51
|
Belevych AE, Bogdanov V, Terentyev DA, Gyorke S. Acute Detubulation of Ventricular Myocytes Amplifies the Inhibitory Effect of Cholinergic Agonist on Intracellular Ca 2+ Transients. Front Physiol 2021; 12:725798. [PMID: 34512394 PMCID: PMC8427700 DOI: 10.3389/fphys.2021.725798] [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: 06/15/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022] Open
Abstract
Muscarinic receptors expressed in cardiac myocytes play a critical role in the regulation of heart function by the parasympathetic nervous system. How the structural organization of cardiac myocytes affects the regulation of Ca2+ handling by muscarinic receptors is not well-defined. Using confocal Ca2+ imaging, patch-clamp techniques, and immunocytochemistry, the relationship between t-tubule density and cholinergic regulation of intracellular Ca2+ in normal murine ventricular myocytes and myocytes with acute disruption of the t-tubule system caused by formamide treatment was studied. The inhibitory effect of muscarinic receptor agonist carbachol (CCh, 10 μM) on the amplitude of Ca2+ transients, evoked by field-stimulation in the presence of 100 nM isoproterenol (Iso), a β-adrenergic agonist, was directly proportional to the level of myocyte detubulation. The timing of the maximal rate of fluorescence increase of fluo-4, a Ca2+-sensitive dye, was used to classify image pixels into the regions functionally coupled or uncoupled to the sarcolemmal Ca2+ influx (ICa). CCh decreased the fraction of coupled regions and suppressed Ca2+ propagation from sarcolemma inside the cell. Formamide treatment reduced ICa density and decreased sarcoplasmic reticulum (SR) Ca2+ content. CCh did not change SR Ca2+ content in Iso-stimulated control and formamide-treated myocytes. CCh inhibited peak ICa recorded in the presence of Iso by ∼20% in both the control and detubulated myocytes. Reducing ICa amplitude up to 40% by changing the voltage step levels from 0 to –25 mV decreased Ca2+ transients in formamide-treated but not in control myocytes in the presence of Iso. CCh inhibited CaMKII activity, whereas CaMKII inhibition with KN93 mimicked the effect of CCh on Ca2+ transients in formamide-treated myocytes. It was concluded that the downregulation of t-tubules coupled with the diminished efficiency of excitation–contraction coupling, increases the sensitivity of Ca2+ release and propagation to muscarinic receptor-mediated inhibition of both ICa and CaMKII activity.
Collapse
Affiliation(s)
- Andriy E Belevych
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Dmitry A Terentyev
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sandor Gyorke
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
52
|
Ang L, Kidwell KM, Dillon B, Reiss J, Fang F, Leone V, Mizokami-Stout K, Pop-Busui R. Dapagliflozin and measures of cardiovascular autonomic function in patients with type 2 diabetes (T2D). J Diabetes Complications 2021; 35:107949. [PMID: 34024686 DOI: 10.1016/j.jdiacomp.2021.107949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 01/08/2023]
Abstract
AIMS Sodium-glucose cotransporter-2 (SGLT-2) inhibitors reduce blood pressure without compensatory heart rate elevation, possibly by modulating sympathetic/parasympathetic activity. This may contribute to their cardiovascular benefits in type 2 diabetes (T2D). We evaluated the effects of dapagliflozin (DAPA) on measures of cardiovascular autonomic neuropathy (CAN), cardiac function, and glucose variability (GV) in T2D. METHODS Pilot, randomized, two-period crossover trial comparing 12-week DAPA versus 12-week glimepiride treatment on CAN measures (cardiovascular autonomic reflex tests and heart rate variability), B-type natriuretic peptide (BNP), and GV (Abbott's Libre Pro devices) using signed rank tests and mixed models from baseline to 12 weeks within and between each period. RESULTS Forty-five T2D participants on metformin monotherapy (mean age 57 ± 8 years, duration 7 ± 6 years, HbA1c 7.8 ± 1.3%) were enrolled with 41 completing the trial. There were no differences in CAN indices or BNP with each drug compared to baseline and each other. Participants on DAPA demonstrated greater weight loss, reduced time in hypoglycemia, and improved GV compared to glimepiride. CONCLUSIONS Short term treatment with DAPA did not affect CAN measures or BNP in uncomplicated and relatively healthy T2D participants. Longer prospective studies in patients with advanced disease are needed to better understand relationships between SGLT-2 inhibitors and CAN. CLINICAL TRIAL REGISTRATION NCT02973477.
Collapse
Affiliation(s)
- Lynn Ang
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America.
| | - Kelley M Kidwell
- School of Public Health, Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States of America
| | - Brendan Dillon
- University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Jacob Reiss
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America
| | - Fang Fang
- School of Public Health, Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States of America
| | - Virginia Leone
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America
| | - Kara Mizokami-Stout
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America; Ann Arbor Veteran Affairs Hospital, Ann Arbor, MI, United States of America
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
53
|
Abstract
Cancer and cardiovascular diseases, including heart failure (HF), are the main causes of death in Western countries. Several anticancer drugs and radiotherapy have adverse effects on the cardiovascular system, promoting left ventricular dysfunction and ultimately HF. Nonetheless, the relationship between cancer and HF is likely not unidirectional. Indeed, cancer and HF share common risk factors, and both have a bidirectional relationship with systemic inflammation, metabolic disturbances, and neurohormonal and immune activation. Few studies have assessed the impact of untreated cancer on the heart. The presence of an active cancer has been associated with elevated cardiac biomarkers, an initial impairment of left ventricular structure and function, autonomic dysfunction, and reduced exercise tolerance. In turn, these conditions might increase the risk of cardiac damage from chemotherapy and radiotherapy. HF drugs such as beta-blockers or inhibitors of the renin–angiotensin–aldosterone system might exert a protective effect on the heart even before the start of cancer therapies. In this review, we recapitulate the evidence of cardiac involvement in cancer patients naïve from chemotherapy and radiotherapy and no history of cardiac disease. We also focus on the perspectives for an early diagnosis and treatment to prevent the progression to cardiac dysfunction and clinical HF, and the potential benefits of cardioactive drugs on cancer progression.
Collapse
|
54
|
Floras JS. The 2021 Carl Ludwig Lecture. Unsympathetic autonomic regulation in heart failure: patient-inspired insights. Am J Physiol Regul Integr Comp Physiol 2021; 321:R338-R351. [PMID: 34259047 DOI: 10.1152/ajpregu.00143.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Defined as a structural or functional cardiac abnormality accompanied by symptoms, signs, or biomarkers of altered ventricular pressures or volumes, heart failure also is a state of autonomic disequilibrium. A large body of evidence affirms that autonomic disturbances are intrinsic to heart failure; basal or stimulated sympathetic nerve firing or neural norepinephrine (NE) release more often than not exceed homeostatic need, such that an initially adaptive adrenergic or vagal reflex response becomes maladaptive. The magnitude of such maladaptation predicts prognosis. This Ludwig lecture develops two theses: the elucidation and judiciously targeted amelioration of maladaptive autonomic disturbances offers opportunities to complement contemporary guideline-based heart failure therapy, and serendipitous single-participant insights, acquired in the course of experimental protocols with entirely different intent, can generate novel insight, inform mechanisms, and launch entirely new research directions. I précis six elements of our current synthesis of the causes and consequences of maladaptive sympathetic disequilibrium in heart failure, shaped by patient-inspired epiphanies: arterial baroreceptor reflex modulation, excitation stimulated by increased cardiac filling pressure, paradoxical muscle sympathetic activation as a peripheral neurogenic constraint on exercise capacity, renal sympathetic restraint of natriuresis, coexisting sleep apnea, and augmented chemoreceptor reflex sensitivity and then conclude by envisaging translational therapeutic opportunities.
Collapse
Affiliation(s)
- John S Floras
- University Health Network and Sinai Health Division of Cardiology, Toronto General Hospital Research Institute and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
55
|
Grassi G, Mancia G, Esler M. CENTRAL AND PERIPHERAL SYMPATHETIC ACTIVATION IN HEART FAILURE. Cardiovasc Res 2021; 118:1857-1871. [PMID: 34240147 DOI: 10.1093/cvr/cvab222] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/02/2021] [Indexed: 11/12/2022] Open
Abstract
The sympathetic nervous system overdrive occurring in heart failure has been reported since more than half a century. Refinements in the methodological approaches to assess human sympathetic neural function have allowed during recent years to better define various aspects related to the neuroadrenergic alteration. These include 1) the different participation of the individual regional sympathetic cardiovascular districts at the process, 2) the role of the central nervous system in determining the neuroadrenergic overdrive, 3) the involvement of baroreflex, cardiopulmonary reflex and chemoreflex mechanisms in the phoenomenon, which is also closely linked to inflammation and the immune reaction, 4) the relationships with the severity of the disease, its ischaemic or idiopathic nature and the preserved or reduced left ventricular ejection fraction and 5) the adverse functional and structural impact of the sympathetic activation on cardiovascular organs, such as the brain, the heart and the kidneys. Information have been also gained on the active role exerted by the sympathetic activation on the disease outcome and its potential relevance as target of the therapeutic interventions based on non-pharmacological, pharmacological and invasive approaches, including the renal denervation, the splanchnic sympathetic nerve ablation and the carotid baroreflex stimulation. The still undefined aspects of the neurogenic alterations and the unmet goals of the therapeutic approach having the sympathetic activation as a target of the intervention will be finally mentioned.
Collapse
Affiliation(s)
- Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca
| | - Giuseppe Mancia
- Policlinico di Monza and University Milano-Bicocca, Milan, Italy
| | - Murray Esler
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
56
|
Bunsawat K, Ratchford SM, Alpenglow JK, Stehlik J, Smith AS, Richardson RS, Wray DW. Sympathoinhibitory effect of sacubitril-valsartan in heart failure with reduced ejection fraction: A pilot study. Auton Neurosci 2021; 235:102834. [PMID: 34186274 DOI: 10.1016/j.autneu.2021.102834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/07/2021] [Accepted: 06/13/2021] [Indexed: 01/08/2023]
Abstract
Chronic sympathetic nervous system (SNS) overactivity, characteristic of heart failure (HF) with reduced ejection fraction (HFrEF), is associated with poor prognosis and contributes to increased mortality risk. Sacubitril-valsartan is a recently approved, first-in-class, angiotensin receptor neprilysin inhibitor (ARNI) drug that markedly reduces the risks of death from cardiovascular causes and hospitalization for HF in patients with HFrEF, but the physiological mechanisms underlying these benefits are not fully understood. This single-arm, open-label, prospective study sought to test the hypothesis that short-term treatment with sacubitril-valsartan reduces SNS activity, measured directly via muscle sympathetic nerve activity (MSNA), in patients with HFrEF. MSNA, heart rate (HR), and arterial blood pressure (BP) were assessed in stable Class II and III patients with HFrEF (n = 9, 69 ± 8 yrs.; 28.6 ± 3.6 kg/m2) on contemporary, guideline-directed medical treatment who were subsequently started on sacubitril-valsartan. These measurements were repeated after two months of treatment with sacubitril-valsartan. Sacubitril-valsartan reduced MSNA burst frequency (baseline: 43 ± 10 bursts/min; 2-month: 36 ± 10 bursts/min, p = 0.05) and burst incidence (baseline: 68 ± 16 bursts/100 heartbeats; 2-month: 55 ± 16 bursts/100 heartbeats, p = 0.02), while HR and BP were unchanged following of treatment (p > 0.05). These preliminary findings provide new evidence regarding the ability of sacubitril-valsartan to rapidly reduce SNS activity in patients with HFrEF, suggesting the presence of a novel sympathoinhibitory effect of this new drug class.
