101
|
Lerner Y, Hanout W, Ben-Uliel SF, Gani S, Leshem MP, Qvit N. Natriuretic Peptides as the Basis of Peptide Drug Discovery for Cardiovascular Diseases. Curr Top Med Chem 2020; 20:2904-2921. [PMID: 33050863 DOI: 10.2174/1568026620666201013154326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 01/14/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading global cause of death, accounting for more than 17.6 million deaths per year in 2016, a number that is expected to grow to more than 23.6 million by 2030. While many technologies are currently under investigation to improve the therapeutic outcome of CVD complications, only a few medications have been approved. Therefore, new approaches to treat CVD are urgently required. Peptides regulate numerous physiological processes, mainly by binding to specific receptors and inducing a series of signals, neurotransmissions or the release of growth factors. Importantly, peptides have also been shown to play an important role in the circulatory system both in physiological and pathological conditions. Peptides, such as angiotensin II, endothelin, urotensin-II, urocortins, adrenomedullin and natriuretic peptides have been implicated in the control of vascular tone and blood pressure as well as in CVDs such as congestive heart failure, atherosclerosis, coronary artery disease, and pulmonary and systemic hypertension. Hence it is not surprising that peptides are becoming important therapeutic leads in CVDs. This article will review the current knowledge on peptides and their role in the circulatory system, focusing on the physiological roles of natriuretic peptides in the cardiovascular system and their implications in CVDs.
Collapse
Affiliation(s)
- Yana Lerner
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed, Israel
| | - Wessal Hanout
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed, Israel
| | - Shulamit Fluss Ben-Uliel
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed, Israel
| | - Samar Gani
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed, Israel
| | - Michal Pellach Leshem
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed, Israel
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed, Israel
| |
Collapse
|
102
|
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: 12.5] [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
|
103
|
Pazos F. Range of adiposity and cardiorenal syndrome. World J Diabetes 2020; 11:322-350. [PMID: 32864046 PMCID: PMC7438185 DOI: 10.4239/wjd.v11.i8.322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/27/2020] [Accepted: 06/14/2020] [Indexed: 02/05/2023] Open
Abstract
Obesity and obesity-related co-morbidities, diabetes mellitus, and hypertension are among the fastest-growing risk factors of heart failure and kidney disease worldwide. Obesity, which is not a unitary concept, or a static process, ranges from alterations in distribution to the amount of adiposity. Visceral adiposity, which includes intraabdominal visceral fat mass and ectopic fat deposition such as hepatic, cardiac, or renal, was robustly associated with a greater risk for cardiorenal morbidity than subcutaneous adiposity. In addition, morbid obesity has also demonstrated a negative effect on cardiac and renal functioning. The mechanisms by which adipose tissue is linked with the cardiorenal syndrome (CRS) are hemodynamic and mechanical changes, as well neurohumoral pathways such as insulin resistance, endothelial dysfunction, nitric oxide bioavailability, renin-angiotensin-aldosterone, oxidative stress, sympathetic nervous systems, natriuretic peptides, adipokines and inflammation. Adiposity and other associated co-morbidities induce adverse cardiac remodeling and interstitial fibrosis. Heart failure with preserved ejection fraction has been associated with obesity-related functional and structural abnormalities. Obesity might also impair kidney function through hyperfiltration, increased glomerular capillary wall tension, and podocyte dysfunction, which leads to tubulointerstitial fibrosis and loss of nephrons and, finally, chronic kidney disease. The development of new treatments with renal and cardiac effects in the context of type 2 diabetes, which improves mortality outcome, has highlighted the importance of CRS and its prevalence. Increased body fat triggers cellular, neuro-humoral and metabolic pathways, which create a phenotype of the CRS with specific cellular and biochemical biomarkers. Obesity has become a single cardiorenal umbrella or type of cardiorenal metabolic syndrome. This review article provides a clinical overview of the available data on the relationship between a range of adiposity and CRS, the support for obesity as a single cardiorenal umbrella, and the most relevant studies on the recent therapeutic approaches.
