A, Beneki E, Tsioufis P, Hering D, Kollias A, Konstantinidis D, Tsioufis K. Renal Denervation in Heart Failure Treatment: Data for a Self-Fulfilling Prophecy. J Clin Med 2024; 13:6656. [PMID: 39597800 PMCID: PMC11594571 DOI: 10.3390/jcm13226656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Renal denervation (RDN), a transcatheter renal sympathetic nerve ablation procedure, is a relatively novel established procedure for the treatment of hypertension, with it being recognized as a third option for hypertension management in the most recent European guidelines, together with pharmacotherapy, for achieving blood pressure targets. Given the relationship between both hypertension and sympathetic overdrive and the development of heart failure (HF), even studies at the dawn of research on RDN explored it as a treatment to overcome diuretic resistance in those patients. As it is now recognized that RDN does not only have organ-specific but also systemic effects, several investigators have aimed to delineate whether renal sympathetic denervation could alter the prognosis, symptoms, and adverse events of HF patients. Data are available in both HF patients with reduced and preserved ejection fraction. As the significance of neuromodulation is gaining grounds in the HF therapeutic arsenal, in this review, we aim to provide a rationale for using RDN in HF and an up-to-date overview of available data in both HF phenotypes, as well as discuss the future of neuromodulatory therapy in HF management.
Collapse
Affiliation(s)
- Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Panagiotis Iliakis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Fotis Tatakis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Christos Fragkoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Vasileios Mantziaris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Aristides Plaitis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Panagiotis Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Dagmara Hering
- Department of Hypertension and Diabetology, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Anastasios Kollias
- Hypertension Center STRIDE-7, School of Medicine, Third Department of Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, 115 27 Athens, Greece;
| | - Dimitrios Konstantinidis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (P.I.); (N.P.); (F.T.); (C.F.); (V.M.); (A.P.); (E.B.); (P.T.); (D.K.); (K.T.)
| |
Collapse
|
27
|
Lopes Soares L, Portes AMO, Costa SFF, Leite LB, Natali AJ. Autonomic Dysregulation in Pulmonary Hypertension: Role of Physical Exercise. Hypertension 2024; 81:2228-2236. [PMID: 39234679 DOI: 10.1161/hypertensionaha.124.23573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Pulmonary hypertension (PH) is a rare and severe condition characterized by increased pressure in the pulmonary circulation, often resulting in right ventricular failure and death. The autonomic nervous system (ANS) plays a crucial role in the cardiovascular and pulmonary controls. Dysfunction of ANS has been implicated in the pathogenesis of cardiopulmonary diseases. Conversely, dysfunctions in ANS can arise from these diseases, impacting cardiac and pulmonary autonomic functions and contributing to disease progression. The complex interaction between ANS dysfunction and PH plays a crucial role in the disease progression, making it essential to explore interventions that modulate ANS, such as physical exercise, to improve the treatment and prognosis of patients with PH. This review addresses autonomic dysfunctions found in PH and their implications for the cardiopulmonary system. Furthermore, we discuss how physical exercise, a significant modulator of ANS, may contribute to the prognosis of PH. Drawing from evidence of aerobic and resistance exercise training in patients and experimental models of PH, potential cardiovascular benefits of exercise are presented. Finally, we highlight emerging therapeutic targets and perspectives to better cope with the complex condition. A comprehensive understanding of the interaction between ANS and PH, coupled with targeted physical exercise interventions, may pave the way for innovative therapeutic strategies and significantly improve the treatment and prognosis of vulnerable patients.
Collapse
Affiliation(s)
- Leôncio Lopes Soares
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| | | | | | - Luciano Bernardes Leite
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| | - Antônio José Natali
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| |
Collapse
|
28
|
Parichatikanond W, Duangrat R, Kurose H, Mangmool S. Regulation of β-Adrenergic Receptors in the Heart: A Review on Emerging Therapeutic Strategies for Heart Failure. Cells 2024; 13:1674. [PMID: 39451192 PMCID: PMC11506672 DOI: 10.3390/cells13201674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
The prolonged overstimulation of β-adrenergic receptors (β-ARs), a member of the G protein-coupled receptor (GPCR) family, causes abnormalities in the density and functionality of the receptor and contributes to cardiac dysfunctions, leading to the development and progression of heart diseases, especially heart failure (HF). Despite recent advancements in HF therapy, mortality and morbidity rates continue to be high. Treatment with β-AR antagonists (β-blockers) has improved clinical outcomes and reduced overall hospitalization and mortality rates. However, several barriers in the management of HF remain, providing opportunities to develop new strategies that focus on the functions and signal transduction of β-ARs involved in the pathogenesis of HF. As β-AR can signal through multiple pathways influenced by different receptor subtypes, expression levels, and signaling components such as G proteins, G protein-coupled receptor kinases (GRKs), β-arrestins, and downstream effectors, it presents a complex mechanism that could be targeted in HF management. In this narrative review, we focus on the regulation of β-ARs at the receptor, G protein, and effector loci, as well as their signal transductions in the physiology and pathophysiology of the heart. The discovery of potential ligands for β-AR that activate cardioprotective pathways while limiting off-target signaling is promising for the treatment of HF. However, applying findings from preclinical animal models to human patients faces several challenges, including species differences, the genetic variability of β-ARs, and the complexity and heterogeneity of humans. In this review, we also summarize recent updates and future research on the regulation of β-ARs in the molecular basis of HF and highlight potential therapeutic strategies for HF.
Collapse
Affiliation(s)
| | - Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Hitoshi Kurose
- Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan;
- Pharmacology for Life Sciences, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Supachoke Mangmool
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
29
|
Chaverra Kornerup S, Parotto M. Extubation-Related Complications. Int Anesthesiol Clin 2024; 62:82-90. [PMID: 39233574 DOI: 10.1097/aia.0000000000000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Extubation represents an essential component of airway management. While being a common procedure in anesthesiology and critical care medicine, it is accompanied by a significant risk of morbidity and mortality. Safe extubation requires considerable skills, risk stratification and advanced planning. It is important to emphasize that intentional extubation is always an elective procedure, and as such should only be executed when conditions are optimal. The purpose of this review is to discuss the complications associated with planned extubation in the adult patient, including risk factors and management strategies, mainly focusing on the postoperative setting.