Collapse
Affiliation(s)
- Kanokwan Bunsawat
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, United States of America.
| | - Stephen M Ratchford
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, United States of America; Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America; Department of Health and Exercise Science, Appalachian State University, Boone, NC, United States of America
| | - Jeremy K Alpenglow
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States of America
| | - Josef Stehlik
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, United States of America
| | - Adam S Smith
- Department of Pharmacy Services, University of Utah, Salt Lake City, UT, United States of America
| | - Russell S Richardson
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, United States of America; Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States of America
| | - D Walter Wray
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, United States of America; Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
57
|
Martin TP, MacDonald EA, Elbassioni AAM, O'Toole D, Zaeri AAI, Nicklin SA, Gray GA, Loughrey CM. Preclinical models of myocardial infarction: from mechanism to translation. Br J Pharmacol 2021; 179:770-791. [PMID: 34131903 DOI: 10.1111/bph.15595] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 11/28/2022] Open
Abstract
Approximately 7 million people are affected by acute myocardial infarction (MI) each year, and despite significant therapeutic and diagnostic advancements, MI remains a leading cause of mortality worldwide. Preclinical animal models have significantly advanced our understanding of MI and have enabled the development of therapeutic strategies to combat this debilitating disease. Notably, some drugs currently used to treat MI and heart failure (HF) in patients had initially been studied in preclinical animal models. Despite this, preclinical models are limited in their ability to fully reproduce the complexity of MI in humans. The preclinical model must be carefully selected to maximise the translational potential of experimental findings. This review describes current experimental models of MI and considers how they have been used to understand drug mechanisms of action and support translational medicine development.
Collapse
Affiliation(s)
- Tamara P Martin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - Eilidh A MacDonald
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - Ali Ali Mohamed Elbassioni
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK.,Suez Canal University, Arab Republic of Egypt
| | - Dylan O'Toole
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - Ali Abdullah I Zaeri
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - Stuart A Nicklin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - Gillian A Gray
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Christopher M Loughrey
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
58
|
Abstract
Despite multiple attempts to develop a unifying hypothesis that explains the pathophysiology of heart failure with a reduced ejection fraction (HFrEF), no single conceptual model has withstood the test of time. In the present review, we discuss how the results of recent successful phase III clinical development programs in HFrEF are built upon existing conceptual models for drug development. We will also discuss where recent successes in clinical trials do not fit existing models to identify areas where further refinement of current paradigms may be needed. To provide the necessary structure for this review, we will begin with a brief overview of the pathophysiology of HFrEF, followed by an overview of the current conceptual models for HFrEF, and end with an analysis of the scientific rationale and clinical development programs for 4 new therapeutic classes of drugs that have improved clinical outcomes in HFrEF. The 4 new therapeutic classes discussed are ARNIs, SGLT2 (sodium-glucose cotransporter 2) inhibitors, soluble guanylate cyclase stimulators, and myosin activators. With the exception of SGLT2 inhibitors, each of these therapeutic advances was informed by the insights provided by existing conceptual models of heart failure. Although the quest to determine the mechanism of action of SGLT2 inhibitors is ongoing, this therapeutic class of drugs may represent the most important advance in cardiovascular therapeutics of recent decades and may lead to rethinking or expanding our current conceptual models for HFrEF.
Collapse
Affiliation(s)
- Douglas L. Mann
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63108
| | - G. Michael Felker
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63108
| |
Collapse
|
59
|
Dasari TW, Csipo T, Amil F, Lipecz A, Fulop GA, Jiang Y, Samannan R, Johnston S, Zhao YD, Silva-Palacios F, Stavrakis S, Yabluchanskiy A, Po SS. Effects of Low-Level Tragus Stimulation on Endothelial Function in Heart Failure With Reduced Ejection Fraction. J Card Fail 2021; 27:568-576. [PMID: 33387632 PMCID: PMC9473302 DOI: 10.1016/j.cardfail.2020.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Autonomic dysregulation in heart failure with reduced ejection fraction plays a major role in endothelial dysfunction. Low-level tragus stimulation (LLTS) is a novel, noninvasive method of autonomic modulation. METHODS AND RESULTS We enrolled 50 patients with heart failure with reduced ejection fraction (left ventricular ejection fraction of ≤40%) in a randomized, double-blinded, crossover study. On day 1, patients underwent 60 minutes of LLTS with a transcutaneous stimulator (20 Hz, 200 μs pulse width) or sham (ear lobule) stimulation. Macrovascular function was assessed using flow-mediated dilatation in the brachial artery and cutaneous microcirculation with laser speckle contrast imaging in the hand and nail bed. On day 2, patients were crossed over to the other study arm and underwent sham or LLTS; vascular tests were repeated before and after stimulation. Compared with the sham, LLTS improved flow-mediated dilatation by increasing the percent change in the brachial artery diameter (from 5.0 to 7.5, LLTS on day 1, P = .02; and from 4.9 to 7.1, LLTS on day 2, P = .003), compared with no significant change in the sham group (from 4.6 to 4.7, P = .84 on day 1; and from 5.6 to 5.9 on day 2, P = .65). Cutaneous microcirculation in the hand showed no improvement and perfusion of the nail bed showed a trend toward improvement. CONCLUSIONS Our study demonstrated the beneficial effects of acute neuromodulation on macrovascular function. Larger studies to validate these findings and understand mechanistic links are warranted.
Collapse
Affiliation(s)
- Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine; Heart Rhythm Institute.
| | - Tamas Csipo
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | - Faris Amil
- Cardiovascular Section, Department of Internal Medicine
| | - Agnes Lipecz
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | - Gabor A Fulop
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | | | | | - Sarah Johnston
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yan D Zhao
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | | | - Stavros Stavrakis
- Cardiovascular Section, Department of Internal Medicine; Heart Rhythm Institute
| | - Andriy Yabluchanskiy
- Department of Biochemistry and Molecular Biology; Section of Geriatrics, Department of Internal Medicine
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine; Heart Rhythm Institute
| |
Collapse
|
60
|
Scridon A, Halaţiu VB, Balan AI, Cozac DA, Moldovan V, Bănescu C, Perian M, Şerban RC. Long-Term Effects of Ivabradine on Cardiac Vagal Parasympathetic Function in Normal Rats. Front Pharmacol 2021; 12:596956. [PMID: 33897414 PMCID: PMC8061748 DOI: 10.3389/fphar.2021.596956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background: The complex interactions that exist between the pacemaker current, I f, and the parasympathetic nervous system could significantly influence the course of patients undergoing chronic therapy with the I f blocker ivabradine. We thus aimed to assess the effects of chronic ivabradine therapy on autonomic modulation and on the cardiovascular response to in situ and in vitro parasympathetic stimulation. The right atrial expression of HCN genes, encoding proteins for I f, was also evaluated. Methods: Sympathetic and parasympathetic heart rate variability parameters and right atrial HCN(1-4) RNA levels were analyzed in 6 Control and 10 ivabradine-treated male Wistar rats (IVA; 3 weeks, 10 mg/kg/day). The heart rate (HR) and systolic blood pressure (SBP) responses to in situ electrical stimulation of the vagus nerve (2-20 Hz) were assessed in 6 additional Control and 10 IVA rats. The spontaneous sinus node discharge rate (SNDR) response to in vitro cholinergic receptors stimulation using carbamylcholine (10-9-10-6 mol/L) was also assessed in these later rats. Results: Ivabradine significantly increased vagal modulation and shifted the sympatho-vagal balance toward vagal dominance. In Control, in situ vagus nerve stimulation induced progressive decrease in both the SBP (p = 0.0001) and the HR (p< 0.0001). Meanwhile, in IVA, vagal stimulation had no effect on the HR (p = 0.16) and induced a significantly lower drop in SBP (p< 0.05). IVA also displayed a significantly lower SNDR drop in response to carbamylcholine (p< 0.01) and significantly higher right atrial HCN4 expression (p = 0.02). Conclusion: Chronic ivabradine administration enhanced vagal modulation in healthy rats. In addition, ivabradine reduced the HR response to direct muscarinic receptors stimulation, canceled the cardioinhibitory response and blunted the hemodynamic response to in situ vagal stimulation. These data bring new insights into the mechanisms of ivabradine-related atrial proarrhythmia and suggest that long-term I f blockade may protect against excessive bradycardia induced by acute vagal activation.
Collapse
Affiliation(s)
- Alina Scridon
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Vasile Bogdan Halaţiu
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania
| | - Alkora Ioana Balan
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania
| | - Dan Alexandru Cozac
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Emergency Institute for Cardiovascular Diseases and Transplantation Târgu Mureş, Târgu Mureş, Romania
| | - Valeriu Moldovan
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Claudia Bănescu
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Marcel Perian
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Răzvan Constantin Şerban
- Emergency Institute for Cardiovascular Diseases and Transplantation Târgu Mureş, Târgu Mureş, Romania
| |
Collapse
|
61
|
Floras JS. From Brain to Blood Vessel: Insights From Muscle Sympathetic Nerve Recordings: Arthur C. Corcoran Memorial Lecture 2020. Hypertension 2021; 77:1456-1468. [PMID: 33775112 DOI: 10.1161/hypertensionaha.121.16490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiunit recordings of postganglionic sympathetic outflow to muscle yield otherwise imperceptible insights into sympathetic neural modulation of human vascular resistance and blood pressure. This Corcoran Lecture will illustrate the utility of microneurography to investigate neurogenic cardiovascular regulation; review data concerning muscle sympathetic nerve activity of women and men with normal and high blood pressure; explore 2 concepts, central upregulation of muscle sympathetic outflow and cortical autonomic neuroplasticity; present sleep apnea as an imperfect model of neurogenic hypertension; and expose the paradox of sympathetic excitation without hypertension. In awake healthy normotensive individuals, resting muscle sympathetic nerve activity increases with age, sleep fragmentation, and obstructive apnea. Its magnitude is not signaled by heart rate. Age-related changes are nonlinear and differ by sex. In men, sympathetic nerve activity increases with age but without relation to their blood pressure, whereas in women, both rise concordantly after age 40. Mean values for muscle sympathetic nerve activity burst incidence are consistently higher in cohorts with hypertension than in matched normotensives, yet women's sympathetic nerve traffic can increase 3-fold between ages 30 and 70 without causing hypertension. Thus, increased sympathetic nerve activity may be necessary but is insufficient for primary hypertension. Moreover, its inhibition does not consistently decrease blood pressure. Despite a half-century of microneurographic research, large gaps remain in our understanding of the content of the sympathetic broadcast from brain to blood vessel and its specific individual consequences for circulatory regulation and cardiovascular, renal, and metabolic risk.