Collapse
Affiliation(s)
- Fernando Pazos
- Department of Medicine, Medicine Faculty, Cantabria University, Valdecilla Hospital, Santander 39080, Cantabria, Spain
| |
Collapse
|
104
|
Mohammed El Tabaa M, Mohammed El Tabaa M. Targeting Neprilysin (NEP) pathways: A potential new hope to defeat COVID-19 ghost. Biochem Pharmacol 2020; 178:114057. [PMID: 32470547 PMCID: PMC7250789 DOI: 10.1016/j.bcp.2020.114057] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 is an ongoing viral pandemic disease that is caused by SARS-CoV2, inducing severe pneumonia in humans. However, several classes of repurposed drugs have been recommended, no specific vaccines or effective therapeutic interventions for COVID-19 are developed till now. Viral dependence on ACE-2, as entry receptors, drove the researchers into RAS impact on COVID-19 pathogenesis. Several evidences have pointed at Neprilysin (NEP) as one of pulmonary RAS components. Considering the protective effect of NEP against pulmonary inflammatory reactions and fibrosis, it is suggested to direct the future efforts towards its potential role in COVID-19 pathophysiology. Thus, the review aimed to shed light on the potential beneficial effects of NEP pathways as a novel target for COVID-19 therapy by summarizing its possible molecular mechanisms. Additional experimental and clinical studies explaining more the relationships between NEP and COVID-19 will greatly benefit in designing the future treatment approaches.
Collapse
Affiliation(s)
- Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute, University of Sadat City, Egypt.
| | | |
Collapse
|
105
|
Geng Q, Yan R, Wang Z, Hou F. Effects of LCZ696 (Sacubitril/Valsartan) on Blood Pressure in Patients with Hypertension: A Meta-Analysis of Randomized Controlled Trials. Cardiology 2020; 145:589-598. [PMID: 32726791 DOI: 10.1159/000507327] [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] [Received: 12/05/2019] [Accepted: 03/14/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND LCZ696 (sacubitril/valsartan), a first-in-class angiotensin receptor-neprilysin inhibitor, can significantly reduce blood pressure in patients with heart failure. We performed this meta-analysis to determine the antihypertensive effect of LCZ696 in patients with hypertension. METHODS Randomized controlled trials were searched in MEDLINE, the Cochrane Library, and Clinicaltrials.gov. Twelve studies with a total of 6,064 participants were included. RESULTS Compared with angiotensin receptor blockers (ARBs), LCZ696 100 mg caused a significant reduction in systolic blood pressure (SBP) (mean difference [MD] -1.58 mm Hg, 95% confidence interval [CI] -2.09 to -1.07, p < 0.05) and diastolic blood pressure (DBP) (MD -0.66 mm Hg, 95% CI -0.98 to -0.33, p < 0.05). LCZ696 200 mg caused a significant reduction in SBP (MD -4.94 mm Hg, 95% CI -6.54 to -3.35, p < 0.05), DBP (MD -2.24 mm Hg, 95% CI -2.74 to -1.75, p < 0.05), 24-h ambulatory SBP (24 h ASBP; MD -3.69 mm Hg, 95% CI -4.80 to -2.58, p < 0.05), and 24-h ADBP (MD -1.71 mm Hg, 95% CI -2.13 to -1.28, p < 0.05). LCZ696 400 mg caused a significant reduction in SBP (MD -6.25 mm Hg, 95% CI -7.90 to -4.61, p < 0.05), DBP (MD -2.30 mm Hg, 95% CI -2.80 to -1.80, p < 0.05), 24-h ASBP (MD -4.31 mm Hg, 95% CI -6.56 to -2.07, p < 0.05), and 24 h ADBP (MD -1.69 mm Hg, 95% CI -2.59 to -0.79, p < 0.05). Compared with LCZ696 200 mg, LCZ696 400 mg caused a significant reduction in SBP (MD 1.71 mm Hg, 95% CI 1.15 to 2.27, p < 0.05), DBP (MD 0.90 mm Hg, 95% CI 0.65 to 1.16, p < 0.05), 24-h ASBP (MD 1.50 mm Hg, 95% CI 0.84 to 2.17, p < 0.05), and 24-h ADBP (MD 0.76 mm Hg, 95% CI 0.47 to 1.06, p < 0.05). CONCLUSIONS The blood pressure-lowering effect of LCZ696 is dose-related. This meta-analysis confirms the antihypertensive effects of LCZ696.