Collapse
Affiliation(s)
- Santiago Chaverra Kornerup
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
- Department of Anesthesia and Pain Management, Toronto General Hospital, Toronto, ON, Canada
| | - Matteo Parotto
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
- Department of Anesthesia and Pain Management, Toronto General Hospital, Toronto, ON, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Wang M, Hou C, Jia F, Zhong C, Xue C, Li J. Aging-associated atrial fibrillation: A comprehensive review focusing on the potential mechanisms. Aging Cell 2024; 23:e14309. [PMID: 39135295 PMCID: PMC11464128 DOI: 10.1111/acel.14309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 10/11/2024] Open
Abstract
Atrial fibrillation (AF) has been receiving a lot of attention from scientists and clinicians because it is an extremely common clinical condition. Due to its special hemodynamic changes, AF has a high rate of disability and mortality. So far, although AF has some therapeutic means, it is still an incurable disease because of its complex risk factors and pathophysiologic mechanisms, which is a difficult problem for global public health. Age is an important independent risk factor for AF, and the incidence of AF increases with age. To date, there is no comprehensive review on aging-associated AF. In this review, we systematically discuss the pathophysiologic evidence for aging-associated AF, and in particular explore the pathophysiologic mechanisms of mitochondrial dysfunction, telomere attrition, cellular senescence, disabled macroautophagy, and gut dysbiosis involved in recent studies with aging-associated AF. We hope that by exploring the various dimensions of aging-associated AF, we can better understand the specific relationship between age and AF, which may be crucial for innovative treatments of aging-associated AF.
Collapse
Affiliation(s)
- Meng‐Fei Wang
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Can Hou
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Fang Jia
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Cheng‐Hao Zhong
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Cong Xue
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Jian‐Jun Li
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
31
|
Namazi M, Eftekhar SP, Mosaed R, Shiralizadeh Dini S, Hazrati E. Pulmonary Hypertension and Right Ventricle: A Pathophysiological Insight. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241274744. [PMID: 39257563 PMCID: PMC11384539 DOI: 10.1177/11795468241274744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024]
Abstract
Background Pulmonary hypertension (PH) is a pulmonary vascular disease characterized by elevated pulmonary vascular pressure. Long-term PH, irrespective of its etiology, leads to increased right ventricular (RV) pressure, RV hypertrophy, and ultimately, RV failure. Main body Research indicates that RV failure secondary to hypertrophy remains the primary cause of mortality in pulmonary arterial hypertension (PAH). However, the impact of PH on RV structure and function under increased overload remains incompletely understood. Several mechanisms have been proposed, including extracellular remodeling, RV hypertrophy, metabolic disturbances, inflammation, apoptosis, autophagy, endothelial-to-mesenchymal transition, neurohormonal dysregulation, capillary rarefaction, and ischemia. Conclusions Studies have demonstrated the significant role of oxidative stress in the development of RV failure. Understanding the interplay among these mechanisms is crucial for the prevention and management of RV failure in patients with PH.
Collapse
Affiliation(s)
- Mehrshad Namazi
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Hazrati
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Fernandes IA, Stavres J, Hamaoka T, Ojikutu QA, Sabino-Carvalho JL, Vianna LC, Luck JC, Blaha C, Cauffman AE, Dalton PC, Herr MD, Ruiz-Velasco V, Carr ZJ, Janicki PK, Cui J. Does a single oral administration of amiloride affect spontaneous arterial baroreflex sensitivity and blood pressure variability in healthy young adults? J Neurophysiol 2024; 132:922-928. [PMID: 39110514 DOI: 10.1152/jn.00264.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Preclinical models indicate that amiloride (AMD) reduces baroreflex sensitivity and perturbs homeostatic blood pressure (BP) regulation. However, it remains unclear whether these findings translate to humans. This study investigated whether oral administration of AMD reduces spontaneous cardiac and sympathetic baroreflex sensitivity and perturbs BP regulation in healthy young humans. Heart rate (HR; electrocardiography), beat-to-beat BP (photoplethysmography), and muscle sympathetic activity (MSNA, microneurography) were continuously measured in 10 young subjects (4 females) during rest across two randomized experimental visits: 1) after 3 h of oral administration of placebo (PLA, 10 mg of methylcellulose within a gelatin capsule) and 2) after 3 h of oral administration of AMD (10 mg). Visits were separated for at least 48 h. We calculated the standard deviation and other indices of BP variability. Spontaneous cardiac baroreflex was assessed via the sequence technique and cardiac autonomic modulation through time- and frequency-domain HR variability. The sensitivity (gain) of the sympathetic baroreflex was determined via weighted linear regression analysis between MSNA and diastolic BP. AMD did not affect HR, BP, and MSNA compared with PLA. Indexes of cardiac autonomic modulation (time- and frequency-domain HR variability) and BP variability were also unchanged after AMD ingestion. Likewise, AMD did not modify the gain of both spontaneous cardiac and sympathetic arterial baroreflex. A single oral dose of AMD does not affect spontaneous arterial baroreflex sensitivity and BP variability in healthy young adults.NEW & NOTEWORTHY Preclinical models indicate that amiloride (AMD), a nonselective antagonist of the acid-sensing ion channels (ASICs), impairs baroreflex sensitivity and perturbs blood pressure regulation. We translated these findings into humans, investigating the impact of acute oral ingestion of AMD on blood pressure variability and spontaneous cardiac and sympathetic baroreflex sensitivity in healthy young humans. In contrast to preclinical evidence, AMD does not impair spontaneous arterial baroreflex sensitivity and blood pressure variability in healthy young adults.
Collapse
Affiliation(s)
- Igor A Fernandes
- Human Neurovascular Control Laboratory, Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, United States
| | - Jon Stavres
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
- School of Kinesiology and Nutrition, University of Southern Mississippi, Hattiesburg, Mississippi, United States
| | - Takuto Hamaoka
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Qudus A Ojikutu
- Human Neurovascular Control Laboratory, Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, United States
| | - Jeann L Sabino-Carvalho
- Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Lauro C Vianna
- NeuroV̇ASQ̇-Integrative Physiology Laboratory, Faculty of Physical Education, University of Brasília, Federal District, Brazil
| | - J Carter Luck
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Cheryl Blaha
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Aimee E Cauffman
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Paul C Dalton
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Michael D Herr
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Zyad J Carr
- Department of Anesthesiology, Yale School of Medicine, Yale New Haven Hospital, New Haven, Connecticut
| | - Piotr K Janicki
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Jian Cui
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
| |
Collapse
|
33
|
Abou Kamar S, Andrzejczyk K, Petersen TB, Chin JF, Aga YS, de Bakker M, Akkerhuis KM, Geleijnse M, Brugts JJ, Sorop O, de Boer RA, Rizopoulos D, Asselbergs FW, Boersma E, den Ruijter H, van Dalen BM, Kardys I. The plasma proteome is linked with left ventricular and left atrial function parameters in patients with chronic heart failure. Eur Heart J Cardiovasc Imaging 2024; 25:1206-1215. [PMID: 38597740 PMCID: PMC11346355 DOI: 10.1093/ehjci/jeae098] [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: 11/27/2023] [Revised: 02/26/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
AIMS Examining the systemic biological processes in the heterogeneous syndrome of heart failure with reduced ejection fraction (HFrEF), as reflected by circulating proteins, in relation to echocardiographic characteristics, may provide insights into heart failure pathophysiology. We investigated the link of 4210 repeatedly measured circulating proteins with repeatedly measured echocardiographic parameters as well as with elevated left atrial pressure (LAP), in patients with HFrEF, to provide insights into underlying mechanisms. METHODS AND RESULTS In 173 patients with HFrEF, we performed 6-monthly echocardiography and trimonthly blood sampling during a median follow-up of 2.7 (inter-quartile range: 2.5-2.8) years. We investigated circulating proteins in relation to echocardiographic parameters of left ventricular [left ventricular ejection fraction (LVEF), global longitudinal strain (GLS)] and left atrial function [left atrial reservoir strain (LASr)] and elevated LAP (E/e' ratio >15) and used gene enrichment analyses to identify underlying pathophysiological processes. We found 723, 249, 792, and 427 repeatedly measured proteins, with significant associations with LVEF, GLS, LASr, and E/e' ratio, respectively. Proteins associated with LASr reflected pathophysiological mechanisms mostly related to the extracellular matrix. Proteins associated with GLS reflected cardiovascular biological processes and diseases, whereas those associated with LVEF reflected processes involved in the sympathetic nervous system. Moreover, 49 proteins were associated with elevated LAP; after correction for LVEF, three proteins remained: cystatin-D, fibulin-5, and HSP40. CONCLUSION Circulating proteins show varying associations with different echocardiographic parameters in patients with HFrEF. These findings suggest that pathways involved in atrial and ventricular dysfunction, as reflected by the plasma proteome, are distinct.