Collapse
Affiliation(s)
- John S Floras
- Sinai Health and University Health Network Division of Cardiology, Toronto General Hospital Research Institute, and the Department of Medicine, University of Toronto
| |
Collapse
|
62
|
Pelat M, Barbe F, Daveu C, Ly-Nguyen L, Lartigue T, Marque S, Tavares G, Ballet V, Guillon JM, Steinmeyer K, Wirth K, Gögelein H, Arndt P, Rackelmann N, Weston J, Bellevergue P, McCort G, Trellu M, Lucats L, Beauverger P, Pruniaux-Harnist MP, Janiak P, Chézalviel-Guilbert F. SAR340835, a Novel Selective Na +/Ca 2+ Exchanger Inhibitor, Improves Cardiac Function and Restores Sympathovagal Balance in Heart Failure. J Pharmacol Exp Ther 2021; 377:293-304. [PMID: 33602875 DOI: 10.1124/jpet.120.000238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/08/2021] [Indexed: 11/22/2022] Open
Abstract
In failing hearts, Na+/Ca2+ exchanger (NCX) overactivity contributes to Ca2+ depletion, leading to contractile dysfunction. Inhibition of NCX is expected to normalize Ca2+ mishandling, to limit afterdepolarization-related arrhythmias, and to improve cardiac function in heart failure (HF). SAR340835/SAR296968 is a selective NCX inhibitor for all NCX isoforms across species, including human, with no effect on the native voltage-dependent calcium and sodium currents in vitro. Additionally, it showed in vitro and in vivo antiarrhythmic properties in several models of early and delayed afterdepolarization-related arrhythmias. Its effect on cardiac function was studied under intravenous infusion at 250,750 or 1500 µg/kg per hour in dogs, which were either normal or submitted to chronic ventricular pacing at 240 bpm (HF dogs). HF dogs were infused with the reference inotrope dobutamine (10 µg/kg per minute, i.v.). In normal dogs, NCX inhibitor increased cardiac contractility (dP/dtmax) and stroke volume (SV) and tended to reduce heart rate (HR). In HF dogs, NCX inhibitor significantly and dose-dependently increased SV from the first dose (+28.5%, +48.8%, and +62% at 250, 750, and 1500 µg/kg per hour, respectively) while significantly increasing dP/dtmax only at 1500 (+33%). Furthermore, NCX inhibitor significantly restored sympathovagal balance and spontaneous baroreflex sensitivity (BRS) from the first dose and reduced HR at the highest dose. In HF dogs, dobutamine significantly increased dP/dtmax and SV (+68.8%) but did not change HR, sympathovagal balance, or BRS. Overall, SAR340835, a selective potent NCX inhibitor, displayed a unique therapeutic profile, combining antiarrhythmic properties, capacity to restore systolic function, sympathovagal balance, and BRS in HF dogs. NCX inhibitors may offer new therapeutic options for acute HF treatment. SIGNIFICANCE STATEMENT: HF is facing growing health and economic burden. Moreover, patients hospitalized for acute heart failure are at high risk of decompensation recurrence, and no current acute decompensated HF therapy definitively improved outcomes. A new potent, Na+/Ca2+ exchanger inhibitor SAR340835 with antiarrhythmic properties improved systolic function of failing hearts without creating hypotension, while reducing heart rate and restoring sympathovagal balance. SAR340835 may offer a unique and attractive pharmacological profile for patients with acute heart failure as compared with current inotrope, such as dobutamine.
Collapse
Affiliation(s)
- Michel Pelat
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Fabrice Barbe
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Cyril Daveu
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Laetitia Ly-Nguyen
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Thomas Lartigue
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Suzanne Marque
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Georges Tavares
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Véronique Ballet
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Jean-Michel Guillon
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Klaus Steinmeyer
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Klaus Wirth
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Heinz Gögelein
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Petra Arndt
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Nils Rackelmann
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - John Weston
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Patrice Bellevergue
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Gary McCort
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Marc Trellu
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Laurence Lucats
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Philippe Beauverger
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Marie-Pierre Pruniaux-Harnist
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Philip Janiak
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Frédérique Chézalviel-Guilbert
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| |
Collapse
|
63
|
Chen J, Abbod M, Shieh JS. Pain and Stress Detection Using Wearable Sensors and Devices-A Review. SENSORS (BASEL, SWITZERLAND) 2021; 21:1030. [PMID: 33546235 PMCID: PMC7913347 DOI: 10.3390/s21041030] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
Pain is a subjective feeling; it is a sensation that every human being must have experienced all their life. Yet, its mechanism and the way to immune to it is still a question to be answered. This review presents the mechanism and correlation of pain and stress, their assessment and detection approach with medical devices and wearable sensors. Various physiological signals (i.e., heart activity, brain activity, muscle activity, electrodermal activity, respiratory, blood volume pulse, skin temperature) and behavioral signals are organized for wearables sensors detection. By reviewing the wearable sensors used in the healthcare domain, we hope to find a way for wearable healthcare-monitoring system to be applied on pain and stress detection. Since pain leads to multiple consequences or symptoms such as muscle tension and depression that are stress related, there is a chance to find a new approach for chronic pain detection using daily life sensors or devices. Then by integrating modern computing techniques, there is a chance to handle pain and stress management issue.
Collapse
Affiliation(s)
- Jerry Chen
- Department of Mechanical Engineering, Yan Ze University, Taoyuan 32003, Taiwan;
| | - Maysam Abbod
- Department of Electronic and Computer Engineering, Brunel University London, Uxbridge UB8 3PH, UK
| | - Jiann-Shing Shieh
- Department of Mechanical Engineering, Yan Ze University, Taoyuan 32003, Taiwan;
| |
Collapse
|
64
|
A Step further-The Role of Trigeminocardiac Reflex in Therapeutic Implications: Hypothesis, Evidence, and Experimental Models. J Neurosurg Anesthesiol 2021; 34:364-371. [PMID: 33538537 DOI: 10.1097/ana.0000000000000760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/04/2021] [Indexed: 11/27/2022]
Abstract
The trigeminocardiac reflex (TCR) is a well-recognized brainstem reflex that represents a unique interaction between the brain and the heart through the Vth and Xth cranial nerves and brainstem nuclei. The TCR has mainly been reported as an intraoperative phenomenon causing cardiovascular changes during skull-base surgeries. However, it is now appreciated that the TCR is implicated during non-neurosurgical procedures and in nonsurgical conditions, and its complex reflex pathways have been explored as potential therapeutic options in various neurological and cardiovascular diseases. This narrative review presents an in-depth overview of hypothetical and experimental models of the TCR phenomenon in relation to the Vth and Xth cranial nerves. In addition, primitive interactions between these 2 cranial nerves and their significance are highlighted. Finally, therapeutic models of the complex interactions of the TCR and areas for further research will be considered.
Collapse
|
65
|
Ausoni S, Calamelli S, Saccà S, Azzarello G. How progressive cancer endangers the heart: an intriguing and underestimated problem. Cancer Metastasis Rev 2021; 39:535-552. [PMID: 32152913 DOI: 10.1007/s10555-020-09869-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since it came into being as a discipline, cardio-oncology has focused on the prevention and treatment of cardiotoxicity induced by antitumor chemotherapy and radiotherapy. Over time, it has been proved that even more detrimental is the direct effect generated by cancer cells that release pro-cachectic factors in the bloodstream. Secreted molecules target different organs at a distance, including the heart. Inflammatory and neuronal modulators released by the tumor bulk, either as free molecules or through exosomes, contribute to the pathogenesis of cardiac disease. Progressive cancer causes cachexia and severe cardiac muscle wasting accompanied by cardiomyocyte atrophy, tissue fibrosis, and several functional impairments up to heart failure. The molecular mechanisms responsible for such a cardiac muscle wasting have been partially elucidated in animal models, but minimally investigated in humans, although severe cardiac dysfunction exacerbates global cachexia and hampers efficient anti-cancer treatments. This review provides an overview of cancer-induced structural cardiac and functional damage, drawing on both clinical and scientific research. We start by looking at the pathophysiological mechanisms and evolving epidemiology and go on to discuss prevention, diagnosis, and a multimodal policy of intervention aimed at providing overall prognosis and global care for patients. Despite much interest in the cardiotoxicity of cancer therapies, the direct tumor effect on the heart remains poorly explored. There is still a lack of diagnostic criteria for the identification of the early stages of cardiac disease in cancer patients, while the possibilities that there are for effective prevention are largely underestimated. Research on innovative therapies has claimed considerable advances in preclinical studies, but none of the molecular targets suitable for clinical application has been approved for therapy. These issues are critically discussed here.
Collapse
Affiliation(s)
- Simonetta Ausoni
- Department of Biomedical Sciences, University of Padua, Padova, Italy.
| | - Sara Calamelli
- Department of Cardiology, Local Health Unit 3 Serenissima, Mirano Hospital, Mirano, Venice, Italy
| | - Salvatore Saccà
- Department of Cardiology, Local Health Unit 3 Serenissima, Mirano Hospital, Mirano, Venice, Italy
| | - Giuseppe Azzarello
- Department of Medical Oncology, Local Health Unit 3 Serenissima, Mirano Hospital, Mirano, Venice, Italy.
| |
Collapse
|
66
|
Keir DA, Duffin J, Floras JS. Measuring Peripheral Chemoreflex Hypersensitivity in Heart Failure. Front Physiol 2020; 11:595486. [PMID: 33447244 PMCID: PMC7802759 DOI: 10.3389/fphys.2020.595486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/30/2020] [Indexed: 01/08/2023] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) induces chronic sympathetic activation. This disturbance is a consequence of both compensatory reflex disinhibition in response to lower cardiac output and patient-specific activation of one or more excitatory stimuli. The result is the net adrenergic output that exceeds homeostatic need, which compromises cardiac, renal, and vascular function and foreshortens lifespan. One such sympatho-excitatory mechanism, evident in ~40-45% of those with HFrEF, is the augmentation of carotid (peripheral) chemoreflex ventilatory and sympathetic responsiveness to reductions in arterial oxygen tension and acidosis. Recognition of the contribution of increased chemoreflex gain to the pathophysiology of HFrEF and to patients' prognosis has focused attention on targeting the carotid body to attenuate sympathetic drive, alleviate heart failure symptoms, and prolong life. The current challenge is to identify those patients most likely to benefit from such interventions. Two assumptions underlying contemporary test protocols are that the ventilatory response to acute hypoxic exposure quantifies accurately peripheral chemoreflex sensitivity and that the unmeasured sympathetic response mirrors the determined ventilatory response. This Perspective questions both assumptions, illustrates the limitations of conventional transient hypoxic tests for assessing peripheral chemoreflex sensitivity and demonstrates how a modified rebreathing test capable of comprehensively quantifying both the ventilatory and sympathoneural efferent responses to peripheral chemoreflex perturbation, including their sensitivities and recruitment thresholds, can better identify individuals most likely to benefit from carotid body intervention.
