101
|
Dewaswala N, Mishra V, Bhopalwala H, Minhas AK, Keshavamurthy S. Pathophysiology and Management of Heart Failure in the Elderly. Int J Angiol 2022; 31:251-259. [PMID: 36588873 PMCID: PMC9803556 DOI: 10.1055/s-0042-1758357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The population of elderly adults is increasing globally. It has been projected that the population of adults aged 65 years will increase by approximately 80% by 2050 in the United States. Similarly, the elderly population is rising in other countries; a notable example being Japan where approximately 30% of the population are aged above 65 years. The pathophysiology and management of heart failure (HF) in this age group tend to have more intricacies than in younger age groups owing to the presence of multiple comorbidities. The normal aging biology includes progressive disruption at cellular and genetic levels and changes in molecular signaling and mechanical activities that contribute to myocardial abnormalities. Older adults with HF secondary to ischemic or valvular heart disease may benefit from surgical therapy, valve replacement or repair for valvular heart disease and coronary artery bypass grafting for coronary artery disease. While referring these patients for surgery, patient and family expectations and life expectations should be taken into account. In this review, we will cover the pathophysiology and the management of HF in the elderly, specifically discussing important geriatric domains such as frailty, cognitive impairment, delirium, polypharmacy, and multimorbidity.
Collapse
Affiliation(s)
- Nakeya Dewaswala
- Department of Cardiovascular Diseases, University of Kentucky, Lexington, Kentucky
| | - Vinayak Mishra
- Grant Medical College and Sir JJ Group of Hospitals, Mumbai, Maharashtra, India
| | - Huzefa Bhopalwala
- Department of Internal Medicine, Appalachian Regional Healthcare, Whitesburg, Kentucky
| | - Abdul Khan Minhas
- Department of Internal Medicine, Forrest General Hospital, Hattiesburg, Mississippi
| | - Suresh Keshavamurthy
- Department of Surgery, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
102
|
Hu Z, Gao S, Yang J, Xu B, Tang W, Bradley JL, Peberdy MA, Ornato JP. IVABRADINE-INDUCED HEART RATE REDUCTION INCREASES THE SEVERITY OF POSTRESUSCITATION MYOCARDIAL DYSFUNCTION IN A RAT MODEL OF CARDIOPULMONARY RESUSCITATION. Shock 2022; 58:573-581. [PMID: 36548647 PMCID: PMC9803391 DOI: 10.1097/shk.0000000000002020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 10/19/2022] [Indexed: 12/24/2022]
Abstract
ABSTRACT Aims: A rapid heart rate (HR) that occurs after cardiopulmonary resuscitation (CPR) is a short-term compensatory mechanism preserving cardiac output. However, if of long duration, it is unfavorable for myocardial function postresuscitation because of disrupted balance between myocardial oxygen supply and demand. This raises the assumption that such a sustained fast HR should be regulated. The present study aimed to investigate the follow-on effect of ivabradine (a specific inhibitor of the I f current of the sinoatrial node)-induced HR reduction (HRR) on postresuscitation myocardial function in a rat model of CPR. Methods and results: Six minutes of ventricular fibrillation and 8 min of CPR were performed on Sprague-Dawley rats. All 32 resuscitated animals were then randomized into saline and ivabradine groups, each group having nonsurvival and survival subgroups (n = 8 each). Saline or ivabradine (0.5 mL/kg) was administered at 1 h postresuscitation. Heart rate, myocardial function as expressed by cardiac output, ejection fraction, and myocardial performance index were assessed at baseline and hourly from 1 to 5 h postresuscitation. Heart rate variability was analyzed at baseline and at 1, 3, and 5 h postresuscitation. Serum epinephrine and cardiac troponin I at baseline and at 1, 3, and 5 h postresuscitation in nonsurvival subgroup were measured. Survival duration in the survival subgroup was observed. The baseline HR was approximately 390 beats/min (bpm). After resuscitation, an average increase of Δ ≈ +15 bpm (relative ratio ≈ +3.8%) with a resultant HR of 405 bpm lasting more than 5 h occurred. Ivabradine group achieved a steady HRR of Δ ≈ -30 bpm (relative ratio ≈ -7.4%) as compared with saline group ( P < 0.01). Postresuscitation myocardial function was significantly worse in the ivabradine group (all P < 0.01). Heart rate variability was significantly impaired in the ivabradine group (all P < 0.05). Serum cardiac troponin I and epinephrine concentration were significantly higher in the ivabradine group (all P < ?0.01). Survival duration was significantly shortened in the ivabradine group as compared with the saline group (388 vs. 526 min, P < ?0.01). Conclusions: Ivabradine-induced HRR increases the severity of postresuscitation myocardial dysfunction and shortens survival duration in a rat model of CPR.
Collapse
Affiliation(s)
- Zhangle Hu
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
- Department of Cardiology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Jin Yang
- Department of Respiratory Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Banglong Xu
- Department of Cardiology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wanchun Tang
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Jennifer L. Bradley
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Mary Ann Peberdy
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Joseph P. Ornato
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
103
|
Li YL. Stellate Ganglia and Cardiac Sympathetic Overactivation in Heart Failure. Int J Mol Sci 2022; 23:ijms232113311. [PMID: 36362099 PMCID: PMC9653702 DOI: 10.3390/ijms232113311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Heart failure (HF) is a major public health problem worldwide, especially coronary heart disease (myocardial infarction)-induced HF with reduced ejection fraction (HFrEF), which accounts for over 50% of all HF cases. An estimated 6 million American adults have HF. As a major feature of HF, cardiac sympathetic overactivation triggers arrhythmias and sudden cardiac death, which accounts for nearly 50–60% of mortality in HF patients. Regulation of cardiac sympathetic activation is highly integrated by the regulatory circuitry at multiple levels, including afferent, central, and efferent components of the sympathetic nervous system. Much evidence, from other investigators and us, has confirmed the afferent and central neural mechanisms causing sympathoexcitation in HF. The stellate ganglion is a peripheral sympathetic ganglion formed by the fusion of the 7th cervical and 1st thoracic sympathetic ganglion. As the efferent component of the sympathetic nervous system, cardiac postganglionic sympathetic neurons located in stellate ganglia provide local neural coordination independent of higher brain centers. Structural and functional impairments of cardiac postganglionic sympathetic neurons can be involved in cardiac sympathetic overactivation in HF because normally, many effects of the cardiac sympathetic nervous system on cardiac function are mediated via neurotransmitters (e.g., norepinephrine) released from cardiac postganglionic sympathetic neurons innervating the heart. This review provides an overview of cardiac sympathetic remodeling in stellate ganglia and potential mechanisms and the role of cardiac sympathetic remodeling in cardiac sympathetic overactivation and arrhythmias in HF. Targeting cardiac sympathetic remodeling in stellate ganglia could be a therapeutic strategy against malignant cardiac arrhythmias in HF.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; ; Tel.: +1-402-559-3016; Fax: +1-402-559-9659
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
104
|
Matsuda Y, Masuda M, Sakio T, Asai M, Iida O, Okamoto S, Ishihara T, Nanto K, Tsujimura T, Hata Y, Uematsu H, Mano T. Heart Rate Decrease After Atrial Fibrillation Catheter Ablation Predicts Decompensated Heart Failure After the Procedure. Circ Rep 2022; 4:461-468. [PMID: 36304430 PMCID: PMC9535125 DOI: 10.1253/circrep.cr-22-0078] [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: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Decompensated heart failure (DHF) can complicate catheter ablation for atrial fibrillation (AF). We investigated the association between heart rate and DHF in AF patients undergoing catheter ablation. Methods and Results: In all, 1,004 consecutive patients who underwent initial ablation for AF (mean [±SD] age 68±10 years; 34% female; persistent AF n=513 [51%]) were enrolled in the study. Heart rate was assessed before and after ablation. DHF was defined as heart failure requiring medical therapy within 2 days after the procedure. The incidence of DHF was 2% (22 of 1,004 patients). Patients with DHF had a higher prevalence of a history of symptomatic heart failure (11/22 [50%] vs. 160/982 [16%]; P<0.0001) and a greater degree of heart rate decrease after the procedure (-21±29 vs. 2±21 beats/min; P=0.001) than those without DHF. On multivariate analysis, heart rate decrease was a significant independent predictor of DHF (hazard ratio 0.8; 95% confidence interval 0.7-0.9; P=0.004; 10 beats/min-increment). Conclusions: In patients undergoing AF ablation, a decrease in heart rate after the procedure was an independent predictor of DHF.
Collapse
Affiliation(s)
| | | | | | | | - Osamu Iida
- Kansai Rosai Hospital Cardiovascular Center Amagasaki Japan
| | - Shin Okamoto
- Kansai Rosai Hospital Cardiovascular Center Amagasaki Japan
| | | | - Kiyonori Nanto
- Kansai Rosai Hospital Cardiovascular Center Amagasaki Japan
| | | | - Yosuke Hata
- Kansai Rosai Hospital Cardiovascular Center Amagasaki Japan
| | | | - Toshiaki Mano
- Kansai Rosai Hospital Cardiovascular Center Amagasaki Japan
| |
Collapse
|
105
|
Xia Y, Wang S, Wu W, Liang J. Association between serum sodium level trajectories and survival in patients with heart failure. ESC Heart Fail 2022; 10:255-263. [PMID: 36193558 PMCID: PMC9871655 DOI: 10.1002/ehf2.14187] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 01/27/2023] Open
Abstract
AIMS The effect of changes in serum sodium levels on the survival of patients with heart failure (HF) is unclear. We aimed to analyse the impact of serum sodium level trajectories on survival in intensive care unit (ICU) patients with HF. METHODS A total of 4760 patients diagnosed with HF between 2001 and 2012 from the Medical Information Mart for Intensive Care III (MIMIC-III) database were extracted. Of these patients, 1132 patients who died within 48 h of ICU admission were excluded, and 3628 patients were included in this retrospective cohort study. Sodium levels were measured at baseline, 6, 12, 18, 24, 30, 36, 42, and 48 h. Patients were divided into hyponatremia, normal, and hypernatremia groups based on baseline sodium levels, and trajectory modelling was performed for each group separately. Group-based trajectory model (GBTM) method was utilized to identify serum sodium levels trajectories. RESULTS The number of patients with hyponatremia (<135 mmol/L), normal sodium levels (135-145 mmol/L), and hypernatremia (>145 mmol/L) at baseline were 594 (16.37%), 2,738 (75.47%), and 296 (8.16%), respectively. A total of seven trajectory groups were identified, including hyponatremia-slow rise group [initial levels (IL), 128.48 ± 5.42 mmol/L; end levels (EL), 131.23 ± 3.83 mmol/L], hyponatremia-rapid rise to normal group (IL, 132.13 ± 2.18 mmol/L; EL, 137.46 ± 3.68 mmol/L), normal-slow decline group (IL, 137.65 ± 2.15 mmol/L; EL, 134.50 ± 2.54 mmol/L), normal-steady-state group (IL, 139.20 ± 2.26 mmol/L; EL, 139.04 ± 2.58 mmol/L), normal-slow rise group (IL, 140.94 ± 2.37 mmol/L; EL, 143.43 ± 2.89 mmol/L), hypernatremia-rapid decline to normal group (IL, 146.31 ± 1.98 mmol/L; EL, 140.71 ± 3.61 mmol/L), and hypernatremia-slow decline group (IL, 148.89 ± 5.54 mmol/L; EL, 146.28 ± 3.90 mmol/L). The results showed that hyponatremia-slow rise group [hazard ratio (HR) = 1.35; 95% confidence interval (CI), 1.01-1.80, P = 0.040], hyponatremia-rapid rise to normal group (HR = 1.37; 95% CI, 1.11-1.71, P = 0.004), hypernatremia-rapid decline to normal group (HR = 1.46; 95% CI, 1.08-1.97, P = 0.014), and hypernatremia-slow decline group (HR = 1.49; 95% CI, 1.07-2.07, P = 0.018) trajectories were associated with an increased risk of 1-year mortality in HF patients compared with normal-steady-state group. After adjustment for all confounders, hyponatremia-rapid rise to normal group (HR = 1.26, 95% CI; 1.01-1.57, P = 0.038) and hypernatremia-rapid decline to normal group (HR = 1.36; 95% CI, 1.01-1.84, P = 0.047) trajectories were still related to an increased risk of 1-year mortality in patients with HF. CONCLUSIONS Serum sodium level trajectories were associated with mortality in patients with HF. Association between serum sodium level trajectories and prognosis in patients with HF deserve further study.