Collapse
Affiliation(s)
- S Abou Kamar
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
- The Netherlands Heart Institute, Moreelsepark 1, 3511 EP Utrecht, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM Rotterdam, The Netherlands
| | - K Andrzejczyk
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - T B Petersen
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
- Department of Biostatistics, University Medical Center Rotterdam, Erasmus University, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - J F Chin
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM Rotterdam, The Netherlands
| | - Y S Aga
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM Rotterdam, The Netherlands
| | - M de Bakker
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - K M Akkerhuis
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - M Geleijnse
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - J J Brugts
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - O Sorop
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - R A de Boer
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - D Rizopoulos
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
- Department of Biostatistics, University Medical Center Rotterdam, Erasmus University, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - F W Asselbergs
- Department of Cardiology, Amsterdam Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - E Boersma
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| | - H den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - B M van Dalen
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM Rotterdam, The Netherlands
| | - I Kardys
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, PO Box 2040 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
34
|
Chen Q, Huang Z, Chen J, Tian X, Zhang R, Liang Q, Liu Z, Cheng Y. Notoginsenoside R1 attenuates ischemic heart failure by modulating MDM2/β arrestin2-mediated β2-adrenergic receptor ubiquitination. Biomed Pharmacother 2024; 177:117004. [PMID: 38955084 DOI: 10.1016/j.biopha.2024.117004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
β2 adrenergic receptor (β2AR) is a G-protein-coupled receptor involved in cardiac protection. In chronic heart failure (CHF), persistent sympathetic nervous system activation occurs, resulting in prolonged β2AR activation and subsequent receptor desensitization and downregulation. Notoginsenoside R1 (NGR1) has the functions of enhancing myocardial energy metabolism and mitigating myocardial fibrosis. The mechanisms of NGR1 against ischemic heart failure are unclear. A left anterior descending (LAD) artery ligation procedure was performed on C57BL/6 J mice for four weeks. From the 4th week onwards, they were treated with various doses (3, 10, 30 mg/kg/day) of NGR1. Subsequently, the impacts of NGR1 on ischemic heart failure were evaluated by assessing cardiac function, morphological changes in cardiac tissue, and the expression of atrial natriuretic peptide (ANP) and beta-myosin heavy chain (β-MHC). H9c2 cells were protected by NGR1 when exposed to OGD/R conditions. H9c2 cells were likewise protected from OGD/R damage by NGR1. Furthermore, NGR1 increased β2AR levels and decreased β2AR ubiquitination. Mechanistic studies revealed that NGR1 enhanced MDM2 protein stability and increased the expression of MDM2 and β-arrestin2 while inhibiting their interaction. Additionally, under conditions produced by OGD/R, the protective benefits of NGR1 on H9c2 cells were attenuated upon administration of the MDM2 inhibitor SP141. According to these findings, NGR1 impedes the interplay between β-arrestin2 and MDM2, thereby preventing the ubiquitination and degradation of β2AR to improve CHF.
Collapse
Affiliation(s)
- Qi Chen
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ziwei Huang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jing Chen
- Department of Cardiovascular Disease, The First Afliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoyu Tian
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qi Liang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital Group, Shenzhen 518000, China.
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Yuanyuan Cheng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
35
|
Llerena-Velastegui J, Santamaria-Lasso M, Mejia-Mora M, Santander-Aldean M, Granda-Munoz A, Hurtado-Alzate C, de Jesus ACFS, Baldelomar-Ortiz J. Efficacy of Beta-Blockers and Angiotensin-Converting Enzyme Inhibitors in Non-Ischemic Dilated Cardiomyopathy: A Systematic Review and Meta-Analysis. Cardiol Res 2024; 15:281-297. [PMID: 39205958 PMCID: PMC11349132 DOI: 10.14740/cr1653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 09/04/2024] Open
Abstract
Background Non-ischemic dilated cardiomyopathy (NIDCM) is a form of heart failure with a poor prognosis and unclear optimal management. The aim of the study was to systematically review the literature and assess the efficacy and safety of beta-blockers and angiotensin-converting enzyme (ACE) inhibitors in the management of chronic heart failure secondary to NIDCM and explore their putative mechanisms of action. Methods Studies from 1990 to 2023 were reviewed using PubMed and EMBASE, focusing on their effects on left ventricular ejection fraction (LVEF) in NIDCM patients, according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results Beta-blockers showed a significant beneficial effect on LVEF improvement in NIDCM, with an overall effect size of Cohen's d = 1.30, 95% confidence interval (CI) (0.76, 1.84), high heterogeneity (Tau2 = 0.90; Chi2 = 162.05, df = 13, P < 0.00001; I2 = 92%), and a significant overall effect (Z = 4.72, P < 0.00001). ACE inhibitors also showed a beneficial role, but with less heterogeneity (Tau2 = 0.02; Chi2 = 1.09, df = 1, P = 0.30; I2 = 8%) and a nonsignificant overall effect (Z = 1.36, P = 0.17), 95% CI (-0.24, 1.31). Conclusions The study highlights the efficacy of carvedilol in improving LVEF in NIDCM patients over ACE inhibitors, recommends beta-blockers as first-line therapy, and advocates further research on ACE inhibitors.