Collapse
Affiliation(s)
- Daniel A. Keir
- University Health Network and Mount Sinai Hospital Division of Cardiology and Department of Medicine, University of Toronto, Toronto General Research Institute, Toronto, ON, Canada
- School of Kinesiology, The University of Western Ontario, London, ON, Canada
| | - James Duffin
- Department of Anesthesia and Pain Management, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Thornhill Research Inc., Toronto, ON, Canada
| | - John S. Floras
- University Health Network and Mount Sinai Hospital Division of Cardiology and Department of Medicine, University of Toronto, Toronto General Research Institute, Toronto, ON, Canada
| |
Collapse
|
67
|
Badrov MB, Mak S, Floras JS. Cardiovascular Autonomic Disturbances in Heart Failure With Preserved Ejection Fraction. Can J Cardiol 2020; 37:609-620. [PMID: 33310140 DOI: 10.1016/j.cjca.2020.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/09/2023] Open
Abstract
In heart failure with reduced ejection fraction (HFrEF), diminished tonic and reflex vagal heart rate modulation and exaggerated sympathetic outflow and neural norepinephrine release are evident from disease inception. Each of these disturbances of autonomic regulation has been independently associated with shortened survival, and β-adrenoceptor antagonism and therapeutic autonomic modulation by other means have been demonstrated, in clinical trials, to lessen symptoms and prolong survival. In contrast, data concerning the autonomic status of patients with heart failure with preserved ejection fraction (HFpEF) are comparatively sparse. Little is known concerning the prognostic consequences of autonomic dysregulation in such individuals, and therapies applied with success in HFrEF have in most trials failed to improve symptoms or survival of those with HFpEF. A recent HFpEF Expert Scientific Panel report emphasised that without a deeper understanding of the pathophysiology of HFpEF, establishing effective treatment will be challenging. One aspect of such pathology may be cardiovascular autonomic disequilibrium, often worsened by acute exercise or routine daily activity. This review aims to summarise existing knowledge concerning parasympathetic and sympathetic function of patients with HFpEF, consider potential mechanisms and specific consequences of autonomic disturbances that have been identified, and propose hypotheses for future investigation.
Collapse
Affiliation(s)
- Mark B Badrov
- Division of Cardiology, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Susanna Mak
- Division of Cardiology, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - John S Floras
- Division of Cardiology, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
68
|
Grassi G, Seravalle G, Esler M. Sympathomodulation in congestive heart failure: From drugs to devices. Int J Cardiol 2020; 321:118-125. [DOI: 10.1016/j.ijcard.2020.07.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/15/2023]
|
69
|
Maack C, Eschenhagen T, Hamdani N, Heinzel FR, Lyon AR, Manstein DJ, Metzger J, Papp Z, Tocchetti CG, Yilmaz MB, Anker SD, Balligand JL, Bauersachs J, Brutsaert D, Carrier L, Chlopicki S, Cleland JG, de Boer RA, Dietl A, Fischmeister R, Harjola VP, Heymans S, Hilfiker-Kleiner D, Holzmeister J, de Keulenaer G, Limongelli G, Linke WA, Lund LH, Masip J, Metra M, Mueller C, Pieske B, Ponikowski P, Ristić A, Ruschitzka F, Seferović PM, Skouri H, Zimmermann WH, Mebazaa A. Treatments targeting inotropy. Eur Heart J 2020; 40:3626-3644. [PMID: 30295807 DOI: 10.1093/eurheartj/ehy600] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/06/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Acute heart failure (HF) and in particular, cardiogenic shock are associated with high morbidity and mortality. A therapeutic dilemma is that the use of positive inotropic agents, such as catecholamines or phosphodiesterase-inhibitors, is associated with increased mortality. Newer drugs, such as levosimendan or omecamtiv mecarbil, target sarcomeres to improve systolic function putatively without elevating intracellular Ca2+. Although meta-analyses of smaller trials suggested that levosimendan is associated with a better outcome than dobutamine, larger comparative trials failed to confirm this observation. For omecamtiv mecarbil, Phase II clinical trials suggest a favourable haemodynamic profile in patients with acute and chronic HF, and a Phase III morbidity/mortality trial in patients with chronic HF has recently begun. Here, we review the pathophysiological basis of systolic dysfunction in patients with HF and the mechanisms through which different inotropic agents improve cardiac function. Since adenosine triphosphate and reactive oxygen species production in mitochondria are intimately linked to the processes of excitation-contraction coupling, we also discuss the impact of inotropic agents on mitochondrial bioenergetics and redox regulation. Therefore, this position paper should help identify novel targets for treatments that could not only safely improve systolic and diastolic function acutely, but potentially also myocardial structure and function over a longer-term.
Collapse
Affiliation(s)
- Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, Würzburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Partner site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Frank R Heinzel
- Department of Cardiology, Charité University Medicine, Berlin, Germany
| | - Alexander R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany.,Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Joseph Metzger
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - M Birhan Yilmaz
- Department of Cardiology, Cumhuriyet University, Sivas, Turkey
| | - Stefan D Anker
- Department of Cardiology and Pneumology, University Medical Center Göttingen and DZHK (German Center for Cardiovascular Research), Göttingen, Germany.,Division of Cardiology and Metabolism - Heart Failure, Cachexia and Sarcopenia, Department of Internal Medicine and Cardiology, Berlin-Brandenburg Center for Regenerative Therapies (BCRT) at Charité University Medicine, Berlin, Germany
| | - Jean-Luc Balligand
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Universite Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover D-30625, Germany
| | | | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Partner site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Stefan Chlopicki
- Department of Pharmacology, Medical College, Jagiellonian University, Krakow, Poland
| | - John G Cleland
- University of Hull, Kingston upon Hull, UK.,National Heart and Lung Institute, Royal Brompton and Harefield Hospitals NHS Trust, Imperial College, London, UK
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander Dietl
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Rodolphe Fischmeister
- Inserm UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | | | | | | | | | - Gilles de Keulenaer
- Laboratory of Physiopharmacology (University of Antwerp) and Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Giuseppe Limongelli
- Department of Cardiothoracic Sciences, Second University of Naples, Naples, Italy
| | | | - Lars H Lund
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josep Masip
- Intensive Care Department, Consorci Sanitari Integral, University of Barcelona, Spain
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel (CRIB), University Hospital Basel, University of Basel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin, and German Centre for Cardiovascular Research (DZHK), Partner site Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Piotr Ponikowski
- Department of Cardiology, Medical University, Clinical Military Hospital, Wroclaw, Poland
| | - Arsen Ristić
- Department of Cardiology of the Clinical Center of Serbia and Belgrade University School of Medicine, Belgrade, Serbia
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Switzerland
| | | | - Hadi Skouri
- Division of Cardiology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner siteGöttingen, Göttingen, Germany
| | - Alexandre Mebazaa
- Hôpital Lariboisière, Université Paris Diderot, Inserm U 942, Paris, France
| |
Collapse
|
70
|
Abstract
The view that chronic heart failure was exclusively a disease of the heart dominated the cardiovascular literature until relatively recently. However, over the last 40 years it has increasingly come to be seen as a multisystem disease. Aside from changes in the sympathetic and parasympathetic nervous systems and the renin-angiotensin-aldosterone system, adaptations to the lungs, muscles and gastrointestinal tract have been clearly documented. It is clear that the brain and CNS are also affected in patients with heart failure, although this is often under recognized. The purpose of this review is to summarize the changes in the structure and biochemical function of the CNS in patients with chronic heart failure and to discuss their potential importance.
Collapse
Affiliation(s)
- Mark Dayer
- Department of Cardiology, Musgrove Park Hospital, Taunton, TA1 5DA, UK
| | - David H MacIver
- Department of Cardiology, Musgrove Park Hospital, Taunton, TA1 5DA, UK.,Biological Physics Group, School of Physics & Astronomy, University of Manchester, Manchester, M13 9PL, UK
| | - Stuart D Rosen
- Ealing & Royal Brompton Hospitals, Uxbridge Rd, Southall, UB1 3HW, UK.,Imperial College London, South Kensington, London, SW7 2BU, UK
| |
Collapse
|
71
|
Borovac JA, D'Amario D, Bozic J, Glavas D. Sympathetic nervous system activation and heart failure: Current state of evidence and the pathophysiology in the light of novel biomarkers. World J Cardiol 2020; 12:373-408. [PMID: 32879702 PMCID: PMC7439452 DOI: 10.4330/wjc.v12.i8.373] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/19/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome characterized by the activation of at least several neurohumoral pathways that have a common role in maintaining cardiac output and adequate perfusion pressure of target organs and tissues. The sympathetic nervous system (SNS) is upregulated in HF as evident in dysfunctional baroreceptor and chemoreceptor reflexes, circulating and neuronal catecholamine spillover, attenuated parasympathetic response, and augmented sympathetic outflow to the heart, kidneys and skeletal muscles. When these sympathoexcitatory effects on the cardiovascular system are sustained chronically they initiate the vicious circle of HF progression and become associated with cardiomyocyte apoptosis, maladaptive ventricular and vascular remodeling, arrhythmogenesis, and poor prognosis in patients with HF. These detrimental effects of SNS activity on outcomes in HF warrant adequate diagnostic and treatment modalities. Therefore, this review summarizes basic physiological concepts about the interaction of SNS with the cardiovascular system and highlights key pathophysiological mechanisms of SNS derangement in HF. Finally, special emphasis in this review is placed on the integrative and up-to-date overview of diagnostic modalities such as SNS imaging methods and novel laboratory biomarkers that could aid in the assessment of the degree of SNS activation and provide reliable prognostic information among patients with HF.
Collapse
Affiliation(s)
- Josip Anđelo Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
- Working Group on Heart Failure of Croatian Cardiac Society, Zagreb 10000, Croatia
| | - Domenico D'Amario
- Department of Cardiovascular and Thoracic Sciences, IRCCS Fondazione Policlinico A. Gemelli, Universita Cattolica Sacro Cuore, Rome 00168, Italy
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
| | - Duska Glavas
- Working Group on Heart Failure of Croatian Cardiac Society, Zagreb 10000, Croatia
- Clinic for Cardiovascular Diseases, University Hospital of Split, Split 21000, Croatia
| |
Collapse
|
72
|
Dorey TW, Moghtadaei M, Rose RA. Altered heart rate variability in angiotensin II–mediated hypertension is associated with impaired autonomic nervous system signaling and intrinsic sinoatrial node dysfunction. Heart Rhythm 2020; 17:1360-1370. [DOI: 10.1016/j.hrthm.2020.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/16/2020] [Indexed: 10/24/2022]
|
73
|
Machhada A, Hosford PS, Dyson A, Ackland GL, Mastitskaya S, Gourine AV. Optogenetic Stimulation of Vagal Efferent Activity Preserves Left Ventricular Function in Experimental Heart Failure. JACC Basic Transl Sci 2020; 5:799-810. [PMID: 32875170 PMCID: PMC7452237 DOI: 10.1016/j.jacbts.2020.06.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022]
Abstract
This study was designed to determine the effect of selective optogenetic simulation of vagal efferent activity on left ventricular function in an animal (rat) model of MI-induced heart failure. Optogenetic stimulation of dorsal brainstem vagal pre-ganglionic neurons transduced to express light-sensitive channels preserved LV function and exercise capacity in animals with MI. The data suggest that activation of vagal efferents is critically important to deliver the therapeutic benefit of VNS in chronic heart failure.