Collapse
Affiliation(s)
- Yan‐Mei Xia
- Department of Intensive Care Unit, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan 030032ShanxiChina,Department of Intensive Care Unit, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan 430030HubeiChina
| | - Shan Wang
- Shanxi University of Chinese MedicineJinzhong 030619ShanxiChina
| | - Wei‐Dong Wu
- Department of Intensive Care Unit, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan 030032ShanxiChina,Department of Intensive Care Unit, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan 430030HubeiChina
| | - Ji‐Fang Liang
- Department of Intensive Care Unit, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan 030032ShanxiChina,Department of Intensive Care Unit, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan 430030HubeiChina
| |
Collapse
|
106
|
Zhang J, Ou C, Chen M. Curcumin attenuates cadmium-induced atherosclerosis by regulating trimethylamine-N-oxide synthesis and macrophage polarization through remodeling the gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114057. [PMID: 36084504 DOI: 10.1016/j.ecoenv.2022.114057] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/23/2022] [Accepted: 09/03/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Studies have shown that cadmium (Cd) exposure primarily occurs through diet, and Cd ingestion is a risk factor for atherosclerosis (AS). However, the underlying mechanism remains unclear. As a target organ, the gastrointestinal tract may play a key role in Cd-induced AS. Additionally, as curcumin is insoluble in water but stable in the stomach of acidic pH, it may play regulative roles in the gut. OBJECTIVES We assess the effect of Cd exposure on gut flora, trimethylamine-N-oxide (TMAO) metabolism and macrophage polarization, further investigate whether curcumin protects against Cd-induced AS by remodeling gut microbiota. METHODS AND RESULTS The results of 16 S rRNA sequencing show that Cd exposure causes diversity reduction and compositional alteration of the microbial community, resulting in the increasing TMAO synthesis, the imbalance of lipid metabolism, and the M1-type macrophage polarization in the mouse model (ApoE-/-) of AS. As a result, the plaque area is increased with Cd exposure, shown by oil red O staining. TMAO synthesis is positively correlated with the concentration of blood Cd, and the dynamics of specific bacteria in this process were revealed at the phylum to genus levels. Moreover, the effects of intestinal flora and TMAO on Cd-induced AS are further confirmed via microbial transplantation from a mouse model not exposed to Cd, as the transplantation decreases plaque area. Finally, the gavage with curcumin reverses the Cd-induced pathological progression via gut flora restoration. CONCLUSIONS We first demonstrate that Cd exposure worsens the progression of AS via intestinal flora imbalance and increased TMAO synthesis. Curcumin was verified as a potential novel intervention for preventing Cd-induced AS via remodeling gut microbiota. This study elucidates a new approach for treating AS in regions with significant Cd exposure.
Collapse
Affiliation(s)
- Jiexin Zhang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510280, China
| | - Caiwen Ou
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510280, China.
| | - Minsheng Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510280, China.
| |
Collapse
|
107
|
Piayda K, Sievert K, Sievert H, Shaburishvili T, Gogorishvili I, Rothman M, Januzzi JL, Lindenfeld J, Stone GW. Endovascular Baroreflex Amplification With the MobiusHD Device in Patients With Heart Failure and Reduced Ejection Fraction: Interim Analysis of the First-in-Human Results. STRUCTURAL HEART : THE JOURNAL OF THE HEART TEAM 2022; 6:100086. [PMID: 37288061 PMCID: PMC10242580 DOI: 10.1016/j.shj.2022.100086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 06/09/2023]
Abstract
Background Endovascular baroreflex amplification with the MobiusHD, a self-expanding stent-like device that is implanted in the internal carotid artery, was designed to reduce the sympathetic overactivity that contributes to progressive heart failure with reduced ejection fraction. Methods Symptomatic patients (New York Heart Association class III) with heart failure with reduced ejection fraction (left ventricular ejection fraction [LVEF] ≤40%) despite guideline directed medical therapy and n-terminal pro-B type natriuretic peptide (NT-proBNP) levels ≥400 pg/mL in whom carotid ultrasound and computed tomographic angiography demonstrated absence of carotid plaque were enrolled. Baseline and follow-up measures included 6-minute walk distance (6MWD), Kansas City Cardiomyopathy Questionnaire overall summary score (KCCQ OSS), and repeat biomarkers and transthoracic echocardiography. Results Twenty-nine patients underwent device implantation. The mean age was 60.6 ± 11.4 years, and all had New York Heart Association class III symptoms. Mean KCCQ OSS was 41.4 ± 12.7, mean 6MWD was 216.0 ± 43.7 m, median NT-proBNP was 1005.9 pg/mL (894, 1294), and mean LVEF was 34.7 ± 2.9%. All device implantations were successful. Two patients died (161 days and 195 days) and one stroke occurred (170 days) during follow-up. For the 17 patients with 12-month follow-up, mean KCCQ OSS improved by 17.4 ± 9.1 points, mean 6MWD increased by 97.6 ± 51.1 meters, a mean 28.4% reduction from the baseline NT-proBNP concentration was found, and mean LVEF improved by 5.6% ± 2.9 (paired data). Conclusion Endovascular baroreflex amplification with the MobiusHD device was safe and effected positive changes in quality of life, exercise capacity, and LVEF, consistent with observed reductions in NT-proBNP levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | - James L. Januzzi
- Massachusetts General Hospital, Harvard Medical School, Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | | | - Gregg W. Stone
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
108
|
Abstract
BACKGROUND Chronic heart failure (CHF) is associated with redox imbalance. Downregulation of Nrf2 (nuclear factor [erythroid-derived 2]-like 2) plays important roles in disrupting myocardial redox homeostasis and mediating sympathetic nerve activity in the setting of CHF. However, it is unclear if circulating extracellular vesicles (EVs) elicit sympathetic excitation in CHF by disrupting central redox homeostasis. We tested the hypothesis that cardiac-derived EVs circulate to the presympathetic rostral ventrolateral medulla and contribute to oxidative stress and sympathetic excitation via EV-enriched microRNA-mediated Nrf2 downregulation. METHODS Data were collected on rats with CHF post-myocardial infarction (MI) and on human subjects with ischemic CHF. EVs were isolated from tissue and plasma, and we determined the miRNAs cargo that related to targeting Nrf2 translation. We tracked the distribution of cardiac-derived EVs using in vitro labeled circulating EVs and cardiac-specific membrane GFP+ transgenic mice. Finally, we tested the impact of exogenously loading of antagomirs to specific Nrf2-related miRNAs on CHF-EV-induced pathophysiological phenotypes in normal rats (eg, sympathetic and cardiac function). RESULTS Nrf2 downregulation in CHF rats was associated with an upregulation of Nrf2-targeting miRNAs, which were abundant in cardiac-derived and circulating EVs from rats and humans. EVs isolated from the brain of CHF rats were also enriched with Nrf2-targeting miRNAs and cardiac-specific miRNAs. Cardiac-derived EVs were taken up by neurons in the rostral ventrolateral medulla. The administration of cardiac-derived and circulating EVs from CHF rats into the rostral ventrolateral medulla of normal rats evoked an increase in renal sympathetic nerve activity and plasma norepinephrine compared with Sham-operated rats, which were attenuated by exogenously preloading CHF-EVs with antagomirs to Nrf2-targeting miRNAs. CONCLUSIONS Cardiac microRNA-enriched EVs from animals with CHF can mediate crosstalk between the heart and the brain in the regulation of sympathetic outflow by targeting the Nrf2/antioxidant signaling pathway. This new endocrine signaling pathway regulating sympathetic outflow in CHF may be exploited for novel therapeutics.
Collapse
Affiliation(s)
- Changhai Tian
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536
| | - Lie Gao
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tara L. Rudebush
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Li Yu
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
109
|
Paolisso P, Dagan A, Gallinoro E, De Colle C, Bertolone DT, Moya A, Penicka M, Degrieck I, Vanderheyden M, Bartunek J. Aortic thoracic neuromodulation in heart failure with preserved ejection fraction. ESC Heart Fail 2022; 10:699-704. [PMID: 36151858 PMCID: PMC9871658 DOI: 10.1002/ehf2.14136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/07/2022] [Accepted: 08/24/2022] [Indexed: 01/27/2023] Open
Abstract
The inadequacy of medical therapies for heart failure with preserved ejection fraction (HFpEF) is driving the development of device-based solutions targeting underlying pathophysiologic abnormalities. The maladaptive autonomic imbalance with a reduction in vagal parasympathetic activity and increased sympathetic signalling contributes to the deterioration of cardiac performance, patient fitness, and the increased overall morbidity and mortality. Thoracic aortic vagal afferents mediate parasympathetic signalling, and their stimulation has been postulated to restore autonomic balance. In this first-in-man experience with chronic stimulation of aortic vagal afferents (Harmony™ System, Enopace, Israel), we demonstrate improved left atrial remodelling and function parallel with improved left ventricular performance. The observed favourable structural and functional cardiac changes remained stable throughout the 1 year follow-up and were associated with improved symptoms and physical fitness. The current experience warrants further validation of the endovascular stimulation of aortic thoracic afferents as a new interventional approach for device-based treatment in HFpEF.
Collapse
Affiliation(s)
- Pasquale Paolisso
- Cardiovascular Center Aalst, OLV HospitalAalstBelgium,Department of Advanced Biomedical SciencesUniversity of Naples Federico IINaplesItaly
| | | | - Emanuele Gallinoro
- Cardiovascular Center Aalst, OLV HospitalAalstBelgium,Department of Translational Medical SciencesUniversity of Campania' Luigi Vanvitelli'NaplesItaly
| | - Cristina De Colle
- Cardiovascular Center Aalst, OLV HospitalAalstBelgium,Department of Advanced Biomedical SciencesUniversity of Naples Federico IINaplesItaly
| | - Dario Tino Bertolone
- Cardiovascular Center Aalst, OLV HospitalAalstBelgium,Department of Advanced Biomedical SciencesUniversity of Naples Federico IINaplesItaly
| | - Ana Moya
- Cardiovascular Center Aalst, OLV HospitalAalstBelgium
| | | | - Ivan Degrieck
- Cardiovascular and Thoracic Surgery, OLV‐ClinicAalstBelgium
| | | | | |
Collapse
|
110
|
Popa IP, Haba MȘC, Mărănducă MA, Tănase DM, Șerban DN, Șerban LI, Iliescu R, Tudorancea I. Modern Approaches for the Treatment of Heart Failure: Recent Advances and Future Perspectives. Pharmaceutics 2022; 14:1964. [PMID: 36145711 PMCID: PMC9503448 DOI: 10.3390/pharmaceutics14091964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure (HF) is a progressively deteriorating medical condition that significantly reduces both the patients' life expectancy and quality of life. Even though real progress was made in the past decades in the discovery of novel pharmacological treatments for HF, the prevention of premature deaths has only been marginally alleviated. Despite the availability of a plethora of pharmaceutical approaches, proper management of HF is still challenging. Thus, a myriad of experimental and clinical studies focusing on the discovery of new and provocative underlying mechanisms of HF physiopathology pave the way for the development of novel HF therapeutic approaches. Furthermore, recent technological advances made possible the development of various interventional techniques and device-based approaches for the treatment of HF. Since many of these modern approaches interfere with various well-known pathological mechanisms in HF, they have a real ability to complement and or increase the efficiency of existing medications and thus improve the prognosis and survival rate of HF patients. Their promising and encouraging results reported to date compel the extension of heart failure treatment beyond the classical view. The aim of this review was to summarize modern approaches, new perspectives, and future directions for the treatment of HF.
Collapse
Affiliation(s)
- Irene Paula Popa
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Mihai Ștefan Cristian Haba
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Minela Aida Mărănducă
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Maria Tănase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700115 Iași, Romania
| | - Dragomir N. Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Radu Iliescu
- Department of Pharmacology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ionuț Tudorancea
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
111
|
Arruda-Junior DF, Salles TA, Martins FL, Antonio EL, Tucci PJF, Gowdak LHW, Tavares CAM, Girardi AC. Unraveling the interplay between dipeptidyl peptidase 4 and the renin-angiotensin system in heart failure. Life Sci 2022; 305:120757. [PMID: 35780844 DOI: 10.1016/j.lfs.2022.120757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
Abstract
AIMS Emerging evidence suggests the existence of a crosstalk between dipeptidyl peptidase 4 (DPP4) and the renin-angiotensin system (RAS). Therefore, combined inhibition of DPP4 and RAS may produce similar pharmacological effects rather than being additive. This study tested the hypothesis that combining an inhibitor of DPP4 with an angiotensin II (Ang II) receptor blocker does not provide additional cardioprotection compared to monotherapy in heart failure (HF) rats. MAIN METHODS Male Wistar rats were subjected to left ventricle (LV) radiofrequency ablation or sham operation. Six weeks after surgery, radiofrequency-ablated rats who developed HF were assigned into four groups and received vehicle (water), vildagliptin, valsartan, or both drugs, for four weeks by oral gavage. KEY FINDINGS Vildagliptin and valsartan in monotherapy reduced LV hypertrophy, alleviated cardiac interstitial fibrosis, and improved systolic and diastolic function in HF rats, with no additional effect of combination treatment. HF rats displayed higher cardiac and serum DPP4 activity and abundance than sham. Surprisingly, not only vildagliptin but also valsartan in monotherapy downregulated the catalytic function and expression levels of systemic and cardiac DPP4. Moreover, vildagliptin and valsartan alone or in combination comparably upregulate the components of the cardiac ACE2/Ang-(1-7)/MasR while downregulating the ACE/Ang II/AT1R axis. SIGNIFICANCE Vildagliptin or valsartan alone is as effective as combined to treat cardiac dysfunction and remodeling in experimental HF. DPP4 inhibition downregulates classic RAS components, and pharmacological RAS blockade downregulates DPP4 in the heart and serum of HF rats. This interplay between DPP4 and RAS may affect HF progression and pharmacotherapy.