Collapse
Affiliation(s)
- Jordan Llerena-Velastegui
- Medical School, Pontifical Catholic University of Ecuador, Quito, Ecuador
- Research Center, Center for Health Research in Latin America (CISeAL), Quito, Ecuador
| | | | - Melany Mejia-Mora
- Medical School, Pontifical Catholic University of Ecuador, Quito, Ecuador
| | | | | | | | | | | |
Collapse
|
36
|
Underwood L, Jiang CS, Oh JY, Sato PY. Unheralded Adrenergic Receptor Signaling in Cellular Oxidative Stress and Death. CURRENT OPINION IN PHYSIOLOGY 2024; 40:100766. [PMID: 39070968 PMCID: PMC11271747 DOI: 10.1016/j.cophys.2024.100766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Catecholamines (CAs) bind and activate adrenergic receptors (ARs), thus exuding a key role in cardiac adaptations to global physiological queues. Prolonged exposure to high levels of CAs promotes deleterious effects on the cardiovascular system, leading to organ dysfunction and heart failure (HF). In addition to the prominent role of ARs in inotropic and chronotropic responses, recent studies have delved into elucidating mechanisms contributing to CA toxicity and cell death. Central to this process is understanding the involvement of α1AR and βAR in cardiac remodeling and mechanisms of cellular survival. Here, we highlight the complexity of AR signaling and the fundamental need for a better understanding of its contribution to oxidative stress and cell death. This crucial informational nexus remains a barrier to the development of new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Lilly Underwood
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Chun-Sun Jiang
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Joo-Yeun Oh
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Priscila Y Sato
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
37
|
Simko F, Baka T. Pharmacological stimulation of the parasympathetic system - a promising means of cardioprotection in heart failure. Hypertens Res 2024; 47:2217-2220. [PMID: 38773336 PMCID: PMC11298403 DOI: 10.1038/s41440-024-01726-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/21/2024] [Indexed: 05/23/2024]
Affiliation(s)
- Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108, Bratislava, Slovak Republic.
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University, 83305, Bratislava, Slovak Republic.
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 84505, Bratislava, Slovak Republic.
| | - Tomas Baka
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108, Bratislava, Slovak Republic
| |
Collapse
|
38
|
Zamorano M, Monteagudo JM, González E, Rayo I, Fernández S, Castillo M, Zhou Q, de la Villa P, Zamorano JL. Pupil reflex as a marker of activity and prognosis in heart failure: a longitudinal and prospective study. ESC Heart Fail 2024; 11:1947-1954. [PMID: 38494834 PMCID: PMC11287362 DOI: 10.1002/ehf2.14746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/26/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
AIMS Compensatory mechanisms in heart failure (HF) are triggered to maintain adequate cardiac output. Among them, hyperactivation of the sympathetic nervous system (SNS) is one of the main ones and carries a worse prognosis. The pupillary reflex depends on the SNS, and we can evaluate it through pupillometry. The aim of the study was to compare the differences in pupillary reflex between patients with acute HF and controls and to analyse whether these differences in pupillary reflex may offer a new and easy prognostic factor in such patients. METHODS AND RESULTS We prospectively and consecutively included 107 patients admitted with decompensated HF. Quantitative pupillometry was performed with the NeuroOptics pupillometry during the first 24 h after admission and prior to discharge. The results were compared with those of a group of 100 healthy volunteers who also underwent pupillometry. The maximum baseline pupil size (MBPS) and the minimum pupil diameter (MPD) were measured. Patients with decompensated HF have a higher MBPS (3.64 ± 0.81) and higher MPD (2.60 ± 0.58) than HF patients at discharge and in the control group (P-value = 0.01 and 0.01, respectively). Also, HF patients presented an improvement in pupillometric values at discharge [MBPS (3.47 ± 0.79) and MPD (2.51 ± 0.58)] and showed no differences compared with the control group [MBPS (3.34 ± 0.82) and MPD (2.40 ± 0.53)] (P-value = 0.19 and 0.14, respectively). In addition, MBPS provides a good independent predictor of in-hospital and 1 month mortality in patients admitted with HF. Six patients (5.61%) died during hospital admission, and 11 patients (10.2%) died in the first month after discharge. Also, four patients (3.74%) were readmitted within 1 month of discharge. The receiver operating characteristic (ROC) curve for predicting in-hospital mortality through MBPS was 0.823. No patient with an MBPS < 3.7 mm died. The ROC curve for predicting combined mortality or readmission within the first month for MBPS was 0.698. CONCLUSIONS Pupillometry may be a new, non-invasive, and simple tool to determine the status of SNS, help in the prognostic stratification of acute HF patients, and improve therapeutic management.
Collapse
Affiliation(s)
- Margarita Zamorano
- Department of OphthalmologyUniversity Hospital of GuadalajaraCalle Donante de SangreGuadalajaraSpain
| | | | - Eduardo González
- Department of CardiologyRamón y Cajal University HospitalMadridSpain
| | - Isabel Rayo
- Department of CardiologyRamón y Cajal University HospitalMadridSpain
| | - Sara Fernández
- Department of CardiologyRamón y Cajal University HospitalMadridSpain
| | - Miguel Castillo
- Department of CardiologyRamón y Cajal University HospitalMadridSpain
| | - Qiheng Zhou
- Department of CardiologyRamón y Cajal University HospitalMadridSpain
| | - Pedro de la Villa
- Physiology Unit, Department of Systems Biology, School of MedicineUniversity of AlcaláMadridSpain
- Visual Neurophysiology Group‐IRYCISMadridSpain
| | | |
Collapse
|
39
|
Eraky AM, Yerramalla Y, Khan A, Mokhtar Y, Alamrosy M, Farag A, Wright A, Grounds M, Gregorich NM. Beta-Blockers as an Immunologic and Autonomic Manipulator in Critically Ill Patients: A Review of the Recent Literature. Int J Mol Sci 2024; 25:8058. [PMID: 39125627 PMCID: PMC11311757 DOI: 10.3390/ijms25158058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
The autonomic nervous system plays a key role in maintaining body hemostasis through both the sympathetic and parasympathetic nervous systems. Sympathetic overstimulation as a reflex to multiple pathologies, such as septic shock, brain injury, cardiogenic shock, and cardiac arrest, could be harmful and lead to autonomic and immunologic dysfunction. The continuous stimulation of the beta receptors on immune cells has an inhibitory effect on these cells and may lead to immunologic dysfunction through enhancing the production of anti-inflammatory cytokines, such as interleukin-10 (IL-10), and inhibiting the production of pro-inflammatory factors, such as interleukin-1B IL-1B and tissue necrotizing factor-alpha (TNF-alpha). Sympathetic overstimulation-induced autonomic dysfunction may also happen due to adrenergic receptor insensitivity or downregulation. Administering anti-adrenergic medication, such as beta-blockers, is a promising treatment to compensate against the undesired effects of adrenergic surge. Despite many misconceptions about beta-blockers, beta-blockers have shown a promising effect in decreasing mortality in patients with critical illness. In this review, we summarize the recently published articles that have discussed using beta-blockers as a promising treatment to decrease mortality in critically ill patients, such as patients with septic shock, traumatic brain injury, cardiogenic shock, acute decompensated heart failure, and electrical storm. We also discuss the potential pathophysiology of beta-blockers in various types of critical illness. More clinical trials are encouraged to evaluate the safety and effectiveness of beta-blockers in improving mortality among critically ill patients.