Collapse
Affiliation(s)
- Qiang Geng
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Rongqiang Yan
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Zhengzhong Wang
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Fangjie Hou
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China,
| |
Collapse
|
106
|
Grubić Rotkvić P, Cigrovski Berković M, Bulj N, Rotkvić L, Ćelap I. Sodium-glucose cotransporter 2 inhibitors’ mechanisms of action in heart failure. World J Diabetes 2020; 11:269-279. [PMID: 32843930 PMCID: PMC7415232 DOI: 10.4239/wjd.v11.i7.269] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Three major cardiovascular outcome trials (CVOTs) with a new class of antidiabetic drugs - sodium-glucose cotransporter 2 (SGLT2) inhibitors (EMPA-REG OUTCOME trial with empagliflozin, CANVAS Program with canagliflozin, DECLARE-TIMI 58 with dapagliflozin) unexpectedly showed that cardiovascular outcomes could be improved possibly due to a reduction in heart failure risk, which seems to be the most sensitive outcome of SGLT2 inhibition. No other CVOT to date has shown any significant benefit on heart failure events. Even more impressive findings came recently from the DAPA-HF trial in patients with confirmed and well-treated heart failure: Dapagliflozin was shown to reduce heart failure risk for patients with heart failure with reduced ejection fraction regardless of diabetes status. Nevertheless, despite their possible wide clinical implications, there is much doubt about the mechanisms of action and a lot of questions to unravel, especially now when their benefits translated to non-diabetic patients, rising doubts about the validity of some current mechanistic assumptions.The time frame of their cardiovascular benefits excludes glucose-lowering and antiatherosclerotic-mediated effects and multiple other mechanisms, direct cardiac as well as systemic, are suggested to explain their early cardiorenal benefits. These are: Anti-inflammatory, antifibrotic, antioxidative, antiapoptotic properties, then renoprotective and hemodynamic effects, attenuation of glucotoxicity, reduction of uric acid levels and epicardial adipose tissue, modification of neurohumoral system and cardiac fuel energetics, sodium-hydrogen exchange inhibition. The most logic explanation seems that SGLT2 inhibitors timely target various mechanisms underpinning heart failure pathogenesis. All the proposed mechanisms of their action could interfere with evolution of heart failure and are discussed separately within the main text.