Collapse
Affiliation(s)
- Daniel F Arruda-Junior
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Thiago A Salles
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Flavia L Martins
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ednei L Antonio
- Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paulo J F Tucci
- Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luís Henrique W Gowdak
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Caio A M Tavares
- Unidade de Cardiogeriatria, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Academic Research Organization (ARO), Hospital Israelita Albert Eistein, São Paulo, São Paulo, Brazil
| | - Adriana C Girardi
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
112
|
Jia R, Ji Y, Sun D. Progress and prospects of Sacubitril/Valsartan: Based on heart failure with preserved ejection fraction. Biomed Pharmacother 2022; 155:113701. [PMID: 36116249 DOI: 10.1016/j.biopha.2022.113701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is present in nearly half of patients with heart failure. The prevalence of heart failure with normal or near-normal ejection fractions increases more rapidly than in patients with reduced ejection fractions. Angiotensin-converting enzyme inhibitor (ACEI), angiotensin receptor blocker (ARB), aldosterone antagonist, β-blocker, and calcium channel blocker have not shown significant efficacy in HFpEF clinical trials. Sacubitril/Valsartan, combined angiotensin receptor blocker (Valsartan) with neprilysin inhibitor (Sacubitril), was the first-of-its-kind angiotensin receptor-neprilysin inhibitor (ARNI) to be developed. It has shown significant efficacy on HFpEF in recent studies. It is considered that most of the current Sacubitril/Valsartan studies are still concentrated in the field of heart failure, especially heart failure with reduced ejection fraction (HFrEF). This review discusses the latest advances in cardiovascular, renal, and metabolic aspects of Sacubitril/Valsartan, mainly in HFpEF, providing more evidence for further future research on Sacubitril/Valsartan and raising issues that should be paid attention. At the same time, this review will introduce the academic consensus on Sacubitril/Valsartan in treating HFpEF in China.
Collapse
Affiliation(s)
- Ruoyu Jia
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Graduate School, Xuzhou Medical University, Xuzhou 221002, China
| | - Ying Ji
- Graduate School, Xuzhou Medical University, Xuzhou 221002, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nephrology, Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
113
|
Sandroni PB, Fisher-Wellman KH, Jensen BC. Adrenergic Receptor Regulation of Mitochondrial Function in Cardiomyocytes. J Cardiovasc Pharmacol 2022; 80:364-377. [PMID: 35170492 PMCID: PMC9365878 DOI: 10.1097/fjc.0000000000001241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/01/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Adrenergic receptors (ARs) are G protein-coupled receptors that are stimulated by catecholamines to induce a wide array of physiological effects across tissue types. Both α1- and β-ARs are found on cardiomyocytes and regulate cardiac contractility and hypertrophy through diverse molecular pathways. Acute activation of cardiomyocyte β-ARs increases heart rate and contractility as an adaptive stress response. However, chronic β-AR stimulation contributes to the pathobiology of heart failure. By contrast, mounting evidence suggests that α1-ARs serve protective functions that may mitigate the deleterious effects of chronic β-AR activation. Here, we will review recent studies demonstrating that α1- and β-ARs differentially regulate mitochondrial biogenesis and dynamics, mitochondrial calcium handling, and oxidative phosphorylation in cardiomyocytes. We will identify potential mechanisms of these actions and focus on the implications of these findings for the modulation of contractile function in the uninjured and failing heart. Collectively, we hope to elucidate important physiological processes through which these well-studied and clinically relevant receptors stimulate and fuel cardiac contraction to contribute to myocardial health and disease.
Collapse
Affiliation(s)
- Peyton B. Sandroni
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
| | - Kelsey H. Fisher-Wellman
- East Carolina University Brody School of Medicine, Department of Physiology
- East Carolina University Diabetes and Obesity Institute
| | - Brian C. Jensen
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
- University of North Carolina School of Medicine, Department of Medicine, Division of Cardiology
| |
Collapse
|
114
|
Depes D, Mennander A, Vehniäinen R, Paavonen T, Kholová I. Human Pulmonary Vein Myocardial Sleeve Autonomic Neural Density and Cardiovascular Mortality. J Histochem Cytochem 2022; 70:627-642. [PMID: 36154512 PMCID: PMC9527475 DOI: 10.1369/00221554221129899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
Myocardial sleeves around pulmonary veins (PVs) are highly innervated structures with heterogeneous morphological and electrophysiological characteristics. Autonomic nerve dysfunction in the myocardium may be associated with an increased risk of cardiovascular morbidity and mortality. This article studied autonomic neural remodeling in myocardial sleeves around PVs and atrial-PV ostia with immunohistochemical and morphometric methods with clinicopathological correlations. PVs were collected from 37 and atrial-PV ostia from 17 human autopsy hearts. Immunohistochemical analysis was performed using antibodies against tyrosine hydroxylase (TH), choline acetyltransferase (CHAT), and growth-associated protein 43 (GAP43). In the PV cohort, subjects with immediate cardiovascular cause of death had significantly decreased sympathetic nerve density in fibro-fatty tissue vs those with non-cardiovascular cause of death (1624.53 vs 2522.05 µm2/mm2, p=0.038). In the atrial-PV ostia cohort, parasympathetic nerve density in myocardial sleeves was significantly increased in subjects with underlying cardiovascular cause of death (19.48 µm2/mm2) than subjects with underlying non-cardiovascular cause of death with no parasympathetic nerves detected (p=0.034). Neural growth regionally varied in sympathetic nerves and was present in most of the parasympathetic nerves. Heterogeneous autonomic nerve distribution and growth around PVs and atrial-PV ostia might play a role in cardiovascular morbidity and mortality. No association in nerve density was found with atrial fibrillation.
Collapse
Affiliation(s)
- Denis Depes
- Department of Pathology, Fimlab Laboratories,
Tampere, Finland
- Faculty of Medicine and Health Technology,
Tampere University, Tampere, Finland
| | - Ari Mennander
- Faculty of Medicine and Health Technology,
Tampere University, Tampere, Finland
- Division of Cardiothoracic Surgery, Tampere
University Heart Hospital, Tampere, Finland
| | - Rauha Vehniäinen
- Faculty of Medicine and Health Technology,
Tampere University, Tampere, Finland
| | - Timo Paavonen
- Department of Pathology, Fimlab Laboratories,
Tampere, Finland
- Faculty of Medicine and Health Technology,
Tampere University, Tampere, Finland
| | - Ivana Kholová
- Department of Pathology, Fimlab Laboratories,
Tampere, Finland
- Faculty of Medicine and Health Technology,
Tampere University, Tampere, Finland
| |
Collapse
|
115
|
Iftikhar IH, Khayat RN. Central sleep apnea treatment in patients with heart failure with reduced ejection fraction: a network meta-analysis. Sleep Breath 2022; 26:1227-1235. [PMID: 34698980 DOI: 10.1007/s11325-021-02512-y] [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: 06/22/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Adaptive servo-ventilation (ASV) is contraindicated for the treatment of central sleep apnea (CSA) in patients with heart failure with reduced ejection fraction (HFrEF), limiting treatment options. Though continuous positive airway pressure (CPAP), bi-level PAP with back-up rate (BPAP-BUR), and transvenous phrenic nerve stimulation (TPNS) are alternatives, not much is known about their comparative efficacies, which formed the basis of conducting this network meta-analysis. We sought to analyze their comparative effectiveness in reducing apnea hypopnea index (AHI). Additionally, we also studied their comparative effectiveness on subjective daytime sleepiness as assessed by Epworth sleepiness score (ESS). METHODS Randomized controlled trials (RCTs) from PubMed were analyzed in a network meta-analysis and relative superiority was computed based on P-score ranking and Hasse diagrams. RESULTS Network meta-analysis based on 8 RCTs showed that when compared to guideline-directed medical therapy (GDMT-used as a common comparator across trials), reduction in AHI by ASV (- 26.05 [- 38.80; - 13.31]), TPNS (- 24.90 [- 42.88; - 6.92]), BPAP-BUR (- 20.36 [- 36.47; - 4.25]), and CPAP (- 16.01 [- 25.42; - 6.60]) were statistically significant but not between the interventions. Based on 6 RCTs of all the interventions, only TPNS showed a statistically significant decrease in ESS (- 3.70 (- 5.58; - 1.82)) when compared to GDMT, while also showing significant differences when compared with ASV (- 3.20 (- 5.86; - 0.54)), BPAP-BUR (- 4.00 (- 7.33; - 0.68)), and CPAP (- 4.45 (- 7.75; - 1.14)). Ranking of treatments based on Hasse diagram, accounting for both AHI and ESS as outcomes for relative hierarchy showed relative superiority of both ASV and TPNS over BPAP-BUR and CPAP. CONCLUSIONS Results indicated relative superiority of TPNS and ASV to BPAP-BUR and CPAP in their effects on AHI and ESS.
Collapse
Affiliation(s)
- Imran H Iftikhar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University School of Medicine, 613 Michael St, NE, Atlanta, GA, 30322, USA.
- Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA.
| | - Rami N Khayat
- Department of Medicine, Division of Pulmonary Diseases and Critical Care Medicine, School of Medicine, University of California, Irvine, Irvine, CA, 92617, USA
| |
Collapse
|
116
|
Escobar C, Luis-Bonilla J, Crespo-Leiro MG, Esteban-Fernández A, Farré N, Garcia A, Nuñez J. Individualizing the treatment of patients with heart failure with reduced ejection fraction: a journey from hospitalization to long-term outpatient care. Expert Opin Pharmacother 2022; 23:1589-1599. [PMID: 35995759 DOI: 10.1080/14656566.2022.2116275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Despite the relevant advances achieved thanks to the traditional step-by-step therapeutic approach, heart failure with reduced ejection fraction (HFrEF) remains associated with considerable morbidity and mortality. The pathogenesis of HFrEF is complex, with the implication of various neurohormonal systems, including activation of deleterious pathways (i.e. renin-angiotensin-aldosterone, sympathetic, and sodium-glucose cotransporter-2 [SGLT2] systems) and the inhibition of protective pathways (i.e. natriuretic peptides and the guanylate cyclase system). Therefore, the burden of HF can only be reduced through a comprehensive approach that involves all evidence-based use of available HF drugs targeting the neurohormonal systems involved. AREAS COVERED : We performed a critical analysis of evidence from recent clinical trials and assessed the effects of HF therapies on hemodynamics and renal function. EXPERT OPINION : HF therapy must be adapted to the clinical profile (i.e. congestion, blood pressure, heart rate, renal function, and electrolytes). Consequently, blood pressure is reduced by beta blockers, renin-angiotensin-aldosterone system inhibitors, sacubitril/valsartan, and, minimally, by SGLT2 inhibitors and vericiguat; heart rate decreases with beta blockers and ivabradine; and renal function is impaired and potassium are levels increased with renin-angiotensin-aldosterone system inhibitors and sacubitril/valsartan. Practical recommendations on how to individualize HF therapy according to patient profile are provided.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Service, Hospital Universitario La Paz, Madrid, Spain
| | | | | | | | | | - Ana Garcia
- Hospital Clinic I Provincial De Barcelona, Barcelona, Spain
| | - Julio Nuñez
- Hospital Clinico de Valencia, Valencia, Spain
| |
Collapse
|
117
|
Muslimova E, Rebrova T, Kondratieva D, Korepanov V, Sonduev E, Kozlov B, Afanasiev S. Expression of the β1-adrenergic receptor (ADRB1) gene in the myocardium and β-adrenergic reactivity of the body in patients with a history of myocardium infraction. Gene 2022; 844:146820. [PMID: 35987510 DOI: 10.1016/j.gene.2022.146820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022]
Abstract
β1-adrenergic receptors (β1-AR) directly affect on intracardiac hemodynamic and the ability of the heart to tolerate physical activity by regulating its inotropic and chronotropic functions. Severe hypersympathicotonia, specific to coronary artery disease (CAD) and chronic heart failure (HF), leads to impaired functioning of β1-AR. The aim of this research was to assess the expression level of the β1-AR ADRB1 gene in the myocardium, to evaluate the β-adrenergic reactivity of the membrane (β-ARM) of erythrocytes, and to analyze the association of these parameters with myocardial contractile dysfunction in patients with a myocardial infarction (MI) in the past and without it. The study included 126 patients with chronic CAD. Among the patients, 55.6 % had a history of MI at least 6 months ago. The expression of the ADRB1 gene was assessed using real-time polymerase chain reaction. With this purpose, we isolated RNA from the right atrial appendage, which was excised when a heart-lung machine was connected during a planned coronary bypass surgery. β-ARM was evaluated in 57 patients. This method is based on the fact of inhibition of hemolysis of erythrocytes, placed in a hyposmotic medium, in the presence of a β-blocker. Within the whole sample of patients, the expression of the ADRB1 gene is comparable in different functional classes of HF. There was no linear correlation between the expression of the ADRB1 gene and left ventricle ejection fraction (LVEF). In patients with a history of MI, the expression of the ADRB1 gene was elevated when compared to a group of patients without MI (p = 0.017). Patients with a history of MI had higher values of β-ARM than those without MI (p = 0.017). The reverse correlation between β-ARM and LVEF (r = -0,570, p = 0,002) was revealed in the group of patients without MI but not in the group of patients with a history of MI (r = -0,137, p = 0,479). In the sample of patients with chronic CAD, in the myocardium of subjects with a history of MI, the relative expression of ADRB1 gene was higher compared to the group of patients without MI. In patients with different functional classes (FC) of HF and with different ejection fraction, both with MI and without it, ADRB1 gene expression was comparable. In the group of patients with a history of MI, an increase in β-ARM was observed, i.e. decrease in the number or sensitivity of β-AR. Among patients without MI, an inverse correlation was found between β-ARM and LVEF.