Large clinical trials designed to test the efficacy of vagus nerve stimulation (VNS) in patients with heart failure did not demonstrate benefits with respect to the primary endpoints. The nonselective nature of VNS may account for the failure to translate promising results of preclinical and earlier clinical studies. This study showed that optogenetic stimulation of vagal pre-ganglionic neurons transduced to express light-sensitive channels preserved left ventricular function and exercise capacity in a rat model of myocardial infarction−induced heart failure. These data suggested that stimulation of vagal efferent activity is critically important to deliver the therapeutic benefit of VNS in heart failure.
Collapse
Key Words
- ABP, arterial blood pressure
- DVMN, dorsal motor nucleus of the vagus nerve
- GRK2, G-protein−coupled receptor kinase 2
- LAD, left anterior descending coronary artery
- LV dP/dtMAX, maximum rate of rise of left ventricular pressure
- LV, left ventricle
- LVEDP, left ventricular end-diastolic pressure
- LVESP, left ventricular end-systolic pressure
- LVP, left ventricular pressure
- LVV, lentiviral vector
- MI, myocardial infarction
- VNS, vagus nerve stimulation
- autonomic nervous system
- eGFP, enhanced green fluorescent protein
- heart failure
- myocardial infarction
- neuromodulation
- vagus nerve stimulation
Collapse
Affiliation(s)
- Asif Machhada
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Patrick S Hosford
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom.,Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alex Dyson
- Clinical Physiology, Division of Medicine, University College London, London, United Kingdom
| | - Gareth L Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Svetlana Mastitskaya
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
74
|
Lee JB, Omazic LJ, Kathia M. What happens in vagus, no longer stays in vagus. J Physiol 2020; 598:4435-4437. [PMID: 32662882 DOI: 10.1113/jp280388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Jordan B Lee
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Lucas J Omazic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Muhammad Kathia
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
75
|
Anker MS, Frey MK, Goliasch G, Bartko PE, Prausmüller S, Gisslinger H, Kornek G, Strunk G, Raderer M, Zielinski C, Hülsmann M, Pavo N. Increased resting heart rate and prognosis in treatment-naïve unselected cancer patients: results from a prospective observational study. Eur J Heart Fail 2020; 22:1230-1238. [PMID: 32202022 PMCID: PMC7540544 DOI: 10.1002/ejhf.1782] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/21/2020] [Accepted: 02/16/2020] [Indexed: 12/19/2022] Open
Abstract
AIMS Cancer patients suffer from impaired cardiovascular function. Elevated resting heart rate (RHR) has been identified as a marker for increased long-term mortality in cancer patients prior to the receipt of anticancer treatment. We aimed to establish whether RHR is associated with survival in treatment-naïve cancer patients. METHODS AND RESULTS This prospective study enrolled 548 unselected treatment-naïve cancer patients between 2011 and 2013. The median age of the cohort was 62 years; 40.9% were male and 32.7% had metastatic disease. Median RHR was 72 b.p.m. Most patients were in sinus rhythm (n = 507, 92.5%). Clinical heart failure was noted in 37 (6.8%) patients. RHR was not related to cancer stage (P = 0.504). Patients in the highest RHR tertile had higher levels of high-sensitivity troponin (P = 0.003) and N-terminal pro-B-type natriuretic peptide (P = 0.039). During a median follow-up of 25 months (interquartile range: 16-32 months; range: 0-40 months), 185 (33.8%) patients died from any cause [1-year-mortality: 17%, 95% confidence interval (CI) 13-20%]. In univariate survival analysis, RHR predicted all-cause mortality [crude hazard ratio (HR) for a 5 b.p.m. increase in RHR: 1.09, 95% CI 1.04-1.15; P < 0.001], and remained significantly associated with outcome after adjustment for age, gender, tumour entity, tumour stage, cardiac status and haemoglobin (adjusted HR for a 5 b.p.m. increase in RHR: 1.10, 95% CI 1.04-1.16; P < 0.001). There was no significant impact of metastatic/non-metastatic disease state on the predictive value of RHR (P = 0.433 for interaction). In subgroup analyses, the strongest associations for RHR with mortality were observed in lung (crude HR 1.14; P = 0.007) and gastrointestinal (crude HR 1.31; P < 0.001) cancer. CONCLUSIONS Treatment-naïve cancer patients with higher RHRs display higher levels of cardiovascular biomarkers. RHR was independently associated with all-cause mortality, especially in lung and gastrointestinal cancers. Elevated RHR and cardiovascular biomarkers may represent early signs of incipient cardiac dysfunction.
Collapse
Affiliation(s)
- Markus S. Anker
- Department of Cardiology & Berlin Institute of Health Center for Regenerative Therapies (BCRT)German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Charité-Universitätsmedizin Berlin (Campus CVK)BerlinGermany
- Department of CardiologyCharité Universitätsmedizin Berlin (Campus CBF)BerlinGermany
| | - Maria K. Frey
- Department of Internal Medicine II, Clinical Division of CardiologyMedical University of ViennaViennaAustria
| | - Georg Goliasch
- Department of Internal Medicine II, Clinical Division of CardiologyMedical University of ViennaViennaAustria
| | - Philipp E. Bartko
- Department of Internal Medicine II, Clinical Division of CardiologyMedical University of ViennaViennaAustria
| | - Suriya Prausmüller
- Department of Internal Medicine II, Clinical Division of CardiologyMedical University of ViennaViennaAustria
| | - Heinz Gisslinger
- Department of Internal Medicine I, Clinical Division of OncologyMedical University of ViennaViennaAustria
| | - Gabriela Kornek
- Department of Internal Medicine I, Clinical Division of OncologyMedical University of ViennaViennaAustria
| | - Guido Strunk
- Department of Complexity Research, Complexity Science HubViennaAustria
- Department of Integrated Safety and SecurityUniversity of Applied Sciences, FH Campus ViennaViennaAustria
- Department of Entrepreneurship and Economic EducationFaculty of Business and Economics, Technical University DortmundDortmundGermany
| | - Markus Raderer
- Department of Internal Medicine I, Clinical Division of OncologyMedical University of ViennaViennaAustria
| | - Christoph Zielinski
- Department of Internal Medicine I, Clinical Division of OncologyMedical University of ViennaViennaAustria
| | - Martin Hülsmann
- Department of Internal Medicine II, Clinical Division of CardiologyMedical University of ViennaViennaAustria
| | - Noemi Pavo
- Department of Internal Medicine II, Clinical Division of CardiologyMedical University of ViennaViennaAustria
| |
Collapse
|
76
|
Sympathetic and baroreflex alterations in congestive heart failure with preserved, midrange and reduced ejection fraction. J Hypertens 2020; 37:443-448. [PMID: 30020242 DOI: 10.1097/hjh.0000000000001856] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM Although abnormalities in reflex sympathetic neural function represent a hallmark of heart failure, no information is available on the neuroadrenergic and baroreflex function in heart failure with preserved, midrange and reduced ejection fraction. The current study was designed to assess muscle sympathetic nerve traffic (MSNA) and baroreflex function in the clinical classes of heart failure defined by the new European Society of Cardiology/American College of Cardiology Foundation/American Heart Association guidelines. METHODS In 32 treated heart failure patients aged 69.3 ± 1.1 (mean ± SEM) classified according to new heart failure guidelines, we measured MSNA (microneurography), spontaneous baroreflex sensitivity and venous plasma norepinephrine (HPLC). Fourteen age-matched healthy individuals represented the control group. RESULTS MSNA was progressively and significantly increased from controls to heart failure conditions characterized by preserved, midrange and reduced ejection fraction (40.4 ± 2.5, 55.6 ± 2.1, 70.4 ± 3 and 78.6 ± 2.6 bursts/100 heart beats, P < 0.01). In contrast, plasma norepinephrine was significantly increased in heart failure with reduced ejection fraction only. Baroreflex sensitivity was significantly reduced in the latter two clinical conditions and almost unaltered in heart failure with preserved ejection fraction. There was an inverse relationship between different markers of adrenergic activity (MSNA, heart rate and plasma norepinephrine), left ventricular ejection fraction and baroreflex function. Brain natriuretic peptides were directly and significantly related to MSNA and plasma norepinephrine. CONCLUSION Thus clinical categories of heart failure patients defined by the new European Society of Cardiology/American College of Cardiology Foundation/American Heart Association classification share as a common pathophysiological link the marked overactivity of the sympathetic nervous system, whose magnitude is significantly and strongly related to the impairment of the left ventricular ejection fraction. A baroreflex dysfunction accompanies in the more severe heart failure state the neuroadrenergic activation.
Collapse
|
77
|
Mineralocorticoid receptor antagonists in heart failure patients with chronic kidney disease. Curr Opin Nephrol Hypertens 2020; 29:258-263. [DOI: 10.1097/mnh.0000000000000583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
78
|
Couffignal C, Amour J, Ait-Hamou N, Cholley B, Fellahi JL, Duval X, Costa De Beauregard Y, Nataf P, Dilly MP, Provenchère S, Montravers P, Mentré F, Longrois D. Timing of β-Blocker Reintroduction and the Occurrence of Postoperative Atrial Fibrillation after Cardiac Surgery: A Prospective Cohort Study. Anesthesiology 2020; 132:267-279. [PMID: 31939841 DOI: 10.1097/aln.0000000000003064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND For cardiac surgery patients under chronic β-blocker therapy, guidelines recommend their early postoperative reintroduction to decrease the incidence of postoperative atrial fibrillation. The authors hypothesized that the timing of β-blocker reintroduction affects their effectiveness on the incidence of postoperative atrial fibrillation. METHODS This multicenter prospective French cohort study included patients on β-blockers (more than 30 days before surgery) in sinus rhythm without a pacemaker. The primary outcome, time sequence of β-blocker reintroduction, was analyzed for 192 h after surgery. The secondary outcome, relationship between the occurrence of postoperative atrial fibrillation and timing of β-blocker reintroduction, was analyzed based on pre- and intraoperative predictors (full and selected sets) according to landmark times (patients in whom atrial fibrillation occurred before a given landmark time were not analyzed). RESULTS Of 663 patients, β-blockers were reintroduced for 532 (80%) but for only 261 (39%) patients in the first 48 h after surgery. Median duration before reintroduction was 49.5 h (95% CI, 48 to 51.5 h). Postoperative atrial fibrillation or death (N = 4) occurred in 290 (44%) patients. After performing a landmark analysis to take into account the timing of β-blocker reintroduction, the adjusted odds ratios (95% CI) for predictor full and selected (increased age, history of paroxysmal atrial fibrillation, and duration of aortic cross clamping) sets for the occurrence of postoperative atrial fibrillation were: adjusted odds ratio (full) = 0.87 (0.58 to 1.32; P = 0.517) and adjusted odds ratio (selected) = 0.84 (0.58 to 1.21; P = 0.338) at 48 h; adjusted odds ratio (full) = 0.64 (0.39 to 1.05; P = 0.076) and adjusted odds ratio (selected) = 0.58 (0.38 to 0.89; P = 0.013) at 72 h; adjusted odds ratio (full) = 0.58 (0.31 to 1.07; P = 0.079) and adjusted odds ratio (selected) = 0.53 (0.31 to 0.91; P = 0.021) at 96 h. CONCLUSIONS β-Blockers were reintroduced early (after less than 48 h) in fewer than half of the cardiac surgery patients. Reintroduction decreased postoperative atrial fibrillation occurrence only at later time points and only in the predictor selected set model. These results are an incentive to optimize (timing, doses, or titration) β-blocker reintroduction after cardiac surgery.