Collapse
Affiliation(s)
- Akram M. Eraky
- Emergency Medicine, Freeman Health System, Joplin, MO 64804, USA; (A.W.); (M.G.)
- Medical Education Department, Kansas City University, Kansas City, MO 64106, USA
| | - Yashwanth Yerramalla
- Critical Care Medicine, Freeman Health System, Joplin, MO 64804, USA; (Y.Y.); (A.K.); (Y.M.)
| | - Adnan Khan
- Critical Care Medicine, Freeman Health System, Joplin, MO 64804, USA; (Y.Y.); (A.K.); (Y.M.)
| | - Yasser Mokhtar
- Critical Care Medicine, Freeman Health System, Joplin, MO 64804, USA; (Y.Y.); (A.K.); (Y.M.)
| | - Mostafa Alamrosy
- Cardiology and Angiology Unit, Department of Clinical and Experimental Internal Medicine, Medical Research Institute, Alexandria University, Alexandria 5422031, Egypt;
| | - Amr Farag
- Critical Care Medicine, Portsmouth University Hospital, Portsmouth PO6 3LY, UK;
| | - Alisha Wright
- Emergency Medicine, Freeman Health System, Joplin, MO 64804, USA; (A.W.); (M.G.)
| | - Matthew Grounds
- Emergency Medicine, Freeman Health System, Joplin, MO 64804, USA; (A.W.); (M.G.)
| | - Nicole M. Gregorich
- School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
| |
Collapse
|
40
|
López Quiñones AJ, Vieira LS, Wang J. Cardiac Uptake of the Adrenergic Imaging Agent meta-Iodobenzylguanidine (mIBG) Is Mediated by Organic Cation Transporter 3 (Oct3). Drug Metab Dispos 2024; 52:899-905. [PMID: 38811159 PMCID: PMC11257688 DOI: 10.1124/dmd.124.001709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024] Open
Abstract
Heart failure (HF) is a chronic disease affecting 1%-2% of the global population.123I-labeled meta-iodobenzylguanidine (mIBG) is US Food and Drug Administration-approved for cardiac imaging and prognosis risk assessment in patients with HF. As a norepinephrine analog, mIBG is believed to be transported into adrenergic nerve terminals by the neuronal norepinephrine transporter (NET) and hence image sympathetic innervation of the myocardium. We previously showed that mIBG is an excellent substrate of organic cation transporter 3 (OCT3), an extraneuronal transporter expressed in cardiomyocytes. Here, we evaluated the in vivo impact of Oct3 on mIBG disposition and tissue distribution using Oct3 knockout mice. Oct3 +/+ and Oct3 -/- mice were administered with mIBG intravenously, and mIBG plasma pharmacokinetics and tissue exposures were determined. In Oct3 +/+ mice, mIBG exhibited extensive accumulation in multiple tissues (heart, salivary gland, liver, and adrenal gland). No difference was observed in overall plasma exposure between Oct3 +/+ and Oct3 -/- mice. Strikingly, cardiac mIBG was depleted in Oct3 -/- mice, resulting in 83% reduction in overall cardiac exposure (AUC0-24 h: 12.7 vs. 2.1 μg × h/g). mIBG tissue exposure (AUC0-24 h) was also reduced by 66%, 36%, and 31% in skeletal muscle, salivary gland, and lung, respectively, in Oct3 -/- mice. Our data demonstrated that Oct3 is the primary transporter responsible for cardiac mIBG uptake in vivo and suggested that cardiac mIBG imaging mainly measures OCT3 activity in cardiomyocytes but not NET-mediated uptake in adrenergic nerve endings. Our findings challenge the current paradigm in interpreting cardiac mIBG imaging results and suggest OCT3 as a potential genetic risk marker for HF prognosis. SIGNIFICANCE STATEMENT: 123I-labeled meta-iodobenzylguanidine is used for cardiac imaging and risk assessment in heart failure patients. Contrary to the current belief that meta-iodobenzylguanidine (mIBG) tracks cardiac sympathetic innervation due to its uptake by the neuronal norepinephrine transporter, the authors demonstrated that cardiac mIBG uptake is mediated by the extraneuronal transporter Oct3. Their findings warrant a re-evaluation of the scientific rationale behind cardiac mIBG scan and further suggest organic cation transporter 3 as a risk factor for disease progression in heart failure patients.
Collapse
Affiliation(s)
| | | | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
41
|
Maniaci A, Lavalle S, Parisi FM, Barbanti M, Cocuzza S, Iannella G, Magliulo G, Pace A, Lentini M, Masiello E, La Via L. Impact of Obstructive Sleep Apnea and Sympathetic Nervous System on Cardiac Health: A Comprehensive Review. J Cardiovasc Dev Dis 2024; 11:204. [PMID: 39057624 PMCID: PMC11277108 DOI: 10.3390/jcdd11070204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
A prevalent condition linked to an elevated risk of cardiovascular disease is sleep apnea. This review examines the connections between cardiac risk, the sympathetic nervous system, and sleep apnea. The increased risk of hypertension, arrhythmias, myocardial infarction, and heart failure was highlighted in the pathophysiology of sleep apnea and its effect on sympathetic activation. It is also important to consider potential processes such as oxidative stress, inflammation, endothelial dysfunction, and autonomic imbalance that may relate sleep apnea-induced sympathetic activation to cardiac risk. With implications for creating innovative diagnostic and treatment approaches to lessen the cardiovascular effects of sleep apnea, the goal of this investigation is to improve the understanding of the intricate link between sympathetic activity, cardiac risk, and sleep apnea. This study aimed to clarify the complex relationship between cardiovascular health and sleep apnea by synthesizing the available research and highlighting the crucial role played by the sympathetic nervous system in moderating this relationship. Our thorough investigation may have important therapeutic ramifications that will direct the creation of focused therapies to enhance cardiovascular outcomes in sleep apnea sufferers.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.)
| | - Salvatore Lavalle
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.)
| | - Federica Maria Parisi
- Department of Medical, Surgical Sciences and Advanced Technologies “GF Ingrassia” ENT Section, University of Catania, 95123 Catania, Italy; (F.M.P.); (S.C.)
| | - Marco Barbanti
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.)
| | - Salvatore Cocuzza
- Department of Medical, Surgical Sciences and Advanced Technologies “GF Ingrassia” ENT Section, University of Catania, 95123 Catania, Italy; (F.M.P.); (S.C.)