Collapse
Affiliation(s)
- Elvira Muslimova
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.
| | - Tatiana Rebrova
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Dina Kondratieva
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Viacheslav Korepanov
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Erdeni Sonduev
- Department of Cardiovascular Surgery, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Boris Kozlov
- Department of Cardiovascular Surgery, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey Afanasiev
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
118
|
Gronda E, Dusi V, D’Elia E, Iacoviello M, Benvenuto E, Vanoli E. Sympathetic activation in heart failure. Eur Heart J Suppl 2022; 24:E4-E11. [PMID: 35991621 PMCID: PMC9385124 DOI: 10.1093/eurheartjsupp/suac030] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Sympathetic activation has been long appreciated exclusively as a fundamental compensatory mechanism of the failing heart and, thus, welcome and to be supported. In the initial clinical phases of heart failure (HF), the sympathetic nervous system overdrive plays a compensatory function aimed at maintaining an adequate cardiac output despite the inotropic dysfunction affecting the myocardium. However, when the sympathetic reflex response is exaggerated it triggers a sequence of unfavourable remodelling processes causing a further contractile deterioration that unleashes major adverse cardiovascular consequences, favouring the HF progression and the occurrence of fatal events. Eventually, the sympathetic nervous system in HF was demonstrated to be a ‘lethality factor’ and thus became a prominent therapeutic target. The existence of an effective highly specialized intracardiac neuronal network immediately rules out the old concept that sympathetic activation in HF is merely the consequence of a drop in cardiac output. When a cardiac damage occurs, such as myocardial ischaemia or a primary myocardial disorder, the adaptive capability of the system may be overcame, leading to excessive sympatho-excitation coupled with attenuation till to abolishment of central parasympathetic drive. Myocardial infarction causes, within a very short time, both a functional and anatomical remodelling with a diffuse up-regulation of nerve growth factor (NGF). The subsequent nerve sprouting signal, facilitated by a rise in the levels of NGF in the left stellate ganglion and in the serum, triggers an increase in cardiac nerve density in both peri-infarct and non-infarcted areas. Finally, NFG production decreases over time, supposedly as an adaptative response to the prolonged exposure to sympathetic overactivity, leading in the end to a reduction in sympathetic nerve density. Accordingly, NGF levels were markedly reduced in patients with severe congestive heart failure. The kidney is the other key player of the sympathetic response to HF as it indeed reacts to under-perfusion and to loop diuretics to preserve filtration at the cost of many pathological consequences on its physiology. This vicious loop ultimately participates to the chronic and disruptive sympathetic overdrive. In conclusion, sympathetic activation is the natural physiological consequence to life stressors but also to any condition that may harm our body. It is the first system of reaction to any potential life-threatening event. However, in any aspect of life over reaction is never effective but, in many instances, is, actually, life threatening. One for all is the case of ischaemia-related ventricular fibrillation which is, strongly facilitated by sympathetic hyperactivity. The take home message? When, in a condition of harm, everybody is yelling failure is just around the corner.
Collapse
Affiliation(s)
- E Gronda
- U.O.C. Nefrologia, Dialisi e Trapianto Renale dell’Adulto, Programma Cardiorenale, Dipartimento di Medicina e Specialità Mediche, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico , Milano , Italy
- Area Cardiorenale Metabolica Associazione nazionale Medici Cardiologi Ospedalieri Italia
| | - V Dusi
- Cardiology Division, Department of Medical Sciences, University of Turin , Torino , Italy
| | - E D’Elia
- Cardiovascular Department, Papa Giovanni XXIII Hospital , Bergamo , Italy
| | - M Iacoviello
- Area Cardiorenale Metabolica Associazione nazionale Medici Cardiologi Ospedalieri Italia
- S.C. Cardiologia, AOU Policlinico Riuniti di Foggia, Dipartimento di Scienze Mediche e Chirurgiche, Università degli Studi , Foggia , Italy
| | - E Benvenuto
- Area Cardiorenale Metabolica Associazione nazionale Medici Cardiologi Ospedalieri Italia
- U.O.C. di Cardiologia-UTIC-Emodinamica PO ‘G. Mazzini’ Teramo , Italy
| | - E Vanoli
- Department of Molecular Medicine, University of Pavia , Pavia , Italy
- Department of Medicine, Cardiology and Rehabilitation Sacra Famiglia Hospital , Erba , Italy
| |
Collapse
|
119
|
Jong J, Pinney JR, Packard RRS. Anthracycline-induced cardiotoxicity: From pathobiology to identification of molecular targets for nuclear imaging. Front Cardiovasc Med 2022; 9:919719. [PMID: 35990941 PMCID: PMC9381993 DOI: 10.3389/fcvm.2022.919719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Anthracyclines are a widely used class of chemotherapy in pediatric and adult cancers, however, their use is hampered by the development of cardiotoxic side-effects and ensuing complications, primarily heart failure. Clinically used imaging modalities to screen for cardiotoxicity are mostly echocardiography and occasionally cardiac magnetic resonance imaging. However, the assessment of diastolic and global or segmental systolic function may not be sensitive to detect subclinical or early stages of cardiotoxicity. Multiple studies have scrutinized molecular nuclear imaging strategies to improve the detection of anthracycline-induced cardiotoxicity. Anthracyclines can activate all forms of cell death in cardiomyocytes. Injury mechanisms associated with anthracycline usage include apoptosis, necrosis, autophagy, ferroptosis, pyroptosis, reactive oxygen species, mitochondrial dysfunction, as well as cardiac fibrosis and perturbation in sympathetic drive and myocardial blood flow; some of which have been targeted using nuclear probes. This review retraces the pathobiology of anthracycline-induced cardiac injury, details the evidence to date supporting a molecular nuclear imaging strategy, explores disease mechanisms which have not yet been targeted, and proposes a clinical strategy incorporating molecular imaging to improve patient management.
Collapse
Affiliation(s)
- Jeremy Jong
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - James R. Pinney
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
- Veterans Affairs West Los Angeles Medical Center, Los Angeles, CA, United States
| | - René R. Sevag Packard
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
- Veterans Affairs West Los Angeles Medical Center, Los Angeles, CA, United States
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
120
|
Crnošija L, Moštak I, Višnjić N, Junaković A, Karić A, Adamec I, Krbot Skorić M, Habek M. Blood pressure variability is altered in secondary progressive multiple sclerosis but not in patients with a clinically isolated syndrome. Neurophysiol Clin 2022; 52:290-298. [PMID: 35907760 DOI: 10.1016/j.neucli.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/17/2022] [Accepted: 06/17/2022] [Indexed: 10/16/2022] Open
Abstract
OBJECTIVES To investigate differences in beat-to-beat systolic blood pressure variability (SBPV) in people with secondary progressive MS (pwSPMS), clinically isolated syndrome (pwCIS) and healthy controls (HC). METHODS This retrospective case-control study included 46 pwSPMS, 46 pwCIS and 44 HC. A semi-automated software made with MATLAB R2019b (The MathWorks, Inc.) was used for the evaluation of SBPV. The frequency domain characteristics observed were the power spectrum in the LF and HF bands and the LF/HF ratio. Data is expressed in absolute power (mmHg2) of LF and HF and ratio (LF/HF) during both supine and tilt-up phases of testing. RESULTS There were no significant differences in mean systolic (sBP) or diastolic blood pressure (dBP) values during supine and tilt-up phases of testing between groups. During the supine phase of testing LF and LF/HF were significantly lower in the SPMS group (4.17±5.38 and 3.52±2.34, respectively) compared to the CIS (5.42±3.59, p = 0.015 and 5.92±4.63, p = 0.029, respectively) and HC group (6.03±4.55, p = 0.011 and 6.52 ± 5.09, p = 0.010, respectively), while during the tilt-up phase, LF was significantly lower compared to both the CIS and HC group, and HF was significantly lower only compared to the CIS group. CONCLUSION SBPV is altered in pwSPMS compared to pwCIS and normal controls. Further research in the field of MS related dysautonomia is warranted not only because of its relevance to comorbidities and MS symptoms, but also because of its likely involvement in the pathophysiology of MS.
Collapse
Affiliation(s)
- Luka Crnošija
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia
| | - Ivan Moštak
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nika Višnjić
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anamari Junaković
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia
| | - Antea Karić
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia
| | - Ivan Adamec
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Magdalena Krbot Skorić
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; Faculty of Electrical Engineering and Computing, University of Zagreb, Zagreb, Croatia
| | - Mario Habek
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
121
|
Impact of R-Carvedilol on β2-Adrenergic Receptor-Mediated Spontaneous Calcium Release in Human Atrial Myocytes. Biomedicines 2022; 10:biomedicines10071759. [PMID: 35885069 PMCID: PMC9313410 DOI: 10.3390/biomedicines10071759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 01/14/2023] Open
Abstract
A hallmark of atrial fibrillation is an excess of spontaneous calcium release events, which can be mimicked by β1- or β2-adrenergic stimulation. Because β1-adrenergic receptor blockers (β1-blockers) are primarily used in clinical practice, we here examined the impact of β2-adrenergic stimulation on spontaneous calcium release and assessed whether the R- and S-enantiomers of the non-selective β- blocker carvedilol could reverse these effects. For this purpose, human atrial myocytes were isolated from patients undergoing cardiovascular surgery and subjected to confocal calcium imaging or immunofluorescent labeling of the ryanodine receptor (RyR2). Interestingly, the β2-adrenergic agonist fenoterol increased the incidence of calcium sparks and waves to levels observed with the non-specific β-adrenergic agonist isoproterenol. Moreover, fenoterol increased both the amplitude and duration of the sparks, facilitating their fusion into calcium waves. Subsequent application of the non β-blocking R-Carvedilol enantiomer reversed these effects of fenoterol in a dose-dependent manner. R-Carvedilol also reversed the fenoterol-induced phosphorylation of the RyR2 at Ser-2808 dose-dependently, and 1 µM of either R- or S-Carvedilol fully reversed the effect of fenoterol. Together, these findings demonstrate that β2-adrenergic stimulation alone stimulates RyR2 phosphorylation at Ser-2808 and spontaneous calcium release maximally, and points to carvedilol as a tool to attenuate the pathological activation of β2-receptors.