Collapse
Affiliation(s)
| | - Maja Cigrovski Berković
- Department of Endocrinology, Diabetes, Metabolism and Clinical Pharmacology, University Hospital, Zagreb 10000, Croatia
- Department for Medicine of Sports and Exercise, Faculty of Kinesiology University of Zagreb, Zagreb 10000, Croatia
| | - Nikola Bulj
- Department of Cardiology, University Hospital Centre, Zagreb 10000, Croatia
| | - Luka Rotkvić
- Department of Cardiology, Magdalena Clinic for Cardiovascular Disease, Krapinske Toplice 49217, Croatia
| | - Ivana Ćelap
- Department of Clinical Chemistry, University Hospital Centre, Zagreb 10000, Croatia
| |
Collapse
|
107
|
Oliver-McNeil S, Bowers M, LaRue SJ, Vader J, DeVore AD, Granger BB. Benefits of Optimizing Heart Failure Medication Dosage. J Nurse Pract 2020. [DOI: 10.1016/j.nurpra.2020.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
108
|
Popovic D, Djordjevic T, Jakovljevic D, Ristic A, Lasica R, Arena R, Guazzi M. N-Terminal-pro-Brain natriuretic peptide dynamics during effort phenotypes ischemic heart failure and determines prognosis regardless of ejection fraction. Peptides 2020; 129:170315. [PMID: 32333999 DOI: 10.1016/j.peptides.2020.170315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/07/2020] [Accepted: 04/12/2020] [Indexed: 11/20/2022]
Abstract
Ischemic heart disease leading to heart failure (HF) portends a high overall morbidity and mortality. A higher N-terminal-pro-B-type natriuretic peptide (NT-pro-BNP) at rest reflects HF severity and impaired cardiac output, most often secondary to reduced ejection fraction (EF). As an insufficient increase in cardiac output during exertion is common in all HF phenotypes, we examined the value of NT-pro-BNP during exercise testing as a risk stratification index for ischemic HF secondary to either reduced (HFrEF) or mid-ranged/preserved EF (HFmrEF/HFpEF). 213 patients (123 HFrEF; 90 HFmrEF/HFpEF) underwent cardiopulmonary exercise testing (CPET). NT-pro-BNP was determined at rest and peak exercise. The distribution of HFrEF and HFmrEF/HFpEF etiology in subjects with and without oxygen consumption trajectory flattening during CPET was similar (p > 0.05). Patients with HFrEF had higher plasma levels of NT-pro-BNP at rest and peak exercise than those with HFmrEF/HFpEF (984 vs. 780; 1012 vs. 845 pg/mL, p < 0.01, respectively), whereas ΔNT-pro-BNPpeak/rest was similar (60 vs. 50 pg/mL, p > 0.05). During the tracking period (22.4 ± 20.3 months) 34 patients died, and there were 2 cardiac transplantations and 3 LVAD implantations. In a multivariate regression model only the NT-pro-BNPpeak and ΔNT-pro-BNPpeak/rest were retained in the regression for the prediction of adverse events (Chi-square:8.97, p = 0.003). ROC analysis demonstrated that NT-pro-BNPpeak ≥1506 pg/mL and ΔNT-pro-BNPpeak/rest ≥108 pg/mL were optimal for identifying patients with a risk (Sn = 76.9, 74.4 %; Sp = 84.7, 80.9 %, respectively). NT-pro-BNP changes during effort and absolute peak values reached provide novel insights emerging as new and strong predictors of adverse events in HF of any EF.
Collapse
Affiliation(s)
- Dejana Popovic
- Clinic for Cardiology, Clinical Center Serbia, University of Belgrade, Belgrade, Serbia.
| | - Tea Djordjevic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Djodje Jakovljevic
- Cardiovascular Research Centre, Institute of Cellular Medicine, Medical School, Newcastle University & Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE24HH, United Kingdom
| | - Arsen Ristic
- Clinic for Cardiology, Clinical Center Serbia, University of Belgrade, Belgrade, Serbia
| | - Ratko Lasica
- Clinic for Cardiology, Clinical Center Serbia, University of Belgrade, Belgrade, Serbia
| | - Ross Arena
- Department of Physical Therapy, College of Applied Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Marco Guazzi
- Heart Failure Unit and Cardiopulmonary Laboratory, University Cardiology Department, I.R.C.C.S., Policlinico San Donato University Hospital, Milan, Italy
| |
Collapse
|
109
|
Adverse Cardiac Remodelling after Acute Myocardial Infarction: Old and New Biomarkers. DISEASE MARKERS 2020; 2020:1215802. [PMID: 32626540 PMCID: PMC7306098 DOI: 10.1155/2020/1215802] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 01/06/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022]
Abstract
The prevalence of heart failure (HF) due to cardiac remodelling after acute myocardial infarction (AMI) does not decrease regardless of implementation of new technologies supporting opening culprit coronary artery and solving of ischemia-relating stenosis with primary percutaneous coronary intervention (PCI). Numerous studies have examined the diagnostic and prognostic potencies of circulating cardiac biomarkers in acute coronary syndrome/AMI and heart failure after AMI, and even fewer have depicted the utility of biomarkers in AMI patients undergoing primary PCI. Although complete revascularization at early period of acute coronary syndrome/AMI is an established factor for improved short-term and long-term prognosis and lowered risk of cardiovascular (CV) complications, late adverse cardiac remodelling may be a major risk factor for one-year mortality and postponded heart failure manifestation after PCI with subsequent blood flow resolving in culprit coronary artery. The aim of the review was to focus an attention on circulating biomarker as a promising tool to stratify AMI patients at high risk of poor cardiac recovery and developing HF after successful PCI. The main consideration affects biomarkers of inflammation, biomechanical myocardial stress, cardiac injury and necrosis, fibrosis, endothelial dysfunction, and vascular reparation. Clinical utilities and predictive modalities of natriuretic peptides, cardiac troponins, galectin 3, soluble suppressor tumorogenicity-2, high-sensitive C-reactive protein, growth differential factor-15, midregional proadrenomedullin, noncoding RNAs, and other biomarkers for adverse cardiac remodelling are discussed in the review.