| | - Giannicola Iannella
- Otorhinolaryngology Department, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico, 00161 Rome, Italy; (G.I.); (G.M.); (A.P.)
| | - Giuseppe Magliulo
- Otorhinolaryngology Department, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico, 00161 Rome, Italy; (G.I.); (G.M.); (A.P.)
| | - Annalisa Pace
- Otorhinolaryngology Department, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico, 00161 Rome, Italy; (G.I.); (G.M.); (A.P.)
| | - Mario Lentini
- ASP Ragusa-Hospital Giovanni Paolo II, 97100 Ragusa, Italy;
| | - Edoardo Masiello
- Radiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Luigi La Via
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico–San Marco”, 95123 Catania, Italy
| |
Collapse
|
42
|
Mather M. The emotion paradox in the aging body and brain. Ann N Y Acad Sci 2024; 1536:13-41. [PMID: 38676452 DOI: 10.1111/nyas.15138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
With age, parasympathetic activity decreases, while sympathetic activity increases. Thus, the typical older adult has low heart rate variability (HRV) and high noradrenaline levels. Younger adults with this physiological profile tend to be unhappy and stressed. Yet, with age, emotional experience tends to improve. Why does older adults' emotional well-being not suffer as their HRV decreases? To address this apparent paradox, I present the autonomic compensation model. In this model, failing organs, the initial phases of Alzheimer's pathology, and other age-related diseases trigger noradrenergic hyperactivity. To compensate, older brains increase autonomic regulatory activity in the pregenual prefrontal cortex (PFC). Age-related declines in nerve conduction reduce the ability of the pregenual PFC to reduce hyperactive noradrenergic activity and increase peripheral HRV. But these pregenual PFC autonomic compensation efforts have a significant impact in the brain, where they bias processing in favor of stimuli that tend to increase parasympathetic activity (e.g., stimuli that increase feelings of safety) and against stimuli that tend to increase sympathetic activity (e.g., threatening stimuli). In summary, the autonomic compensation model posits that age-related chronic sympathetic/noradrenergic hyperactivity stimulates regulatory attempts that have the side effect of enhancing emotional well-being.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, Department of Psychology, and Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
43
|
Bode C, Preissl S, Hein L, Lother A. Catecholamine treatment induces reversible heart injury and cardiomyocyte gene expression. Intensive Care Med Exp 2024; 12:48. [PMID: 38733526 PMCID: PMC11088585 DOI: 10.1186/s40635-024-00632-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Catecholamines are commonly used as therapeutic drugs in intensive care medicine to maintain sufficient organ perfusion during shock. However, excessive or sustained adrenergic activation drives detrimental cardiac remodeling and may lead to heart failure. Whether catecholamine treatment in absence of heart failure causes persistent cardiac injury, is uncertain. In this experimental study, we assessed the course of cardiac remodeling and recovery during and after prolonged catecholamine treatment and investigated the molecular mechanisms involved. RESULTS C57BL/6N wild-type mice were assigned to 14 days catecholamine treatment with isoprenaline and phenylephrine (IsoPE), treatment with IsoPE and subsequent recovery, or healthy control groups. IsoPE improved left ventricular contractility but caused substantial cardiac fibrosis and hypertrophy. However, after discontinuation of catecholamine treatment, these alterations were largely reversible. To uncover the molecular mechanisms involved, we performed RNA sequencing from isolated cardiomyocyte nuclei. IsoPE treatment resulted in a transient upregulation of genes related to extracellular matrix formation and transforming growth factor signaling. While components of adrenergic receptor signaling were downregulated during catecholamine treatment, we observed an upregulation of endothelin-1 and its receptors in cardiomyocytes, indicating crosstalk between both signaling pathways. To follow this finding, we treated mice with endothelin-1. Compared to IsoPE, treatment with endothelin-1 induced minor but longer lasting changes in cardiomyocyte gene expression. DNA methylation-guided analysis of enhancer regions identified immediate early transcription factors such as AP-1 family members Jun and Fos as key drivers of pathological gene expression following catecholamine treatment. CONCLUSIONS The results from this study show that prolonged catecholamine exposure induces adverse cardiac remodeling and gene expression before the onset of left ventricular dysfunction which has implications for clinical practice. The observed changes depend on the type of stimulus and are largely reversible after discontinuation of catecholamine treatment. Crosstalk with endothelin signaling and the downstream transcription factors identified in this study provide new opportunities for more targeted therapeutic approaches that may help to separate desired from undesired effects of catecholamine treatment.
Collapse
Affiliation(s)
- Christine Bode
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Interdisciplinary Medical Intensive Care, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
44
|
Bredy C, Werner O, Helena H, Picot MC, Amedro P, Adda J. Cardiac magnetic resonance ventricular parameters correlate with cardiopulmonary fitness in patients with functional single ventricle. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2024; 40:1041-1048. [PMID: 38546925 DOI: 10.1007/s10554-024-03072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/19/2024] [Indexed: 06/05/2024]
Abstract
Owing to advances in medical and surgical fields, patients with single ventricle (SV) have a greatly improved life expectancy. However, progressive functional deterioration is observed over time, with a decrease in cardiopulmonary fitness. This study aimed to identify, in patients with SV, the association between cardiac magnetic resonance imaging (CMR) parameters and change in cardiopulmonary fitness assessed by cardiopulmonary exercise test (CPET), and if certain thresholds could anticipate a decline in aerobic fitness. Patients with an SV physiology were retrospectively screened from 2011 and 2021 in a single-centre observational study. We evaluated (1) the correlation between baseline CMR and CPET parameters, (2) the association between baseline CMR results and change in peak oxygen uptake (peak VO2), and (3) the cut-off values of end-diastolic and end-systolic volume index in patients with an impaired cardiopulmonary fitness (low peak VO2 and/or high VE/VCO2 slope). 32 patients were included in the study. End-systolic volume index (r = 0.37, p = 0.03), end-diastolic volume index (r = 0.45, p = 0.01), and cardiac index (r = 0.46, p = 0.01) correlated with the VE/VCO2 slope. End-systolic ventricular volume (r = - 0.39, p = 0.01), end-diastolic ventricular volume (r = - 0.38, p = 0.01), and cardiac output (r = - 0.45, p < 0.01) inversely correlated with the peak VO2. In multivariate analysis, the cardiac index obtained from baseline CMR was inversely associated with the change in peak VO2 (p < 0.01). An end-diastolic volume index > 101 ml/m2 and an end-systolic volume index > 47 ml/m2 discriminated patients with impaired cardiopulmonary fitness. CMR parameters correlate with cardiopulmonary fitness in patients with SV and can therefore be useful for follow-up and therapeutic management of these patients.