Collapse
|
122
|
Li P, Jin C, Cui C, Cai P, Manohar SA, Jin L, Wei X, Pan S, Dixon RAF, Liu Q. Impact of family history of coronary artery disease on clinical outcomes in Takotsubo cardiomyopathy. J Investig Med 2022; 70:jim-2021-002186. [PMID: 35850971 DOI: 10.1136/jim-2021-002186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 11/03/2022]
Abstract
Family history of coronary artery disease (FHxCAD) is a critical risk factor for CAD, underscoring the contribution of genetic factors to disease pathogenesis and susceptibility. Takotsubo cardiomyopathy (TCM) simulates the clinical features of and frequently coexists with CAD. However, the association between FHxCAD and TCM is unclear. Here, we retrospectively examined the impact of FHxCAD on in-hospital outcomes of patients with TCM. Using the National Inpatient Sample database (2016-2018), we identified 4733 patients admitted to hospital with a primary diagnosis of TCM. We compared in-hospital outcomes and complications between TCM patients with (n=646, 13.7%) and without FHxCAD (n=646) in the unmatched and in a propensity-score matched cohort (1:1 ratio). TCM with FHxCAD patients had a reduced incidence of cardiogenic shock, acute kidney injury (AKI), and acute respiratory failure (ARF); lower mortality rates; shorter length of stay (LOS); and decreased total charge compared with TCM without FHxCAD patients (p<0.05). In the matched cohort, TCM with FHxCAD patients (vs TCM without FHxCAD patients) had a lower incidence of cardiogenic shock (2.2% vs 6.3%, p<0.001; OR 0.33, 95% CI 0.18 to 0.61), AKI (5.1% vs 8.7%, p=0.016; OR 0.57, 95% CI 0.36 to 0.88), and ARF (5.7% vs 12.7%, p<0.001; OR 0.42, 95% CI 0.28 to 0.63); decreased in-hospital mortality (<11% vs 3.1%, p=0.002; OR 0.2, 95% CI 0.07 to 0.57); shorter LOS (2.66±1.96 days vs 3.40±3.05 days, p<0.001); and a reduced total charge (p=0.001), respectively. FHxCAD was associated with favorable outcomes in both unmatched and propensity-matched cohorts.
Collapse
Affiliation(s)
- Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Chengyue Jin
- Department of Medicine, Westchester Medical Center, Valhalla, New York, USA
| | - Can Cui
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Peng Cai
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Shamita Alisa Manohar
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ling Jin
- Department of Medicine, Metrowest Medical Center, Framingham, Massachusetts, USA
| | - Xin Wei
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Su Pan
- Wafic Said Molecular Cardiology Research Laboratory, Texas Heart Institute, Houston, Texas, USA
| | - Richard A F Dixon
- Wafic Said Molecular Cardiology Research Laboratory, Texas Heart Institute, Houston, Texas, USA
| | - Qi Liu
- Wafic Said Molecular Cardiology Research Laboratory, Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
123
|
Wang Y, Zhao M, Xu B, Bahriz SMF, Zhu C, Jovanovic A, Ni H, Jacobi A, Kaludercic N, Di Lisa F, Hell JW, Shih JC, Paolocci N, Xiang YK. Monoamine oxidase A and organic cation transporter 3 coordinate intracellular β 1AR signaling to calibrate cardiac contractile function. Basic Res Cardiol 2022; 117:37. [PMID: 35842861 PMCID: PMC9288959 DOI: 10.1007/s00395-022-00944-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/03/2023]
Abstract
We have recently identified a pool of intracellular β1 adrenergic receptors (β1ARs) at the sarcoplasmic reticulum (SR) crucial for cardiac function. Here, we aim to characterize the integrative control of intracellular catecholamine for subcellular β1AR signaling and cardiac function. Using anchored Förster resonance energy transfer (FRET) biosensors and transgenic mice, we determined the regulation of compartmentalized β1AR-PKA signaling at the SR and plasma membrane (PM) microdomains by organic cation transporter 3 (OCT3) and monoamine oxidase A (MAO-A), two critical modulators of catecholamine uptake and homeostasis. Additionally, we examined local PKA substrate phosphorylation and excitation-contraction coupling in cardiomyocyte. Cardiac-specific deletion of MAO-A (MAO-A-CKO) elevates catecholamines and cAMP levels in the myocardium, baseline cardiac function, and adrenergic responses. Both MAO-A deletion and inhibitor (MAOi) selectively enhance the local β1AR-PKA activity at the SR but not PM, and augment phosphorylation of phospholamban, Ca2+ cycling, and myocyte contractile response. Overexpression of MAO-A suppresses the SR-β1AR-PKA activity and PKA phosphorylation. However, deletion or inhibition of OCT3 by corticosterone prevents the effects induced by MAOi and MAO-A deletion in cardiomyocytes. Deletion or inhibition of OCT3 also negates the effects of MAOi and MAO-A deficiency in cardiac function and adrenergic responses in vivo. Our data show that MAO-A and OCT3 act in concert to fine-tune the intracellular SR-β1AR-PKA signaling and cardiac fight-or-flight response. We reveal a drug contraindication between anti-inflammatory corticosterone and anti-depressant MAOi in modulating adrenergic regulation in the heart, providing novel perspectives of these drugs with cardiac implications.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Meimi Zhao
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- Department of Pharmaceutical Toxicology, China Medical University, Shenyang, 110122, China
| | - Bing Xu
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- VA Northern California Health Care System, Mather, CA, USA
| | - Sherif M F Bahriz
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Chaoqun Zhu
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Aleksandra Jovanovic
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Haibo Ni
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Ariel Jacobi
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy, Padua, Italy
- Institute for Pediatric Research Città Della Speranza, Padua, Italy
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy, Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Johannes W Hell
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA.
- VA Northern California Health Care System, Mather, CA, USA.
| |
Collapse
|
124
|
Kim Y, Canada JM, Kenyon J, Billingsley H, Arena R, Lavie CJ, Carbone S. Physical activity, sedentary behaviors and all-cause mortality in patients with heart failure: Findings from the NHANES 2007–2014. PLoS One 2022; 17:e0271238. [PMID: 35839246 PMCID: PMC9286289 DOI: 10.1371/journal.pone.0271238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/24/2022] [Indexed: 12/03/2022] Open
Abstract
Background Limited data are available examining the effects of both moderate- and vigorous-intensity physical activity (MVPA) and sedentary behavior (SB) on longevity among patients with heart failure (HF). This study examined the associations of MVPA and SB with all-cause mortality in HF patients using a nationally representative survey data. Methods National Health and Nutrition Examination Survey data (2007–2014) were used. 711 adults with self-reported congestive HF, linked to 2015 mortality data were analyzed. Self-reported MVPA and SB minutes were used to create the three MVPA [No-MVPA, insufficient (I-MVPA; <150 min/wk), and sufficient (S-MVPA; ≥150 min/wk)] and two SB (<8 and ≥8 hrs/d) groups. Cox proportional hazard models were constructed to test the associations of MVPA and SB with all-cause mortality. Results 119 deaths occurred over an average of 4.9 years of follow-up. Lower MVPA and higher SB were independently associated with poor survival (P < .001). Joint and stratified analyses showed that the protective effect of MVPA was most pronounced among patients with SB<8 hrs/d. There was no difference in the mortality risk by SB levels within I-MVPA and S-MVPA groups; however, in the No-MVPA group, those with SB≥8 hrs/d had a greater risk of mortality compared to those with <8 hrs/d (Hazard ratio = 1.60). Conclusion In this HF cohort, MVPA and SB were independently and jointly associated with all-cause mortality. The beneficial effect of MVPA is attenuated by excessive SB; however, engaging in some amount of MVPA may provide a protective effect and attenuates the detrimental effects associated with excessive SB.
Collapse
Affiliation(s)
- Youngdeok Kim
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Justin M. Canada
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Jonathan Kenyon
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Hayley Billingsley
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Carl J. Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-the University of Queensland School of Medicine, New Orleans, LA, United States of America
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, United States of America
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- * E-mail:
| |
Collapse
|
125
|
|
126
|
Bhuiyan MHU, Fard M, Robinson SR. Effects of whole-body vibration on driver drowsiness: A review. JOURNAL OF SAFETY RESEARCH 2022; 81:175-189. [PMID: 35589288 DOI: 10.1016/j.jsr.2022.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 09/29/2021] [Accepted: 02/14/2022] [Indexed: 05/19/2023]
Abstract
INTRODUCTION Whole-body vibration has direct impacts on driver vigilance by increasing physical and cognitive stress on the driver, which leads to drowsiness, fatigue and road traffic accidents. Although sleep deprivation, sleep apnoea and alcohol consumption can also lead to driver drowsiness, exposure to steady vibration is the factor most readily controlled by changes to vehicle design, yet it has received comparatively less attention. METHODS This review investigated interrelationships between the various components of whole-body vibration and the physiological and cognitive parameters that lead to driver drowsiness, as well as the effects of vibration parameters (frequency, amplitude, waveform and duration). Vibrations transmitted to the driver body from the vehicle floor and/or seat have been considered for this review, whereas hand-arm vibration, shocks, acute or transient vibration were excluded from consideration. RESULTS Drowsiness is affected by interactions between the frequency, amplitude, waveform and duration of the vibration. Under optimal conditions, whole-body vibration can induce significant drowsiness within 30 min. Low frequency whole-body vibrations, particularly vibrations of 4-10 Hz, are most effective at inducing drowsiness. This review notes some limitations of current studies and suggests directions for future research. CONCLUSIONS This review demonstrated a strong causal link exists between whole-body vibration and driver drowsiness. Since driver drowsiness has been established to be a significant contributor to motor vehicle accidents, research is needed to identify ways to minimise the components of whole-body vibration that contribute to drowsiness, as well as devising more effective ways to counteract drowsiness. PRACTICAL APPLICATIONS By raising awareness of the vibrational factors that contribute to drowsiness, manufacturers will be prompted to design vehicles that reduce the influence of these factors.
Collapse
Affiliation(s)
| | - Mohamad Fard
- School of Engineering, RMIT University, Melbourne, Australia
| | - Stephen R Robinson
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| |
Collapse
|
127
|
Son TK, Toan NH, Thang N, Le Trong Tuong H, Tien HA, Thuy NH, Van Minh H, Valensi P. Prediabetes and insulin resistance in a population of patients with heart failure and reduced or preserved ejection fraction but without diabetes, overweight or hypertension. Cardiovasc Diabetol 2022; 21:75. [PMID: 35568879 PMCID: PMC9107647 DOI: 10.1186/s12933-022-01509-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/07/2022] [Indexed: 12/17/2022] Open
Abstract
Background The relationships between glucose abnormalities, insulin resistance (IR) and heart failure (HF) are unclear, especially regarding to the HF type, i.e., HF with reduced (HFrEF) or preserved (HFpEF) ejection fraction. Overweight, diabetes and hypertension are potential contributors to IR in persons with HF. This study aimed to evaluate the prevalence of prediabetes and IR in a population of Vietnamese patients with HFrEF or HFpEF but no overweight, diabetes or hypertension, in comparison with healthy controls, and the relation between prediabetes or IR and HF severity. Methods We conducted a prospective cross-sectional observational study in 190 non-overweight normotensive HF patients (114 with HFrEF and 76 with HFpEF, 92.6% were ischemic HF, mean age was 70.1 years, mean BMI 19.7 kg/m2) without diabetes (neither known diabetes nor newly diagnosed by OGTT) and 95 healthy individuals (controls). Prediabetes was defined using 2006 WHO criteria. Glucose and insulin levels were measured fasting and 2 h after glucose challenge. IR was assessed using HOMA-IR and several other indexes. Results Compared to controls, HF patients had a higher prevalence of prediabetes (63.2% vs 22.1%) and IR (according to HOMA-IR, 55.3% vs 26.3%), higher HOMA-IR, insulin/glucose ratio after glucose and FIRI, and lower ISIT0 and ISIT120 (< 0.0001 for all comparisons), with no difference for body weight, waist circumference, blood pressure and lipid parameters. Prediabetes was more prevalent (69.3% vs 53.9%, p = 0.03) and HOMA-IR was higher (p < 0.0001) in patients with HFrEF than with HFpEF. Among both HFrEF and HFpEF patients, those with prediabetes or IR had a more severe HF (higher NYHA functional class and NT-proBNP levels, lower ejection fraction; p = 0.04–< 0.0001) than their normoglycemic or non-insulinresistant counterparts, with no difference for blood pressure and lipid parameters. Conclusion In non-diabetic non-overweight normotensive patients with HF, the prevalence of prediabetes is higher with some trend to more severe IR in those with HFrEF than in those with HFpEF. Both prediabetes and IR are associated with a more severe HF. The present data support HF as a culprit for IR. Intervention strategies should be proposed to HF patients with prediabetes aiming to reduce the risk of incident diabetes. Studies should be designed to test whether such strategies may translate into an improvement of further HF-related outcomes.