Collapse
|
110
|
Salazar J, Rojas-Quintero J, Cano C, Pérez JL, Ramírez P, Carrasquero R, Torres W, Espinoza C, Chacín-González M, Bermúdez V. Neprilysin: A Potential Therapeutic Target of Arterial Hypertension? Curr Cardiol Rev 2020; 16:25-35. [PMID: 31241018 PMCID: PMC7062041 DOI: 10.2174/1573403x15666190625160352] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 02/07/2023] Open
Abstract
Arterial hypertension is the most prevalent chronic disease in the adult population of developed countries and it constitutes a significant risk factor in the development of cardiovascular disease, contributing to the emergence of many comorbidities, among which heart failure excels, a clinical syndrome that nowadays represents a major health problem with uncountable hospitalizations and the indolent course of which progressively worsens until quality of life decreases and lastly death occurs prematurely. In the light of this growing menace, each day more efforts are invested in the field of cardiovascular pharmacology, searching for new therapeutic options that allow us to modulate the physiological systems that appear among these pathologies. Therefore, in the later years, the study of natriuretic peptides has become so relevant, which mediate beneficial effects at the cardiovascular level such as diuresis, natriuresis, and decreasing cardiac remodeling; their metabolism is mediated by neprilysin, a metalloproteinase, widely expressed in the human and capable of catalyzing many substrates. The modulation of these functions has been studied by decades, giving room to Sacubitril, the first neprilysin inhibitor, which in conjunction with an angiotensin receptor blocker has provided a high efficacy and tolerability among patients with heart failure, for whom it has already been approved and recommended. Nonetheless, in the matter of arterial hypertension, significant findings have arisen that demonstrate the potential role that it will play among the pharmacological alternatives in the upcoming years.
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Joselyn Rojas-Quintero
- Pulmonary and Critical Care Medicine Department, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Clímaco Cano
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - José L Pérez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Paola Ramírez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Rubén Carrasquero
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Wheeler Torres
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | | | | | - Valmore Bermúdez
- Universidad Simon Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| |
Collapse
|
111
|
Effects of Low-Dose Sacubitril/Valsartan on Different Stages of Cardiac Hypertrophy in Salt-Loaded Hypertensive Rats. J Cardiovasc Pharmacol 2020; 73:282-289. [PMID: 30829732 DOI: 10.1097/fjc.0000000000000662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sacubitril/valsartan was shown to attenuate the development of cardiac hypertrophy with enhanced blood pressure reduction compared with valsartan alone in animal models. We investigated whether a low-dose sacubitril/valsartan has blood pressure-independent effects on cardiac hypertrophy and pulmonary edema using a rat model of hypertension and obesity. METHODS AND RESULTS In plan 1, male SHR/NDmcr-cp rats fed normal or phase-increased high salt were treated with vehicle, 6-mg/kg sacubitril/valsartan or 3-mg/kg valsartan, for 6 months. In plan 2, after high-salt loading for 6 months, drugs were administered for 4 months. Antihypertensive effects of the 2 drugs were similar during all study periods. In plan 1 with normal salt, there were no differences between treatments in the left ventricle weight/body weight (BW), or lung weight/BW as an index of cardiac hypertrophy or pulmonary edema, respectively. These indexes were smaller in high-salt-fed rats with sacubitril/valsartan than vehicle. In plan 2, both indexes did not differ between vehicle and sacubitril/valsartan. Ventricle weight/BW was lower in valsartan than sacubitril/valsartan. In plan 2, gene markers of cardiac dysfunction were upregulated by sacubitril/valsartan compared with the other groups. CONCLUSIONS Low-dose sacubitril/valsartan may have different effects depending on the stage of cardiac hypertrophy in rats.