Collapse
Affiliation(s)
- Charlene Bredy
- Paediatric and Congenital Cardiology Department, M3C Regional Reference CHD Centre, Montpellier University Hospital, Montpellier, France
| | - Oscar Werner
- Paediatric and Congenital Cardiology Department, M3C Regional Reference CHD Centre, Montpellier University Hospital, Montpellier, France
- Pediatric Imaging Department, Montpellier University Hospital, Montpellier, France
| | - Huguet Helena
- Epidemiology and Clinical Research Department, University Hospital, Montpellier, France
- Clinical Investigation Centre, INSERM U1411, Montpellier University Hospital, University of Montpellier, Montpellier, France
| | - Marie-Christine Picot
- Epidemiology and Clinical Research Department, University Hospital, Montpellier, France
- Clinical Investigation Centre, INSERM U1411, Montpellier University Hospital, University of Montpellier, Montpellier, France
| | - Pascal Amedro
- Paediatric and Congenital Cardiology Department, M3C National Reference Centre, Bordeaux University Hospital, Bordeaux, France
- IHU Liryc, Electrophysiology and Heart Modelling Institute, Bordeaux University Foundation, Pessac, France
| | - Jerome Adda
- Cardiology Department, Montpellier University Hospital, Montpellier, France.
| |
Collapse
|
45
|
Luo J, Shi L, Liu J, Li G, Tu L, Hu S. SGLT2 inhibition, plasma proteins, and heart failure: a proteome-wide Mendelian Randomization and colocalization study. Front Cardiovasc Med 2024; 11:1371513. [PMID: 38725835 PMCID: PMC11079590 DOI: 10.3389/fcvm.2024.1371513] [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: 01/16/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Objective To investigate the causal contributions of Sodium-glucose cotransporter 2 (SGLT2) inhibition on Heart Failure (HF) and identify the circulating proteins that mediate SGLT2 inhibition's effects on HF. Methods Applying a two-sample, two-step Mendelian Randomization (MR) analysis, we aimed to estimate: (1) the causal impact of SGLT2 inhibition on HF; (2) the causal correlation of SGLT2 inhibition on 4,907 circulating proteins; (3) the causal association of SGLT2 inhibition-driven plasma proteins on HF. Genetic variants linked to SGLT2 inhibition derived from the previous studies. The 4,907 circulating proteins were derived from the deCODE study. Genetic links to HF were obtained through the Heart Failure Molecular Epidemiology for Therapeutic Targets (HERMES) consortium. Results SGLT2 inhibition demonstrated a lower risk of HF (odds ratio [OR] = 0.44, 95% CI [0.26, 0.76], P = 0.003). Among 4,907 circulating proteins, we identified leucine rich repeat transmembrane protein 2 (LRRTM2), which was related to both SGLT2 inhibition and HF. Mediation analysis revealed that the impact of SGLT2 inhibition on HF operates indirectly through LRRTM2 [β = -0.20, 95% CI (-0.39, -0.06), P = 0.02] with a mediation proportion of 24.6%. Colocalization analysis provided support for the connections between LRRTM2 and HF. Conclusion The study indicated a causative link between SGLT2 inhibition and HF, with plasma LRRTM2 potentially serving as a mediator.
Collapse
Affiliation(s)
- Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Lili Shi
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jingrui Liu
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gen Li
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Shuiqing Hu
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Dolejší E, Janoušková A, Jakubík J. Muscarinic Receptors in Cardioprotection and Vascular Tone Regulation. Physiol Res 2024; 73:S389-S400. [PMID: 38634650 PMCID: PMC11412339 DOI: 10.33549/physiolres.935270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Muscarinic acetylcholine receptors are metabotropic G-protein coupled receptors. Muscarinic receptors in the cardiovascular system play a central role in its regulation. Particularly M2 receptors slow down the heart rate by reducing the impulse conductivity through the atrioventricular node. In general, activation of muscarinic receptors has sedative effects on the cardiovascular system, including vasodilation, negative chronotropic and inotropic effects on the heart, and cardioprotective effects, including antifibrillatory effects. First, we review the signaling of individual subtypes of muscarinic receptors and their involvement in the physiology and pathology of the cardiovascular system. Then we review age and disease-related changes in signaling via muscarinic receptors in the cardiovascular system. Finally, we review molecular mechanisms involved in cardioprotection mediated by muscarinic receptors leading to negative chronotropic and inotropic and antifibrillatory effects on heart and vasodilation, like activation of acetylcholine-gated inward-rectifier K+-currents and endothelium-dependent and -independent vasodilation. We relate this knowledge with well-established cardioprotective treatments by vagal stimulation and muscarinic agonists. It is well known that estrogen exerts cardioprotective effects against atherosclerosis and ischemia-reperfusion injury. Recently, some sex hormones and neurosteroids have been shown to allosterically modulate muscarinic receptors. Thus, we outline possible treatment by steroid-based positive allosteric modulators of acetylcholine as a novel pharmacotherapeutic tactic. Keywords: Muscarinic receptors, Muscarinic agonists, Allosteric modulation, Cardiovascular system, Cardioprotection, Steroids.
Collapse
Affiliation(s)
- E Dolejší
- Laboratory of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | | | | |
Collapse
|
47
|
Olivella A, Almenar-Bonet L, Moliner P, Coloma E, Martínez-Rubio A, Paz Bermejo M, Boixeda R, Cediel G, Méndez Fernández AB, Facila Rubio L. Role of vericiguat in management of patients with heart failure with reduced ejection fraction after worsening episode. ESC Heart Fail 2024; 11:628-636. [PMID: 38158630 DOI: 10.1002/ehf2.14647] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 10/26/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024] Open
Abstract
Worsening heart failure (HF) is a vulnerable period in which the patient has a markedly high risk of death or HF hospitalization (up to 10% and 30%, respectively, within the first weeks after episode). The prognosis of HF patients can be improved through a comprehensive approach that considers the different neurohormonal systems, with the early introduction and optimization of the quadruple therapy with sacubitril-valsartan, beta-blockers, mineralocorticoid receptor antagonists, and inhibitors. Despite that, there is a residual risk that is not targeted with these therapies. Currently, it is recognized that the cyclic guanosine monophosphate deficiency has a negative direct impact on the pathogenesis of HF, and vericiguat, an oral stimulator of soluble guanylate cyclase, can restore this pathway. The effect of vericiguat has been explored in the VICTORIA study, the largest chronic HF clinical trial that has mainly focused on patients with recent worsening HF, evidencing a significant 10% risk reduction of the primary composite endpoint of cardiovascular death or HF hospitalization (number needed to treat 24), after adding vericiguat to standard therapy. This benefit was independent of background HF therapy. Therefore, optimization of treatment should be performed as earlier as possible, particularly within vulnerable periods, considering also the use of vericiguat.