Collapse
Affiliation(s)
- Camille Couffignal
- From the Department of Biostatistics, Bichat-Claude Bernard Hospital, AP-HP.Nord, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France (C.C., F.M.) University of Paris, IAME, UMR1137, Paris, France (C.C., X.D., F.M.) INSERM, IAME, UMR 1137, Paris, France (C.C., X.D., F.M.) Clinical Investigation Center, CIC-1425, AP-HP, INSERM, Paris, France (X.D., Y.C.D.B.) Department of Anesthesiology and Critical Care Medicine, Hôpital Pitié-Salpêtrière, APHP, Sorbonne University, UPMC University, Paris 06, UMR INSERM 1166, IHU ICAN, Paris, France (J.A., N.A.-H.) Department of Anesthesia and Intensive Care, Hôpital Européen Georges Pompidou, APHP Paris-Ouest, University of Paris, Paris, France (B.C.) Department of Anesthesia and Intensive Care, Hôpital Cardiologique Louis Pradel, IHU OPERA Inserm U1060/Faculté de Médecine Lyon Est, University Claude Bernard Lyon 1, Lyon, France (J.-L.F.) Department of Cardiac Surgery, Bichat-Claude Bernard Hospital, AP-HP.Nord, APHP, Paris, France (P.N.) Department of Anesthesia and Intensive Care, Bichat-Claude Bernard Hospital, AP-HP.Nord, APHP, Paris, France (M.-P.D., S.P., P.M., D.L.) University of Paris, Paris, France (P.M., D.L.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Poliwczak AR, Białkowska J, Woźny J, Koziróg M, Bała A, Jabłkowski M. Cardiovascular risk assessment by electrocardiographic Holter monitoring in patients with chronic hepatitis C. Arch Med Sci 2020; 16:1031-1039. [PMID: 32863991 PMCID: PMC7444696 DOI: 10.5114/aoms.2020.96600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/05/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Cardiovascular diseases are ranked as the third cause of mortality among people infected with hepatitis C virus (HCV), but the relationship of infection with cardiovascular risk remains disputable. We have focused on the comprehensive use of parameters obtainable during long-term electrocardiographic (ECG) Holter monitoring. MATERIAL AND METHODS Heart rate variability and turbulence (HRV and HRT), deceleration/acceleration capacity (DC/AC), corrected QT interval (QTc) and late potential (LP) were used. 36 persons were included, and 30 healthy subjects formed a control group. All were submitted to 24-hour Holter ECG-monitoring. RESULTS The studied groups were not statistically significantly different with regards to basic anthropometric parameters. Statistically significantly higher medium and maximum heart rhythm and aminotransferase activities were recorded in patients with hepatitis C. The HRV parameters r-MSSD, p50NN, HF, and absolute DC/AC values were significantly lower in the subjects with hepatitis C than those in the control group. The QTc interval, measured for nocturnal hours, was also significantly longer in that group. There were no differences in the albumin level or basic echocardiographic parameters, including left ventricle ejection fraction. Nor was there any difference in the HRT parameters, or LP. The most interesting observation was the positive correlation among the number of viral RNA copies and DC, and LF. CONCLUSIONS We confirmed the presence of autonomic disorders with prevalence of sympathetic system activity and prolonged QTc interval in patients with chronic hepatitis C. Those parameters significantly correlated with infection intensity. Our results suggest that HCV infection could be an independent cardiovascular risk factor, not associated with the lipid profile. Further prospective studies are needed.
Collapse
Affiliation(s)
- Adam R. Poliwczak
- Department of Human Physiology, Medical University of Lodz, Lodz, Poland
| | - Jolanta Białkowska
- Department of Infectious and Liver Diseases, Medical University of Lodz, Lodz, Poland
| | - Joanna Woźny
- Department of Infectious and Liver Diseases, Medical University of Lodz, Lodz, Poland
| | - Marzena Koziróg
- Department of Internal Diseases and Cardiac Rehabilitation, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Bała
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Maciej Jabłkowski
- Department of Infectious and Liver Diseases, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
80
|
Abstract
Sarcopaenia is defined as reduced skeletal muscle mass associated with either a decline in muscle strength or low physical performance. It has been shown to affect 17.5% of people worldwide, with a prevalence of 20% or higher in patients with heart failure (HF). Sarcopaenia has severe impact on mortality, physical capacity, and quality of life. Even though several mechanisms, such as autonomic imbalance, reduced muscle blood flow, increased inflammation, hormonal alterations, increased apoptosis, and autophagy have been proposed to fuel the pathogenesis of sarcopaenia, additional studies assessing the interaction of these conditions need to be conducted to elucidate how the presence of sarcopaenia can exacerbate the progression of HF and vice-versa. Resistance training combined with nutritional protein intake seems to be effective in the treatment of sarcopaenia, although current pharmacotherapies have not been extensively studied with this endpoint in mind. In conclusion, sarcopaenia is interwoven with HF and leads to worse exercise capacity in these patients. The mechanisms associated with this bilateral relationship between sarcopaenia and HF are still to be elucidated, leading to effective treatment, not only for the heart, but also for the skeletal muscle.
Collapse
Affiliation(s)
- Guilherme Wesley Peixoto da Fonseca
- Cardiovascular Rehabilitation and Exercise Physiology Unit, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr. Enéas de Carvalho Aguiar, 44 - Cerqueira Cesar, 05403-900 São Paulo, Brazil.,Department of Cardiology and Pneumology, University Medicine Göttingen (UMG), Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medicine Göttingen (UMG), Robert-Koch-Straße 40, 37075 Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| |
Collapse
|
81
|
Seferović PM. Introduction to the special issue entitled 'Heart failure management of the elderly patient: focus on frailty, sarcopenia, cachexia, and dementia'. Eur Heart J Suppl 2019; 21:L1-L3. [PMID: 31885503 PMCID: PMC6926411 DOI: 10.1093/eurheartj/suz234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This is a special issue focused on heart failure management of the elderly patient with a focus on frailty, sarcopaenia, cachexia, and dementia, all common problems in the contemporary older heart failure (HF) patient. The Heart Failure Association (HFA) of the European Society of Cardiology (ESC) has brought together experts to discuss these topical and clinically difficult areas. There are papers on ageing, demographics, and heart failure, drug treatment of the older patient, the frail heart failure patient and how to recognize frailty and screen for it without the risk segmenting these patients in a form of discrimination of them as less worthy of treatment through ‘frailtyism’. This is also discussion of the common problems affecting skeletal muscle, both sarcopaenia and cachexia, as well as dementia and cognitive decline and the crucial issue of planning health care for the older patient with HF most effectively by the use of care plans.
Collapse
Affiliation(s)
- Petar M Seferović
- Faculty of Medicine, University of Belgrade, 8 Dr Subotića, 11000 Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
82
|
Abstract
Heart and brain disorders more frequently co-exist than by chance alone, due to having common risk factors and a degree of interaction. In the setting of heart failure (HF) in the elderly strokes, dementia, and depression are all common and can produce a particularly difficult series of clinical problems to manage. Loss of ability to self-care can lead to very poor quality of life and a dramatic increase in health care expenditure. The Heart Failure Association of the ESC as part of its workshop on physiological monitoring of the complex multi-morbid HF patient reviewed screening, monitoring, prevention, and management of cognitive decline within the setting of HF.
Collapse
Affiliation(s)
- Wolfram Doehner
- Department of Cardiology Campus Virchow, Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Föhrer Str. 15, 13353 Berlin, Germany.,Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
83
|
Abstract
Cachexia is a multifactorial disease characterized by a pathologic shift of metabolism towards a more catabolic state. It frequently occurs in patients with chronic diseases such as chronic heart failure and is especially common in the elderly. In patients at risk, cardiac cachexia is found in about 10% of heart failure patients. The negative impact of cardiac cachexia on mortality, morbidity, and quality of life demonstrates the urgent need to find new effective therapies against cardiac cachexia. Furthermore, exercise training and nutritional support can help patients with cardiac cachexia. Despite ongoing efforts to find new therapies for cachexia treatment, also new preventive strategies are needed.
Collapse
Affiliation(s)
- Alessia Lena
- Division of Cardiology and Metabolism, Department of Cardiology, Charité-Campus Virchow Klinikum (CVK), Augustenburger Platz 1, 13353 Berlin, Germany.,Department of Cardiology, Charité-Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Föhrer Str. 15, 13353 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| | - Nicole Ebner
- Department of Cardiology, University Medical Center Goettingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Markus S Anker
- Division of Cardiology and Metabolism, Department of Cardiology, Charité-Campus Virchow Klinikum (CVK), Augustenburger Platz 1, 13353 Berlin, Germany.,Department of Cardiology, Charité-Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Föhrer Str. 15, 13353 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| |
Collapse
|
84
|
Coats AJS. Heart failure management of the elderly patient: focus on frailty, sarcopaenia, cachexia, and dementia: conclusions. Eur Heart J Suppl 2019; 21:L36-L38. [PMID: 31885512 PMCID: PMC6926414 DOI: 10.1093/eurheartj/suz236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2019] [Indexed: 12/30/2022]
Abstract
With the ageing of populations heart failure is becoming more common and more complex. It is affecting ever older patients and the number of prevalent comorbidities is rising. Even as we continue to gain success in large-scale clinical trials with more effective therapies so our patients are becoming more complex. One of the biggest challenges is the effect of age. Frailty, comorbidity, sarcopaenia, cachexia, polypharmacy, and cognitive decline are all challenging our patients as never before and these challenges will be difficult for cash strapped health care systems to manage. For these reasons, the Heart Failure Association brought together a panel of experts to debate and review this complex area, championing the need for us to establish better ways of caring for the patients of the future.