Collapse
Affiliation(s)
- Tran Kim Son
- Department of Internal Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Ngo Hoang Toan
- Department of Internal Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Nguyen Thang
- Science - Technology & External Relations Office, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | | | - Hoang Anh Tien
- Department of Internal Medicine, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Nguyen Hai Thuy
- Department of Internal Medicine, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Huynh Van Minh
- Department of Internal Medicine, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Paul Valensi
- Unit of Endocrinology-Diabetology-Nutrition. Jean Verdier hospital, APHP, Sorbonne Paris Nord University, CINFO, CRNH-IdF, Bondy, France.
| |
Collapse
|
128
|
Ljubojević-Holzer S, Kraler S, Djalinac N, Abdellatif M, Voglhuber J, Schipke J, Schmidt M, Kling KM, Franke GT, Herbst V, Zirlik A, von Lewinski D, Scherr D, Rainer PP, Kohlhaas M, Nickel A, Mühlfeld C, Maack C, Sedej S. Loss of autophagy protein ATG5 impairs cardiac capacity in mice and humans through diminishing mitochondrial abundance and disrupting Ca2+ cycling. Cardiovasc Res 2022; 118:1492-1505. [PMID: 33752242 PMCID: PMC9074988 DOI: 10.1093/cvr/cvab112] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
AIMS Autophagy protects against the development of cardiac hypertrophy and failure. While aberrant Ca2+ handling promotes myocardial remodelling and contributes to contractile dysfunction, the role of autophagy in maintaining Ca2+ homeostasis remains elusive. Here, we examined whether Atg5 deficiency-mediated autophagy promotes early changes in subcellular Ca2+ handling in ventricular cardiomyocytes, and whether those alterations associate with compromised cardiac reserve capacity, which commonly precedes the onset of heart failure. METHODS AND RESULTS RT-qPCR and immunoblotting demonstrated reduced Atg5 gene and protein expression and decreased abundancy of autophagy markers in hypertrophied and failing human hearts. The function of ATG5 was examined using cardiomyocyte-specific Atg5-knockout mice (Atg5-/-). Before manifesting cardiac dysfunction, Atg5-/- mice showed compromised cardiac reserve in response to β-adrenergic stimulation. Consequently, effort intolerance and maximal oxygen consumption were reduced during treadmill-based exercise tolerance testing. Mechanistically, cellular imaging revealed that Atg5 deprivation did not alter spatial and functional organization of intracellular Ca2+ stores or affect Ca2+ cycling in response to slow pacing or upon acute isoprenaline administration. However, high-frequency stimulation exposed stunted amplitude of Ca2+ transients, augmented nucleoplasmic Ca2+ load, and increased CaMKII activity, especially in the nuclear region of hypertrophied Atg5-/- cardiomyocytes. These changes in Ca2+ cycling were recapitulated in hypertrophied human cardiomyocytes. Finally, ultrastructural analysis revealed accumulation of mitochondria with reduced volume and size distribution, meanwhile functional measurements showed impaired redox balance in Atg5-/- cardiomyocytes, implying energetic unsustainability due to overcompensation of single mitochondria, particularly under increased workload. CONCLUSION Loss of cardiac Atg5-dependent autophagy reduces mitochondrial abundance and causes subtle alterations in subcellular Ca2+ cycling upon increased workload in mice. Autophagy-related impairment of Ca2+ handling is progressively worsened by β-adrenergic signalling in ventricular cardiomyocytes, thereby leading to energetic exhaustion and compromised cardiac reserve.
Collapse
Affiliation(s)
- Senka Ljubojević-Holzer
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Simon Kraler
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Nataša Djalinac
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Julia Voglhuber
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Julia Schipke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Marlene Schmidt
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078 Würzburg, Germany
| | - Katharina-Maria Kling
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Greta Therese Franke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Viktoria Herbst
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Dirk von Lewinski
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Daniel Scherr
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078 Würzburg, Germany
| | - Alexander Nickel
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078 Würzburg, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078 Würzburg, Germany
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
129
|
Shi J, Yu X, Liu Z. A review of new-onset ventricular arrhythmia after left ventricular assist device implantation. Cardiology 2022; 147:315-327. [PMID: 35483328 PMCID: PMC9393833 DOI: 10.1159/000524779] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/24/2022] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Heart failure (HF) is a severe and terminal stage of various heart diseases. Left ventricular assist devices (LVADs) are relatively mature and have contributed to the treatment of end-stage HF. Ventricular arrhythmia (VA) is a common complication after LVAD implantation, including ventricular tachycardia and ventricular fibrillation, both of which may cause abnormal circulation in the body. METHODS A literature search was conducted in the PubMed database, "Ventricular Arrhythmia" OR "VA" OR "Arrhythmia" OR "Ventricular Tachycardia," OR "Ventricular Fibrillation" AND "LVAD" OR "Left Ventricular Assist Device" OR "Heart Assist Device" as either keywords or MeSH terms, the authors screened the titles and abstracts of the articles. Eventually, 12 original research articles were retrieved. RESULTS The 0.83 [95%CI: 0.77, 0.89] of patients were male. A whole of 53% [95%CI:0.25, 0.81] of VA patients had a history of atrial fibrillation and 61% [95%CI:0.52, 0.69] had a history of VA. 39% [95%CI:0.29, 0.49] of the participants had no prior history of VA and experienced new VA following CF-LVAD implantation. Following CF-LVAD implantation, 59% [95%CI:0.51, 0.67] of patients developed the early VA (VA ≤ 30 days). The 30-day mortality of patients was 4% [95%CI:0.01, 0.07]. And overall mortality was 28% [95%CI:0.15, 0.41] The reported incidence of VA after LVAD implantation is not identical in different medical centers and ranges from 20% to 60%. The mechanism of VA after LVAD implantation is summarized as primary cardiomyopathy-related, device mechanical stimulation, myocardial scarring, ventricular displacement, electrolyte regulation, and other processes. CONCLUSIONS A preoperative VA history is considered a predictor of VA following LVAD implantation in most studies. Multiple mechanisms and factors, such as prevention of "suction events", ablation, and ICD, should be considered for the prevention and treatment of postoperative VA in patients requiring long-term VAD treatment. This study provides a reference for the clinical application of LAVD and the prevention of postoperative VA after LVAD implantation. Future multicenter prospective studies with uniform patient follow-up are needed to screen for additional potential risk factors and predictors. These studies will help to define the incidence rate of VA after LAVD implantation. As a result, we provide guidance for the selection of preventive intervention.
Collapse
|
130
|
Chen CJ, Jiang C, Yuan J, Chen M, Cuyler J, Xie XQ, Feng Z. How Do Modulators Affect the Orthosteric and Allosteric Binding Pockets? ACS Chem Neurosci 2022; 13:959-977. [PMID: 35298129 PMCID: PMC10496248 DOI: 10.1021/acschemneuro.1c00749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Allosteric modulators (AMs) that bind allosteric sites can exhibit greater selectivity than the orthosteric ligands and can either enhance agonist-induced receptor activity (termed positive allosteric modulator or PAM), inhibit agonist-induced activity (negative AM or NAM), or have no effect on activity (silent AM or SAM). Until now, it is not clear what the exact effects of AMs are on the orthosteric active site or the allosteric binding pocket(s). In the present work, we collected both the three-dimensional (3D) structures of receptor-orthosteric ligand and receptor-orthosteric ligand-AM complexes of a specific target protein. Using our novel algorithm toolset, molecular complex characterizing system (MCCS), we were able to quantify the key residues in both the orthosteric and allosteric binding sites along with potential changes of the binding pockets. After analyzing 21 pairs of 3D crystal or cryo-electron microscopy (cryo-EM) complexes, including 4 pairs of GPCRs, 5 pairs of ion channels, 11 pairs of enzymes, and 1 pair of transcription factors, we found that the binding of AMs had little impact on both the orthosteric and allosteric binding pockets. In return, given the accurately predicted allosteric binding pocket(s) of a drug target of medicinal interest, we can confidently conduct the virtual screening or lead optimization without concern that the huge conformational change of the pocket could lead to the low accuracy of virtual screening.
Collapse
Affiliation(s)
- Chih-Jung Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Chen Jiang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Jiayi Yuan
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Maozi Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Jacob Cuyler
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
131
|
González-Juanatey J, Anguita-Sánchez M, Bayes-Genís A, Comín-Colet J, García-Quintana A, Recio-Mayoral A, Zamorano-Gómez J, Cepeda-Rodrigo J, Manzano L. Vericiguat in heart failure: From scientific evidence to clinical practice. Rev Clin Esp 2022; 222:359-369. [DOI: 10.1016/j.rceng.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022]
|
132
|
Pharmacological assessment of zebrafish-based cardiotoxicity models. Biomed Pharmacother 2022; 148:112695. [DOI: 10.1016/j.biopha.2022.112695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/14/2022] [Accepted: 02/02/2022] [Indexed: 01/03/2023] Open
|
133
|
Wang H, Guo X, Zheng Y, Yang Y. An automatic approach for heart failure typing based on heart sounds and convolutional recurrent neural networks. Phys Eng Sci Med 2022; 45:475-485. [PMID: 35347667 DOI: 10.1007/s13246-022-01112-8] [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: 09/08/2021] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
Abstract
Heart failure (HF) is a complex clinical syndrome that poses a major hazard to human health. Patients with different types of HF have great differences in pathogenesis and treatment options. Therefore, HF typing is of great significance for timely treatment of patients. In this paper, we proposed an automatic approach for HF typing based on heart sounds (HS) and convolutional recurrent neural networks, which provides a new non-invasive and convenient way for HF typing. Firstly, the collected HS signals were preprocessed with adaptive wavelet denoising. Then, the logistic regression based hidden semi-Markov model was utilized to segment HS frames. For the distinction between normal subjects and the HF patients with preserved ejection fraction or reduced ejection fraction, a model based on convolutional neural network and recurrent neural network was built. The model can automatically learn the spatial and temporal characteristics of HS signals. The results show that the proposed model achieved a superior performance with an accuracy of 97.64%. This study suggests the proposed method could be a useful tool for HF recognition and as a supplement for HF typing.
Collapse
Affiliation(s)
- Hui Wang
- Key Laboratory of Biorheology Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xingming Guo
- Key Laboratory of Biorheology Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Yineng Zheng
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yang Yang
- Key Laboratory of Biorheology Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
134
|
Carnevale D. Neuroimmune axis of cardiovascular control: mechanisms and therapeutic implications. Nat Rev Cardiol 2022; 19:379-394. [PMID: 35301456 DOI: 10.1038/s41569-022-00678-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2022] [Indexed: 12/21/2022]
Abstract
Cardiovascular diseases (CVDs) make a substantial contribution to the global burden of disease. Prevention strategies have succeeded in reducing the effect of acute CVD events and deaths, but the long-term consequences of cardiovascular risk factors still represent the major cause of disability and chronic illness, suggesting that some pathophysiological mechanisms might not be adequately targeted by current therapies. Many of the underlying causes of CVD have now been recognized to have immune and inflammatory components. However, inflammation and immune activation were mostly regarded as a consequence of target-organ damage. Only more recent findings have indicated that immune dysregulation can be pathogenic for CVD, identifying a need for novel immunomodulatory therapeutic strategies. The nervous system, through an array of afferent and efferent arms of the autonomic nervous system, profoundly affects cardiovascular function. Interestingly, the autonomic nervous system also innervates immune organs, and neuroimmune interactions that are biologically relevant to CVD have been discovered, providing the foundation to target neural reflexes as an immunomodulatory therapeutic strategy. This Review summarizes how the neural regulation of immunity and inflammation participates in the onset and progression of CVD and explores promising opportunities for future therapeutic strategies.
Collapse
Affiliation(s)
- Daniela Carnevale
- Department of Molecular Medicine, Sapienza University, Rome, Italy. .,Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy.
| |
Collapse
|
135
|
Guimaraes DA, Aquino NSS, Rocha-Resende C, Jesus ICG, Silva MM, Scalzo SA, Fonseca RC, Durand MT, Pereira V, Tezini GCSV, Oliveira A, Prado VF, Stefanon I, Salgado HC, Prado MAM, Szawka RE, Guatimosim S. Neuronal cholinergic signaling constrains norepinephrine activity in the heart. Am J Physiol Cell Physiol 2022; 322:C794-C801. [PMID: 35264016 DOI: 10.1152/ajpcell.00031.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is well known that cholinergic hypofunction contributes to cardiac pathology; yet, the mechanisms involved remain unclear. Our previous publication has shown that genetically engineered model of cholinergic deficit, the vesicular acetylcholine transporter knockdown homozygous (VAChT KDHOM) mice exhibit pathological cardiac remodeling and a gradual increase in cardiac mass with aging. Given that an increase in cardiac mass is often caused by adrenergic hyperactivity, we hypothesized that VAChT KDHOM mice might have an increase in cardiac norepinephrine (NE) levels. We thus investigated the temporal changes in NE content in the heart from 3, 6 and 12 month-old VAChT mutants. Interestingly, mice with cholinergic hypofunction showed a gradual elevation in cardiac NE content, which was already increased at 6 months of age. Consistent with this finding, 6 month-old VAChT KDHOM mice showed enhanced sympathetic activity and a greater abundance of tyrosine hydroxylase positive sympathetic nerves in the heart. VAChT mutants exhibited an increase in peak calcium transient, and mitochondrial oxidative stress in cardiomyocytes along with enhanced GRK5 and NFAT staining in the heart. These are known targets of adrenergic signaling in the cell. Moreover, vagotomized-mice displayed an increase in cardiac NE content confirming the data obtained in VAChT KDHOM mice. Establishing a causal relationship between acetylcholine and NE, VAChT KDHOM mice treated with pyridostigmine, a cholinesterase inhibitor, showed reduced cardiac NE content, rescuing the phenotype. Our findings unveil a yet unrecognized role of cholinergic signaling as a modulator of cardiac NE, providing novel insights into the mechanisms that drive autonomic imbalance.