Collapse
Affiliation(s)
- Aleix Olivella
- Heart Failure Unit, Department of Cardiology, Hospital Universitari Vall d'Hebrón, Vall d'Hebrón Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Almenar-Bonet
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
- Heart Failure and Transplantation Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Pedro Moliner
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
- Community Heart Failure Program (UMICO), Department of Cardiology, Bellvitge University Hospital, Barcelona, Spain
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Emmanuel Coloma
- Heart Failure and Transplantation Unit, Internal Medicine Department and Hospital at Home Unit, Hospital Clinic, Barcelona, Spain
- Instituto de Investigaciones Médicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Antoni Martínez-Rubio
- Department of Cardiology, Hospital Universitario de Sabadell, Sabadell, Spain
- Universidad Autonoma de Barcelona, Sabadell, Spain
| | | | - Ramon Boixeda
- Department of Internal Medicine, Hospital de Mataró, Mataró, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - German Cediel
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
- Heart Failure Unit, Department of Cardiology, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Ana Belén Méndez Fernández
- Heart Failure Unit, Department of Cardiology, Hospital Universitari Vall d'Hebrón, Vall d'Hebrón Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lorenzo Facila Rubio
- Department of Cardiology, Hospital General de Valencia, Universitat de Valencia, Avda. Tres Cruces 2, 46014, Valencia, Spain
| |
Collapse
|
48
|
Goldenberg I, Younis A, McNitt S, Klein H, Goldenberg I, Kutyifa V. Prior history of atrial fibrillation and arrhythmic outcomes: Data from the WEARIT-II prospective registry. J Cardiovasc Electrophysiol 2024; 35:785-793. [PMID: 38383981 DOI: 10.1111/jce.16215] [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: 05/13/2023] [Revised: 11/27/2023] [Accepted: 02/01/2024] [Indexed: 02/23/2024]
Abstract
INTRODUCTION Wearable cardioverter defibrillator (WCD) is utilized in patients with assumed but not yet confirmed risk for sudden cardiac death (SCD). Many of these patients also present with atrial fibrillation (AF). However, the rate of WCD-detected ventricular or atrial arrhythmia events in this specific high-risk cohort is not well understood. METHODS In WEARIT-II, the cumulative probability of any sustained or nonsustained VT/VF (WCD-treated and nontreated), and atrial/supraventricular arrhythmias during WCD use was assessed using the Kaplan-Meier method by prior AF, with comparisons by the log-rank test. The incidence of ventricular and atrial arrhythmia events were expressed as events per 100 patient-years, and were analyzed by prior AF using negative binomial regression. RESULTS WEARIT-II enrolled 2000 patients, 557 (28%) of whom had AF before enrollment. Cumulative probability of any sustained or nonsustained WCD-detected VT/VF during WCD use was significantly higher among patients with a history of AF than without AF (6% vs. 3%, p = .001). Similarly, the recurrent rate of any sustained or nonsustained VT/VF was significantly higher in patients with prior AF versus no prior AF (131.5 events per 100 patient-years vs. 22.7 events per 100 patient-years, p = .001). Patients with prior AF also had a significantly higher burden of any WCD-detected atrial arrhythmias/SVT/inappropriate arrhythmias therapy (183.2 events per 100 patient-years vs. 74.8 events per 100 patient-years, p < .001). CONCLUSION Our results demonstrate that patients with a history of AF wearing the WCD for risk assessment have a higher incidence of ventricular arrhythmias that may facilitate the decision making for ICD implantation.
Collapse
Affiliation(s)
- Ido Goldenberg
- Department of Medicine, Clinical Cardiovascular Research Center, Cardiology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Arwa Younis
- Department of Medicine, Clinical Cardiovascular Research Center, Cardiology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Scott McNitt
- Department of Medicine, Clinical Cardiovascular Research Center, Cardiology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Helmut Klein
- Department of Medicine, Clinical Cardiovascular Research Center, Cardiology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Ilan Goldenberg
- Department of Medicine, Clinical Cardiovascular Research Center, Cardiology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Valentina Kutyifa
- Department of Medicine, Clinical Cardiovascular Research Center, Cardiology Division, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
49
|
Keefe JA, Garber R, McCauley MD, Wehrens XHT. Tachycardia and Atrial Fibrillation-Related Cardiomyopathies: Potential Mechanisms and Current Therapies. JACC. HEART FAILURE 2024; 12:605-615. [PMID: 38206235 DOI: 10.1016/j.jchf.2023.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/06/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024]
Abstract
Atrial fibrillation (AF) is associated with an increased risk of new-onset ventricular contractile dysfunction, termed arrhythmia-induced cardiomyopathy (AIC). Although cardioembolic stroke remains the most feared and widely studied complication of AF, AIC is also a clinically important consequence of AF that portends significant morbidity and mortality to patients with AF. Current treatments are aimed at restoring sinus rhythm through catheter ablation and rate and rhythm control, but these treatments do not target the underlying molecular mechanisms driving the progression from AF to AIC. Here, we describe the clinical features of the various AIC subtypes, discuss the pathophysiologic mechanisms driving the progression from AF to AIC, and review the evidence surrounding current treatment options. In this review, we aim to identify key knowledge gaps that will enable the development of more effective AIC therapies that target cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Rebecca Garber
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mark D McCauley
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown VA Medical Center, Chicago, Illinois, USA.
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA; Departments of Pediatrics, Medicine, and Neuroscience, and Center for Space Medicine, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
50
|
Zhou S, Liu Y, Huang X, Wu C, Pórszász R. Omecamtiv Mecarbil in the treatment of heart failure: the past, the present, and the future. Front Cardiovasc Med 2024; 11:1337154. [PMID: 38566963 PMCID: PMC10985333 DOI: 10.3389/fcvm.2024.1337154] [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: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Heart failure, a prevailing global health issue, imposes a substantial burden on both healthcare systems and patients worldwide. With an escalating prevalence of heart failure, prolonged survival rates, and an aging demographic, an increasing number of individuals are progressing to more advanced phases of this incapacitating ailment. Against this backdrop, the quest for pharmacological agents capable of addressing the diverse subtypes of heart failure becomes a paramount pursuit. From this viewpoint, the present article focuses on Omecamtiv Mecarbil (OM), an emerging chemical compound said to exert inotropic effects without altering calcium homeostasis. For the first time, as a review, the present article uniquely started from the very basic pathophysiology of heart failure, its classification, and the strategies underpinning drug design, to on-going debates of OM's underlying mechanism of action and the latest large-scale clinical trials. Furthermore, we not only saw the advantages of OM, but also exhaustively summarized the concerns in sense of its effects. These of no doubt make the present article the most systemic and informative one among the existing literature. Overall, by offering new mechanistic insights and therapeutic possibilities, OM has carved a significant niche in the treatment of heart failure, making it a compelling subject of study.
Collapse
Affiliation(s)
- Shujing Zhou
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ying Liu
- Department of Cardiology, Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xufeng Huang
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Chuhan Wu
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|