Collapse
Affiliation(s)
- Andrew J Stewart Coats
- Department of Medical Sciences, Centre for Clinical and Basic Research, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Roma, Italy
| |
Collapse
|
85
|
Coats AJ. Heart Failure Association position papers – a new way to advance the field. Eur J Heart Fail 2019; 22:6-7. [DOI: 10.1002/ejhf.1704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023] Open
|
86
|
Grassi G, Quarti-Trevano F, Esler MD. Sympathetic activation in congestive heart failure: an updated overview. Heart Fail Rev 2019; 26:173-182. [PMID: 31832833 DOI: 10.1007/s10741-019-09901-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Conclusive evidence demonstrates that the sympathetic nervous system activation is a hallmark of congestive heart failure. This has been shown via a variety of biochemical, neurophysiological, and neuroimaging approaches for studying human sympathetic neural function. The sympathetic activation appears to be an early phenomenon in the clinical course of the disease, closely related to its severity and potentiated by the concomitant presence of other comorbidities, such as obesity, diabetes mellitus, metabolic syndrome, hypertension, and renal failure. The adrenergic overdrive in heart failure is associated with other sympathetic abnormalities, such as the downregulation of beta-adrenergic adrenoreceptors at cardiac level, and exerts unfavorable consequences on the cardiovascular system. These include the endothelial dysfunction, the development of left ventricular hypertrophy, the atherosclerosis development, as well as the generation of atrial and ventricular arrhythmias, and, at very extreme levels of sympathetic activation, the occurrence of microscopic myocardial necrosis. Given the close direct independent relationships detected in heart failure between sympathetic activation and mortality, the adrenergic overdrive has become a target of neuromodulatory therapeutic interventions, which include non-pharmacological, pharmacological, and device-based interventions. For some of these approaches (specifically bilateral renal nerves ablation and carotid baroreceptor stimulation), additional studies are needed to better define their impact on the clinical course of the disease.
Collapse
Affiliation(s)
- Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Via Pergolesi 33, 20052, Monza, Italy.
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Via Pergolesi 33, 20052, Monza, Italy
| | - Murray D Esler
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
87
|
Anker MS, Hadzibegovic S, Lena A, Belenkov Y, Bergler‐Klein J, de Boer RA, Farmakis D, von Haehling S, Iakobishvili Z, Maack C, Pudil R, Skouri H, Cohen‐Solal A, Tocchetti CG, Coats AJ, Seferović PM, Lyon AR. Recent advances in cardio-oncology: a report from the 'Heart Failure Association 2019 and World Congress on Acute Heart Failure 2019'. ESC Heart Fail 2019; 6:1140-1148. [PMID: 31884717 PMCID: PMC6989292 DOI: 10.1002/ehf2.12551] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
While anti-cancer therapies, including chemotherapy, immunotherapy, radiotherapy, and targeted therapy, are constantly advancing, cardiovascular toxicity has become a major challenge for cardiologists and oncologists. This has led to an increasing demand of cardio-oncology units in Europe and a growing interest of clinicians and researchers. The Heart Failure 2019 meeting of the Heart Failure Association of the European Society of Cardiology in Athens has therefore created a scientific programme that included four dedicated sessions on the topic along with several additional lectures. The major points that were discussed at the congress included the implementation and delivery of a cardio-oncology service, the collaboration among cardio-oncology experts, and the risk stratification, prevention, and early recognition of cardiotoxicity. Furthermore, sessions addressed the numerous different anti-cancer therapies associated with cardiotoxic effects and provided guidance on how to treat cancer patients who develop cardiovascular disease before, during, and after treatment.
Collapse
Affiliation(s)
- Markus S. Anker
- Division of Cardiology and Metabolism, Department of CardiologyCharité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research)partner site Berlin and Department of Cardiology, Charité Campus Benjamin FranklinBerlinGermany
| | - Sara Hadzibegovic
- Division of Cardiology and Metabolism, Department of CardiologyCharité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research)partner site Berlin and Department of Cardiology, Charité Campus Benjamin FranklinBerlinGermany
| | - Alessia Lena
- Division of Cardiology and Metabolism, Department of CardiologyCharité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research)partner site Berlin and Department of Cardiology, Charité Campus Benjamin FranklinBerlinGermany
| | | | | | - Rudolf A. de Boer
- Department of CardiologyUniversity of GroningenUniversity Medical Center Groningen, GroningenThe Netherlands
| | - Dimitrios Farmakis
- University of Cyprus Medical SchoolNicosiaCyprus
- Department of Cardiology, Cardio‐Oncology Clinic, Heart Failure UnitAthens University Hospital ‘Attikon’, National and Kapodistrian University of AthensAthensGreece
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center Göttingen, German Center for Cardiovascular Medicine (DZHK)University of Göttingen Medical Center, Georg‐August‐UniversityGöttingenGermany
| | - Zaza Iakobishvili
- Department of Community Cardiology, Clalit Health Fund, and Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC)University Clinic WürzburgWürzburgGermany
| | - Radek Pudil
- 1st Department of Medicine–Cardioangiology, Faculty of MedicineUniversity HospitalHradec KrálovéCzech Republic
| | - Hadi Skouri
- Department of Internal MedicineAmerican University of Beirut Medical CenterBeirutLebanon
| | - Alain Cohen‐Solal
- Department of Cardiology, Lariboisière Hospital and U942 INSERM, BIOCANVAS (Biomarqueurs Cardiovasculaires)Paris UniversityParisFrance
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Sciences (CIRCET)Federico II UniversityNaplesItaly
| | | | - Petar M. Seferović
- Faculty of Medicine and Heart Failure CenterBelgrade University Medical Center, University of BelgradeBelgradeSerbia
| | | | | |
Collapse
|
88
|
Tomasoni D, Adamo M, Lombardi CM, Metra M. Highlights in heart failure. ESC Heart Fail 2019; 6:1105-1127. [PMID: 31997538 PMCID: PMC6989277 DOI: 10.1002/ehf2.12555] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains a major cause of mortality, morbidity, and poor quality of life. It is an area of active research. This article is aimed to give an update on recent advances in all aspects of this syndrome. Major changes occurred in drug treatment of HF with reduced ejection fraction (HFrEF). Sacubitril/valsartan is indicated as a substitute to ACEi/ARBs after PARADIGM-HF (hazard ratio [HR], 0.80; 95% confidence interval [CI], 0.73 to 0.87 for sacubitril/valsartan vs. enalapril for the primary endpoint and Wei, Lin and Weissfeld HR 0.79, 95% CI 0.71-0.89 for recurrent events). Its initiation was then shown as safe and potentially useful in recent studies in patients hospitalized for acute HF. More recently, dapagliflozin and prevention of adverse-outcomes in DAPA-HF trial showed the beneficial effects of the sodium-glucose transporter type 2 inhibitor dapaglifozin vs. placebo, added to optimal standard therapy [HR, 0.74; 95% CI, 0.65 to 0.85;0.74; 95% CI, 0.65 to 0.85 for the primary endpoint]. Trials with other SGLT 2 inhibitors and in other patients, such as those with HF with preserved ejection fraction (HFpEF) or with recent decompensation, are ongoing. Multiple studies showed the unfavourable prognostic significance of abnormalities in serum potassium levels. Potassium lowering agents may allow initiation and titration of mineralocorticoid antagonists in a larger proportion of patients. Meta-analyses suggest better outcomes with ferric carboxymaltose in patients with iron deficiency. Drugs effective in HFrEF may be useful also in HF with mid-range ejection fraction. Better diagnosis and phenotype characterization seem warranted in HF with preserved ejection fraction. These and other burning aspects of HF research are summarized and reviewed in this article.
Collapse
Affiliation(s)
- Daniela Tomasoni
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public HealthUniversity of BresciaCardiothoracic DepartmentCivil HospitalsBresciaItaly
| | - Marianna Adamo
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public HealthUniversity of BresciaCardiothoracic DepartmentCivil HospitalsBresciaItaly
| | - Carlo Mario Lombardi
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public HealthUniversity of BresciaCardiothoracic DepartmentCivil HospitalsBresciaItaly
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public HealthUniversity of BresciaCardiothoracic DepartmentCivil HospitalsBresciaItaly
| |
Collapse
|
89
|
Renal denervation in patients with symptomatic chronic heart failure despite resynchronization therapy - a pilot study. POSTEPY W KARDIOLOGII INTERWENCYJNEJ = ADVANCES IN INTERVENTIONAL CARDIOLOGY 2019; 15:240-246. [PMID: 31497058 PMCID: PMC6727225 DOI: 10.5114/aic.2019.8601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/14/2019] [Indexed: 01/01/2023]
Abstract
Introduction Renal denervation (RD) has been shown to decrease sympathetic function in patients with hypertension. Its efficacy in symptomatic chronic heart failure (CHF) patients not responding to cardiac resynchronization therapy (CRT) has not been evaluated. Aim To assess whether a less invasive treatment method – renal denervation – is safe in symptomatic heart failure patients despite optimal medical treatment and resynchronization therapy and whether it is associated with an improvement in clinical status, exercise capacity and hemodynamic parameters. Material and methods The study was an open-label, randomized, controlled clinical trial. Patients were divided into an intervention (RD) and a control group. Clinical data collection, blood pressure (BP) measurements, echocardiography, 6-minute walk test (6MWT) and laboratory tests were performed before, 6 and 12 months after RD. The patients were followed-up to 24 months. Results We included 20 patients aged 52.0 to 86.0 years (median age: 71.5 years), 15 males and 5 females with median left ventricular ejection fraction (LVEF) of 32.5%, body mass index 31.3 kg/m2. Renal denervation was safe, no significant adverse effects were registered. There were no significant differences in LVEF, BP, 6MWT and N-terminal prohormone of brain natriuretic peptide (NT-proBNP) concentration 6 and 12 months after RD or control. Conclusions Our results indicate that RD in CHF patients not responding to CRT is safe and does not worsen exercise capacity and hemodynamic parameters.
Collapse
|
90
|
Yogasundaram H, Chappell MC, Braam B, Oudit GY. Cardiorenal Syndrome and Heart Failure-Challenges and Opportunities. Can J Cardiol 2019; 35:1208-1219. [PMID: 31300181 PMCID: PMC9257995 DOI: 10.1016/j.cjca.2019.04.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/23/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiorenal syndromes (CRS) describe concomitant bidirectional dysfunction of the heart and kidneys in which 1 organ initiates, perpetuates, and/or accelerates decline of the other. CRS are common in heart failure and universally portend worsened prognosis. Despite this heavy disease burden, the appropriate diagnosis and classification of CRS remains problematic. In addition to the hemodynamic drivers of decreased renal perfusion and increased renal vein pressure, induction of the renin-angiotensin-aldosterone system, stimulation of the sympathetic nervous system, disruption of balance between nitric oxide and reactive oxygen species, and inflammation are implicated in the pathogenesis of CRS. Medical therapy of heart failure including renin-angiotensin-aldosterone system inhibition and β-adrenergic blockade can blunt these deleterious processes. Renovascular disease can accelerate the progression of CRS. Volume overload and diuretic resistance are common and complicate the management of CRS. In heart failure and CRS being treated with diuretics, worsening creatinine is not associated with worsened outcome if clinical decongestion is achieved. Adjunctive therapy is often required in the management of volume overload in CRS, but evidence for these therapies is limited. Anemia and iron deficiency are importantly associated with CRS and might amplify decline of cardiac and renal function. End-stage cardiac and/or renal disease represents an especially poor prognosis with limited therapeutic options. Overall, worsening renal function is associated with significantly increased mortality. Despite progress in the area of CRS, there are still multiple pathophysiological and clinical aspects of CRS that need further research to eventually develop effective therapeutic options.