Collapse
Affiliation(s)
- Diogo A Guimaraes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nayara S S Aquino
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cibele Rocha-Resende
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Itamar C G Jesus
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mário Morais Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio A Scalzo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Roberta Cristelli Fonseca
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina T Durand
- Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Vanessa Pereira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - André Oliveira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vânia F Prado
- Robarts Research Institute, The University of Western Ontario, Department of Physiology 1and Pharmacology, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Ivanita Stefanon
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, Espirito Santo, Brazil
| | - Helio C Salgado
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marco Antonio Máximo Prado
- Robarts Research Institute, The University of Western Ontario, Department of Physiology 1and Pharmacology, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Raphael E Szawka
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
136
|
Conte M, Petraglia L, Poggio P, Valerio V, Cabaro S, Campana P, Comentale G, Attena E, Russo V, Pilato E, Formisano P, Leosco D, Parisi V. Inflammation and Cardiovascular Diseases in the Elderly: The Role of Epicardial Adipose Tissue. Front Med (Lausanne) 2022; 9:844266. [PMID: 35242789 PMCID: PMC8887867 DOI: 10.3389/fmed.2022.844266] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
Human aging is a complex phenomenon characterized by a wide spectrum of biological changes which impact on behavioral and social aspects. Age-related changes are accompanied by a decline in biological function and increased vulnerability leading to frailty, thereby advanced age is identified among the major risk factors of the main chronic human diseases. Aging is characterized by a state of chronic low-grade inflammation, also referred as inflammaging. It recognizes a multifactorial pathogenesis with a prominent role of the innate immune system activation, resulting in tissue degeneration and contributing to adverse outcomes. It is widely recognized that inflammation plays a central role in the development and progression of numerous chronic and cardiovascular diseases. In particular, low-grade inflammation, through an increased risk of atherosclerosis and insulin resistance, promote cardiovascular diseases in the elderly. Low-grade inflammation is also promoted by visceral adiposity, whose accumulation is paralleled by an increased inflammatory status. Aging is associated to increase in epicardial adipose tissue (EAT), the visceral fat depot of the heart. Structural and functional changes in EAT have been shown to be associated with several heart diseases, including coronary artery disease, aortic stenosis, atrial fibrillation, and heart failure. EAT increase is associated with a greater production and secretion of pro-inflammatory mediators and neuro-hormones, so that thickened EAT can pathologically influence, in a paracrine and vasocrine manner, the structure and function of the heart and is associated to a worse cardiovascular outcome. In this review, we will discuss the evidence underlying the interplay between inflammaging, EAT accumulation and cardiovascular diseases. We will examine and discuss the importance of EAT quantification, its characteristics and changes with age and its clinical implication.
Collapse
Affiliation(s)
- Maddalena Conte
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Casa di Cura San Michele, Maddaloni, Italy
| | - Laura Petraglia
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | | | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Pasquale Campana
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Comentale
- Department of Advanced Biomedical Science, University of Naples Federico II, Naples, Italy
| | - Emilio Attena
- Department of Cardiology, Monaldi Hospital, Naples, Italy
| | - Vincenzo Russo
- Department of Medical Translational Sciences, Monaldi Hospital, University of Campania Luigi Vanvitelli, Campania, Italy
| | - Emanuele Pilato
- Department of Advanced Biomedical Science, University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
137
|
Discovery, synthesis and exploration of N-benzylsulfonyl-2-phenylazepanes as inhibitors of Bim expression in a mouse embryonic fibroblast model. Bioorg Chem 2022; 120:105635. [DOI: 10.1016/j.bioorg.2022.105635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/08/2021] [Accepted: 01/18/2022] [Indexed: 11/17/2022]
|
138
|
Argunhan F, Brain SD. The Vascular-Dependent and -Independent Actions of Calcitonin Gene-Related Peptide in Cardiovascular Disease. Front Physiol 2022; 13:833645. [PMID: 35283798 PMCID: PMC8914086 DOI: 10.3389/fphys.2022.833645] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/21/2022] [Indexed: 12/21/2022] Open
Abstract
The treatment of hypertension and heart failure remains a major challenge to healthcare providers. Despite therapeutic advances, heart failure affects more than 26 million people worldwide and is increasing in prevalence due to an ageing population. Similarly, despite an improvement in blood pressure management, largely due to pharmacological interventions, hypertension remains a silent killer. This is in part due to its ability to contribute to heart failure. Development of novel therapies will likely be at the forefront of future cardiovascular studies to address these unmet needs. Calcitonin gene-related peptide (CGRP) is a 37 amino acid potent vasodilator with positive-ionotropic and -chronotropic effects. It has been reported to have beneficial effects in hypertensive and heart failure patients. Interestingly, changes in plasma CGRP concentration in patients after myocardial infarction, heart failure, and in some forms of hypertension, also support a role for CGRP on hemodynamic functions. Rodent studies have played an important role thus far in delineating mechanisms involved in CGRP-induced cardioprotection. However, due to the short plasma half-life of CGRP, these well documented beneficial effects have often proven to be acute and transient. Recent development of longer lasting CGRP agonists may therefore offer a practical solution to investigating CGRP further in cardiovascular disease in vivo. Furthermore, pre-clinical murine studies have hinted at the prospect of cardioprotective mechanisms of CGRP which is independent of its hypotensive effect. Here, we discuss past and present evidence of vascular-dependent and -independent processes by which CGRP could protect the vasculature and myocardium against cardiovascular dysfunction.
Collapse
|
139
|
Rossi A, Mikail N, Bengs S, Haider A, Treyer V, Buechel RR, Wegener S, Rauen K, Tawakol A, Bairey Merz CN, Regitz-Zagrosek V, Gebhard C. Heart-brain interactions in cardiac and brain diseases: why sex matters. Eur Heart J 2022; 43:3971-3980. [PMID: 35194633 PMCID: PMC9794190 DOI: 10.1093/eurheartj/ehac061] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease and brain disorders, such as depression and cognitive dysfunction, are highly prevalent conditions and are among the leading causes limiting patient's quality of life. A growing body of evidence has shown an intimate crosstalk between the heart and the brain, resulting from a complex network of several physiological and neurohumoral circuits. From a pathophysiological perspective, both organs share common risk factors, such as hypertension, diabetes, smoking or dyslipidaemia, and are similarly affected by systemic inflammation, atherosclerosis, and dysfunction of the neuroendocrine system. In addition, there is an increasing awareness that physiological interactions between the two organs play important roles in potentiating disease and that sex- and gender-related differences modify those interactions between the heart and the brain over the entire lifespan. The present review summarizes contemporary evidence of the effect of sex on heart-brain interactions and how these influence pathogenesis, clinical manifestation, and treatment responses of specific heart and brain diseases.
Collapse
Affiliation(s)
- Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland,Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland,Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland,Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Ahmed Haider
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland,Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland,Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Ronny Ralf Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Susanne Wegener
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Katrin Rauen
- Department of Geriatric Psychiatry, Psychiatric Hospital, Zurich, Switzerland,Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center, Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin Berlin, Berlin, Germany,University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
140
|
Pitoulis FG, Nunez-Toldra R, Xiao K, Kit-Anan W, Mitzka S, Jabbour RJ, Harding SE, Perbellini F, Thum T, de Tombe PP, Terracciano CM. Remodelling of adult cardiac tissue subjected to physiological and pathological mechanical load in vitro. Cardiovasc Res 2022; 118:814-827. [PMID: 33723566 PMCID: PMC8859636 DOI: 10.1093/cvr/cvab084] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/11/2021] [Indexed: 01/14/2023] Open
Abstract
AIMS Cardiac remodelling is the process by which the heart adapts to its environment. Mechanical load is a major driver of remodelling. Cardiac tissue culture has been frequently employed for in vitro studies of load-induced remodelling; however, current in vitro protocols (e.g. cyclic stretch, isometric load, and auxotonic load) are oversimplified and do not accurately capture the dynamic sequence of mechanical conformational changes experienced by the heart in vivo. This limits translational scope and relevance of findings. METHODS AND RESULTS We developed a novel methodology to study chronic load in vitro. We first developed a bioreactor that can recreate the electromechanical events of in vivo pressure-volume loops as in vitro force-length loops. We then used the bioreactor to culture rat living myocardial slices (LMS) for 3 days. The bioreactor operated based on a 3-Element Windkessel circulatory model enabling tissue mechanical loading based on physiologically relevant parameters of afterload and preload. LMS were continuously stretched/relaxed during culture simulating conditions of physiological load (normal preload and afterload), pressure-overload (normal preload and high afterload), or volume-overload (high preload & normal afterload). At the end of culture, functional, structural, and molecular assays were performed to determine load-induced remodelling. Both pressure- and volume-overloaded LMS showed significantly decreased contractility that was more pronounced in the latter compared with physiological load (P < 0.0001). Overloaded groups also showed cardiomyocyte hypertrophy; RNAseq identified shared and unique genes expressed in each overload group. The PI3K-Akt pathway was dysregulated in volume-overload while inflammatory pathways were mostly associated with remodelling in pressure-overloaded LMS. CONCLUSION We have developed a proof-of-concept platform and methodology to recreate remodelling under pathophysiological load in vitro. We show that LMS cultured in our bioreactor remodel as a function of the type of mechanical load applied to them.
Collapse
Affiliation(s)
- Fotios G Pitoulis
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
| | - Raquel Nunez-Toldra
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
| | - Ke Xiao
- Institute for Molecular and Translational Therapeutic Strategies, Hannover Medical School, OE 8886, Carl-Neuberg-Str. 1, J3 Building, Level 1, Room 3030, 30625 Hannover, Germany
| | - Worrapong Kit-Anan
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
| | - Saskia Mitzka
- Institute for Molecular and Translational Therapeutic Strategies, Hannover Medical School, OE 8886, Carl-Neuberg-Str. 1, J3 Building, Level 1, Room 3030, 30625 Hannover, Germany
| | - Richard J Jabbour
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
| | - Filippo Perbellini
- Institute for Molecular and Translational Therapeutic Strategies, Hannover Medical School, OE 8886, Carl-Neuberg-Str. 1, J3 Building, Level 1, Room 3030, 30625 Hannover, Germany
| | - Thomas Thum
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
- Institute for Molecular and Translational Therapeutic Strategies, Hannover Medical School, OE 8886, Carl-Neuberg-Str. 1, J3 Building, Level 1, Room 3030, 30625 Hannover, Germany
| | - Pieter P de Tombe
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Rm E202 (MC901), Chicago, IL 60612-7342, USA
| | - Cesare M Terracciano
- National Heart and Lung Institute, Imperial College London, 72 Du Cane Road, Hammersmith Hospital, ICTEM Building, W12 0NN London, UK
| |
Collapse
|
141
|
Chen X, Zhang Y, Li L, Li S, Zhang J. Treadmill running alleviates adipose tissue browning and lipolysis in rats with heart failure. J Physiol Biochem 2022; 78:323-334. [PMID: 35112327 DOI: 10.1007/s13105-021-00849-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/18/2021] [Indexed: 11/29/2022]
Abstract
This study observed the effects of treadmill running on adipose tissue browning and lipolysis in rats with induced heart failure and elucidated the possible mechanism. Rats underwent abdominal aortic constriction as a model of heart failure. Cardiac function was detected by echocardiography. We detected serum levels of norepinephrine and interleukin 6, cardiac atrial natriuretic peptide and brain natriuretic peptide and marker genes of browning, white adipose tissue (WAT), and lipolysis in adipose tissue. Rats with heart failure showed typical symptoms such as increased heart weight and mRNA levels of atrial natriuretic peptide and brain natriuretic peptide and decreased left ventricular ejection fraction. Exercise partially improved left ventricular diastolic function and significantly decreased atrial natriuretic peptide expression. Rats with heart failure showed significantly reduced body weight and ratios of muscle and fat weight to body weight. Exercise significantly increased body weight and the ratio of muscle weight to body weight. Heart failure stimulated the expression of proliferator-activated receptor-gamma coactivator-1-alpha and uncoupling protein 1 in epididymal WAT, inguinal WAT, and brown adipose tissue but decreased that of adiponectin and leptin in inguinal WAT. Lipolysis, characterized by high adipose triglyceride lipase and hormone-sensitive lipase expression, was activated in all adipose tissues. Exercise reduced browning and lipolysis in adipose tissues. Rats with heart failure had abnormally high levels of serum norepinephrine and interleukin 6, which could be suppressed by exercise. Exercise may improve cardiac cachexia and inhibit the browning and lipolysis of adipose tissue by downregulating sympathetic nervous system activity and inflammation.