Collapse
Affiliation(s)
- Haran Yogasundaram
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Mark C Chappell
- Department of Surgery/Hypertension and Vascular Research, Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Branko Braam
- Division of Nephrology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
91
|
Coats AJS. Activities of the study groups - the Study Group on Takotsubo Syndrome. Eur J Heart Fail 2019; 21:1054-1056. [PMID: 31414523 DOI: 10.1002/ejhf.1583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 07/16/2019] [Indexed: 11/10/2022] Open
|
92
|
Notarius CF, Millar PJ, Keir DA, Murai H, Haruki N, O'Donnell E, Marzolini S, Oh P, Floras JS. Training heart failure patients with reduced ejection fraction attenuates muscle sympathetic nerve activation during mild dynamic exercise. Am J Physiol Regul Integr Comp Physiol 2019; 317:R503-R512. [PMID: 31365304 DOI: 10.1152/ajpregu.00104.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Muscle sympathetic nerve activity (MSNA) decreases during low-intensity dynamic one-leg exercise in healthy subjects but increases in patients with heart failure with reduced ejection fraction (HFrEF). We hypothesized that increased peak oxygen uptake (V̇o2peak) after aerobic training would be accompanied by less sympathoexcitation during both mild and moderate one-leg dynamic cycling, an attenuated muscle metaboreflex, and greater skin vasodilation. We studied 27 stable, treated HFrEF patients (6 women; mean age: 65 ± 2 SE yr; mean left ventricular ejection fraction: 30 ± 1%) and 18 healthy age-matched volunteers (6 women; mean age: 57 ± 2 yr). We assessed V̇o2peak (open-circuit spirometry) and the skin microcirculatory response to reactive hyperemia (laser flowmetry). Fibular MSNA (microneurography) was recorded before and during one-leg cycling (2 min unloaded and 2 min at 50% of V̇o2peak) and, to assess the muscle metaboreflex, during posthandgrip ischemia (PHGI). HFrEF patients were evaluated before and after 6 mo of exercise-based cardiac rehabilitation. Pretraining V̇o2peak and skin vasodilatation were lower (P < 0.001) and resting MSNA higher (P = 0.01) in HFrEF than control subjects. Training improved V̇o2peak (+3.0 ± 1.0 mL·kg-1·min-1; P < 0.001) and cutaneous vasodilation and diminished resting MSNA (-6.0 ± 2.0, P = 0.01) plus exercise MSNA during unloaded (-4.0 ± 2.5, P = 0.04) but not loaded cycling (-1.0 ± 4.0 bursts/min, P = 0.34) and MSNA during PHGI (P < 0.05). In HFrEF patients, exercise training lowers MSNA at rest, desensitizes the sympathoexcitatory metaboreflex, and diminishes MSNA elicited by mild but not moderate cycling. Training-induced downregulation of resting MSNA and attenuated reflex sympathetic excitation may improve exercise capacity and survival.
Collapse
Affiliation(s)
- Catherine F Notarius
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Philip J Millar
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Human Health and Nutritional Sciences, University of Guelph, Ontario, Canada
| | - Daniel A Keir
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Hisayoshi Murai
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Nobuhiko Haruki
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Emma O'Donnell
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.,School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Susan Marzolini
- Cardiovascular Prevention and Rehabilitation Program, Toronto Rehabilitation Institute, University Health Network, Toronto, Ontario, Canada
| | - Paul Oh
- Cardiovascular Prevention and Rehabilitation Program, Toronto Rehabilitation Institute, University Health Network, Toronto, Ontario, Canada
| | - John S Floras
- Division of Cardiology, University Health Network and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
93
|
Device therapy in heart failure with reduced ejection fraction-cardiac resynchronization therapy and more. Herz 2019; 43:415-422. [PMID: 29744528 DOI: 10.1007/s00059-018-4710-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In patients with heart failure with reduced ejection fraction (HFrEF), optimal medical treatment includes beta-blockers, ACE inhibitors/angiotensinreceptor-neprilysin inhibitors (ARNI), mineralocorticoid receptor antagonists, and ivabradine when indicated. In device therapy of HFrEF, implantable cardioverter-defibrillators and cardiac resynchronization therapy (CRT) have been established for many years. CRT is the therapy of choice (class I indication) in symptomatic patients with HFrEF and a broad QRS complex with a left bundle branch block (LBBB) morphology. However, the vast majority of heart failure patients show a narrow QRS complex or a non-LBBB morphology. These patients are not candidates for CRT and alternative electrical therapies such as baroreflex activation therapy (BAT) and cardiac contractility modulation (CCM) may be considered. BAT modulates vegetative dysregulation in heart failure. CCM improves contractility, functional capacity, and symptoms. Although a broad data set is available for BAT and CCM, mortality data are still lacking for both methods. This article provides an overview of the device-based therapeutic options for patients with HFrEF.
Collapse
|
94
|
|
95
|
Keir DA, Duffin J, Millar PJ, Floras JS. Simultaneous assessment of central and peripheral chemoreflex regulation of muscle sympathetic nerve activity and ventilation in healthy young men. J Physiol 2019; 597:3281-3296. [DOI: 10.1113/jp277691] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/13/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Daniel A. Keir
- University Health Network and Mount Sinai Hospital Division of CardiologyDepartment of Medicine, University of Toronto Toronto Ontario Canada
| | - James Duffin
- Departments of Anaesthesia and PhysiologyUniversity of Toronto Toronto Ontario Canada
- Thornhill Research Inc. Toronto Ontario Canada
| | - Philip J. Millar
- University Health Network and Mount Sinai Hospital Division of CardiologyDepartment of Medicine, University of Toronto Toronto Ontario Canada
- Human Health and Nutritional ScienceUniversity of Guelph Guelph Ontario Canada
| | - John S. Floras
- University Health Network and Mount Sinai Hospital Division of CardiologyDepartment of Medicine, University of Toronto Toronto Ontario Canada
| |
Collapse
|
96
|
Coats AJS. Figures of the Heart Failure Association: Professor Dr. med. Johann Bauersachs, Chair of the Clinical Science Section. Eur J Heart Fail 2019; 21:545-548. [PMID: 31069912 DOI: 10.1002/ejhf.1484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 11/05/2022] Open
|
97
|
Inspiratory- and expiratory-gated transcutaneous vagus nerve stimulation have different effects on heart rate in healthy subjects: preliminary results. Clin Auton Res 2019; 31:205-214. [PMID: 30941526 PMCID: PMC8041682 DOI: 10.1007/s10286-019-00604-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022]
Abstract
Purpose Transcutaneous auricular vagus nerve stimulation (taVNS) has been considered for the treatment of sympathetically mediated disorders. However, the optimal mode of stimulation is unknown. This study aimed to compare the cardiovascular effects of respiratory-gated taVNS in healthy subjects. Methods The examination included expiratory-gated, inspiratory-gated, and non-respiratory-gated taVNS trials. Subjects were examined twice (the order of expiratory- and inspiratory-gated taVNS was changed). taVNS trials started with controlled breathing without stimulation (pre-stimulatory recording) followed by controlled breathing with taVNS (stimulatory recording). Synchronizing taVNS with the respiratory phase was computer-controlled. Heart rate (HR) was calculated from ECG. Systolic blood pressure (SBP) and systemic vascular resistance (SVR) were recorded continuously and noninvasively. Baroreflex sensitivity based on rising (BRS-UP) or falling SBP sequences (BRS-DOWN) or all sequences (BRS-ALL) and heart rate variability (HRV) were analyzed. Results Seventy-two taVNS trials were obtained from 12 subjects (age 23 ± 3 years). Pre-stimulatory HR correlated with change in HR (r = − 0.25) and SVR (r = 0.24, both p < 0.05). There were no differences between three stimulatory conditions in (1) the changes of hemodynamic parameters, (2) BRS-UP and BRS-ALL, or (3) HRV indices (all p > 0.20). However, in the group of high pre-stimulatory HR trials, HR change differed between inspiratory-gated (0.11 ± 0.53%) and both expiratory-gated (− 1.30 ± 0.58%, p = 0.06) and non-respiratory-gated taVNS (− 1.69 ± 0.65, p = 0.02). BRS-DOWN was higher in inspiratory- vs. non-respiratory-gated taVNS (15.4 ± 1.3 vs. 14.1 ± 0.9 ms/mmHg, p = 0.03). Conclusions Expiratory-gated and non-respiratory-gated taVNS exert clear cardioinhibitory effects in healthy subjects with high pre-stimulatory HR, whereas inspiratory-gated taVNS does not affect HR. Cardiac and vascular effects of taVNS depend on pre-stimulatory HR.
Collapse
|
98
|
Coats AJ. Heart Failure Atlas, a major project of the Heart Failure Association in concert with the European Society of Cardiology. Eur J Heart Fail 2019; 21:395-397. [DOI: 10.1002/ejhf.1445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 11/11/2022] Open
|
99
|
Caru M, Corbin D, Périé D, Lemay V, Delfrate J, Drouin S, Bertout L, Krajinovic M, Laverdière C, Andelfinger G, Sinnett D, Curnier D. Doxorubicin treatments induce significant changes on the cardiac autonomic nervous system in childhood acute lymphoblastic leukemia long-term survivors. Clin Res Cardiol 2019; 108:1000-1008. [DOI: 10.1007/s00392-019-01427-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022]
|
100
|
Manolis AS, Manolis TA, Manolis AA, Melita H. Neprilysin Inhibitors: Filling a Gap in Heart Failure Management, Albeit Amidst Controversy and at a Significant Cost. Am J Cardiovasc Drugs 2019; 19:21-36. [PMID: 29926350 DOI: 10.1007/s40256-018-0289-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dual angiotensin and neprilysin inhibition using the combination drug sacubitril-valsartan has ushered in a new era in the treatment of heart failure (HF). The randomized controlled PARADIGM-HF trial, which randomized 8399 patients with HF to enalapril or sacubitril-valsartan, showed a 20% reduction in mortality and HF hospitalization with the new drug. This has been heralded as a step toward filling a crucial gap in HF management by providing strong evidence that combined inhibition of the angiotensin receptor and neprilysin is superior to inhibition of the renin-angiotensin system alone in stable patients with chronic HF as it negates the deleterious effects of angiotensin while concomitantly augmenting the beneficial effects of the endogenous natriuretic peptide system. This new therapy is costly, and other confirmatory studies have been lacking for over 2 years since its approval by major regulatory authorities. As such, controversy and heated discussions have amassed, as has detailed information from a plethora of secondary analyses of this pivotal trial about the pros and cons of this promising new therapeutic strategy in HF management. The aim of this review was to provide a critical assessment of all these aspects.
Collapse
Affiliation(s)
- Antonis S Manolis
- Third Department of Cardiology, Athens University School of Medicine, Vas. Sofias 114, 115 27, Athens, Greece.
| | | | | | | |
Collapse
|