Collapse
Affiliation(s)
- Xuefei Chen
- School of P.E. and Sports Science, Beijing Normal University, Beijing, 100875, China
| | - Yuhan Zhang
- School of P.E. and Sports Science, Beijing Normal University, Beijing, 100875, China
| | - Lingjie Li
- School of P.E. and Sports Science, Beijing Normal University, Beijing, 100875, China
| | - Shitian Li
- School of P.E. and Sports Science, Beijing Normal University, Beijing, 100875, China
| | - Jing Zhang
- School of P.E. and Sports Science, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
142
|
Saghafi F, Bagheri N, Salehi-Abargouei A, Sahebnasagh A. Efficacy of combination triple therapy with vasopressin, steroid, and epinephrine in cardiac arrest: a systematic review and meta-analysis of randomized-controlled trials. J Intensive Care 2022; 10:5. [PMID: 35109925 PMCID: PMC8809021 DOI: 10.1186/s40560-022-00597-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background This study investigated whether combination therapy with vasopressin, steroid, and epinephrine (VSE) improves in-hospital survival and return of spontaneous circulation (ROSC) during and after resuscitation in-hospital cardiac arrest (CA). Materials and methods Various databases were explored from inception until October 2021 for relevant published clinical trials and cohort studies. Results Three clinical trials were included. Pooled analysis suggested that VSE was significantly associated with increased ROSC in patients with in-hospital CA (IHCA) (odds ratio (OR): 2.281, 95% confidence interval (CI): 1.304–3.989, P value = 0.004). Meta-analysis of two studies (368 patients) demonstrated a significant difference in the reduction of mean arterial pressure (MAP) during and 15–20 min after cardiopulmonary resuscitation (standardized mean difference (SMD): 1.069, 95% CI: 0.851–1.288, P value < 0.001), renal failure free days (SMD = 0.590; 95% CI: 0.312–0.869 days; P value < 0.001), and coagulation failure free days (SMD = 0.403; 95% CI: 0.128–0.679, P value = 0.004). However, no significant difference was observed for survival-to-discharge ratio (OR: 2.082, 95% CI: 0.638–6.796, P value = 0.225) and ventilator free days (SMD = 0.201, 95% CI: − 0.677, 1.079 days; P value = 0.838). Conclusions VSE combination therapy during and after IHCA may have beneficial effects in terms of the ROSC, renal and circulatory failure free days, and MAP. Prospero registration: CRD42020178297 (05/07/2020). Supplementary Information The online version contains supplementary material available at 10.1186/s40560-022-00597-5.
Collapse
Affiliation(s)
- Fatemeh Saghafi
- Department of Clinical Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Negar Bagheri
- Pharmaceutical Sciences Research Center, School of Pharmacy, Student Research Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amin Salehi-Abargouei
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
143
|
Genetic polymorphisms in ADRB2 and ADRB1 are associated with differential survival in heart failure patients taking β-blockers. THE PHARMACOGENOMICS JOURNAL 2022; 22:62-68. [PMID: 34642472 PMCID: PMC8799480 DOI: 10.1038/s41397-021-00257-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023]
Abstract
Single nucleotide polymorphisms (SNPs) have been associated with differential beta-blocker (BB) effects on heart rate, blood pressure, and left ventricular ejection fraction in various patient populations. This study aimed to determine if SNPs previously associated with BB response are also associated with differential survival in heart failure (HF) patients receiving BBs. HF patient data were derived from electronic health records and the Social Security Death Index. Associations and interactions between BB dose, SNP genotype, and the outcome of death were assessed using a Cox proportional-hazard model adjusting for covariates known to be associated with differential survival in HF patients. Two SNPs, ADRB1 Arg389Gly and ADRB2 Glu27Gln, displayed significant interactions (Pint = 0.043 and Pint = 0.017, respectively) with BB dose and their association with mortality. Our study suggests that ADRB2 27Glu and ADRB1 389Arg may confer a larger survival benefit with higher BB doses in patients with HF.
Collapse
|
144
|
Hiraiwa H, Okumura T, Sawamura A, Araki T, Mizutani T, Kazama S, Kimura Y, Shibata N, Oishi H, Kuwayama T, Kondo T, Furusawa K, Morimoto R, Murohara T. Splenic size as an indicator of hemodynamics and prognosis in patients with heart failure. Heart Vessels 2022; 37:1344-1355. [DOI: 10.1007/s00380-022-02030-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/14/2022] [Indexed: 01/16/2023]
|
145
|
Allen LA, Teerlink JR, Gottlieb SS, Ahmad T, Lam CSP, Psotka MA. Heart Failure Spending Function: An Investment Framework for Sequencing and Intensification of Guideline-Directed Medical Therapies. Circ Heart Fail 2022; 15:e008594. [PMID: 35000432 DOI: 10.1161/circheartfailure.121.008594] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Heart failure with reduced ejection fraction is managed with increasing numbers of guideline-directed medical therapies (GDMT). Benefits tend to be additive. Burdens can also be additive. We propose a heart failure spending function as a conceptual framework for tailored intensification of GDMT that maximizes therapeutic opportunity while limiting adverse events and patient burden. Each patient is conceptualized to have reserve in physiological and psychosocial domains, which can be spent for a future return on investment. Key domains are blood pressure, heart rate, serum creatinine, potassium, and out-of-pocket costs. For each patient, GDMT should be initiated and intensified in a sequence that prioritizes medications with the greatest expected cardiac benefit while drawing on areas where the patient has ample reserves. When reserve is underspent, patients fail to gain the full benefit of GDMT. Conversely, when a reserve is fully spent, addition of new drugs or higher doses that draw upon a domain will lead to patient harm. The benefit of multiple agents drawing upon varied physiological domains should be balanced against cost and complexity. Thresholds for overspending are explored, as are mechanisms for implementing these concepts into routine care, but further health care delivery research is needed to validate and refine clinical use of the spending function. The heart failure spending function also suggests how newer therapies may be considered in terms of relative value, prioritizing agents that draw on different spending domains from existing GDMT.
Collapse
Affiliation(s)
- Larry A Allen
- Division of Cardiology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora (L.A.A.)
| | - John R Teerlink
- Section of Cardiology, San Francisco Veterans Affairs Medical Center and Department of Medicine, School of Medicine, University of California San Francisco (J.R.T.)
| | | | - Tariq Ahmad
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT (T.A.)
| | - Carolyn S P Lam
- National Heart Centre Singapore and Duke-National University of Singapore (C.S.P.L.)
| | | |
Collapse
|
146
|
Thorp EB, Flanagan ME, Popko B, DeBerge M. Resolving inflammatory links between myocardial infarction and vascular dementia. Semin Immunol 2022; 59:101600. [PMID: 35227567 PMCID: PMC10234261 DOI: 10.1016/j.smim.2022.101600] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 01/15/2023]
Abstract
Myocardial infarction is associated with increased risk for vascular dementia. In both myocardial infarction and vascular dementia, there is evidence that elevated inflammatory biomarkers are associated with worsened clinical outcomes. Myocardial infarction leads to a systemic inflammatory response, which may contribute to recruitment or activation of myeloid cells, including monocytes, microglia, and perivascular macrophages, within the central nervous system. However, our understanding of the causative roles for these cells linking cardiac injury to the development and progression of dementia is incomplete. Herein, we provide an overview of inflammatory cellular and molecular links between myocardial infarction and vascular dementia and discuss strategies to resolve inflammation after myocardial infarction to limit neurovascular injury.
Collapse
Affiliation(s)
- Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States.
| | - Margaret E Flanagan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Brian Popko
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Matthew DeBerge
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States.
| |
Collapse
|
147
|
OUP accepted manuscript. Eur J Prev Cardiol 2022; 29:1124-1141. [DOI: 10.1093/eurjpc/zwac006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 11/12/2022]
|
148
|
Nathaniel S, McGinty S, Witman MA, Edwards DG, Farquhar WB, Hosmane V, Wenner MM. A new lead: Sacubitril-valsartan's unique benefit in HFrEF could lie with sympathoinhibition. Auton Neurosci 2022; 238:102949. [DOI: 10.1016/j.autneu.2022.102949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
|
149
|
Schwarz KG, Pereyra KV, Toledo C, Andrade DC, Díaz HS, Díaz-Jara E, Ortolani D, Rios-Gallardo A, Arias P, Las Heras A, Vera I, Ortiz FC, Inestrosa NC, Vio CP, Del Rio R. Effects of enriched-potassium diet on cardiorespiratory outcomes in experimental non-ischemic chronic heart failure. Biol Res 2021; 54:43. [PMID: 34952651 PMCID: PMC8710008 DOI: 10.1186/s40659-021-00365-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background Chronic heart failure (CHF) is a global health problem. Increased sympathetic outflow, cardiac arrhythmogenesis and irregular breathing patterns have all been associated with poor outcomes in CHF. Several studies showed that activation of the renin-angiotensin system (RAS) play a key role in CHF pathophysiology. Interestingly, potassium (K+) supplemented diets showed promising results in normalizing RAS axis and autonomic dysfunction in vascular diseases, lowering cardiovascular risk. Whether subtle increases in dietary K+ consumption may exert similar effects in CHF has not been previously tested. Accordingly, we aimed to evaluate the effects of dietary K+ supplementation on cardiorespiratory alterations in rats with CHF. Methods Adult male Sprague–Dawley rats underwent volume overload to induce non-ischemic CHF. Animals were randomly allocated to normal chow diet (CHF group) or supplemented K+ diet (CHF+K+ group) for 6 weeks. Cardiac arrhythmogenesis, sympathetic outflow, baroreflex sensitivity, breathing disorders, chemoreflex function, respiratory–cardiovascular coupling and cardiac function were evaluated. Results Compared to normal chow diet, K+ supplemented diet in CHF significantly reduced arrhythmia incidence (67.8 ± 15.1 vs. 31.0 ± 3.7 events/hour, CHF vs. CHF+K+), decreased cardiac sympathetic tone (ΔHR to propranolol: − 97.4 ± 9.4 vs. − 60.8 ± 8.3 bpm, CHF vs. CHF+K+), restored baroreflex function and attenuated irregular breathing patterns. Additionally, supplementation of the diet with K+ restores normal central respiratory chemoreflex drive and abrogates pathological cardio-respiratory coupling in CHF rats being the outcome an improved cardiac function. Conclusion Our findings support that dietary K+ supplementation in non-ischemic CHF alleviate cardiorespiratory dysfunction. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00365-z.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Fisiología y Medicina de Altura, Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Rios-Gallardo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Paulina Arias
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexandra Las Heras
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Vera
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos P Vio
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
150
|
Abstract
Motivated by the persistence of uncontrolled blood pressure and its public health impact, the development and evaluation of device-based therapies for hypertension has advanced at an accelerated pace to complement pharmaceutical and lifestyle intervention strategies. Countering widespread interest from early studies, the lack of demonstrable efficacy for renal denervation (RDN) in a large, sham-controlled randomized trial motivated revision of trial design and conduct to account for confounding variables of procedural technique, medication variability, and selection of both patients and end points. Now amidst varied trial design and methods, several sham-controlled, randomized trials have demonstrated clinically meaningful reductions in blood pressure with RDN. With this momentum, additional studies are underway to position RDN as a potential part of standard therapy for the world's leading cause of death and disability. In parallel, further studies will address unresolved issues including durability of blood pressure lowering and reduction in antihypertensive medications, late-term safety, and impact on clinical outcomes. Identifying predictors of treatment effect and surveys of patient-reported outcomes and treatment preferences are also evolving areas of investigation. Aside from confirmatory studies of safety and effectiveness, these additional studies will further inform patient selection, expand experience with RDN in broader populations with hypertension, and provide guidance to how RDN may be incorporated into treatment pathways.
Collapse
|