Collapse
|
112
|
Xu D, Ma M, Xu Y, Su Y, Ong SB, Hu X, Chai M, Zhao M, Li H, Chen Y, Xu X. Single-cell Transcriptome Analysis Indicates New Potential Regulation Mechanism of ACE2 and NPs signaling among heart failure patients infected with SARS-CoV-2. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.04.30.20081257. [PMID: 32511460 PMCID: PMC7241106 DOI: 10.1101/2020.04.30.20081257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND COVID-19 patients with comorbidities such as hypertension or heart failure (HF) are associated with poor clinical outcomes. Angiotensin-converting enzyme 2 (ACE2), the critical enzyme for SARS-CoV-2 infection, is broadly expressed in many organs including heart. However, the cellular distribution of ACE2 in the human heart, particularly the failing heart is unknown. METHODS We analyzed single-cell RNA sequencing (scRNA-seq) data in both normal and failing hearts, and characterized the ACE2 gene expression profile in various cell subsets, especially in cardiomyocyte subsets, as well as its interaction with gene networks relating to various defense and immune responses at the single cell level. RESULTS The results demonstrated that ACE2 is present in cardiomyocytes (CMs), endothelial cells, fibroblasts and smooth muscle cells in the heart, while the number of ACE2-postive (ACE2+) CMs and ACE2 gene expression in these CMs are significantly increased in the failing hearts. Interestingly, both brain natriuretic peptides (BNP) and atrial natriuretic peptide (ANP) are significantly up-regulated in the ACE2+ CMs. Further analysis shows that ANP, BNP and ACE2 may form a negative feedback loop with a group of genes associated with the development of heart failure. To our surprise, we found that genes related to virus entry, virus replication and suppression of interferon-gamma(IFN-γ)signaling are all up-regulated in CMs in failing hearts, and the increases were significantly higher in ACE2+ CMs as compared with ACE2 negative (ACE2-) CMs, suggesting that these ACE2+ CMs may be more vulnerable to virus infection. Since ACE2 expression is correlated with BNP expression, we further performed retrospective analysis of the plasma BNP levels and clinic outcome of 91 COVID-19 patients from a single-center. Patients with higher plasma BNP were associated with significantly higher mortality rate and expression levels of inflammatory and infective markers such as procalcitonin and C-reactive protein. CONCLUSION In the failing heart, the upregulation of ACE2 and virus infection associated genes, as well as the increased expression of ANP and BNP could facilitate SARS-CoV-2 virus entry and replication in these vulnerable cardiomyocyte subsets. These findings may advance our understanding of the underlying molecular mechanisms of myocarditis associated with COVID-19.
Collapse
Affiliation(s)
- Dachun Xu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200072, China
| | - Mengqiu Ma
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200072, China
| | - Yanhua Xu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200072, China
| | - Yang Su
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200072, China
| | - Sang-Bing Ong
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, Chinese University of Hong Kong (CUHK), Hong Kong SAR
- Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Kowloon Bay, Hong Kong SAR
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong SAR
- Institute for Translational Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Xingdong Hu
- Department of Critical Care Medicine, The Third people’s Hospital of Guizhou Province, Guiyang, China
| | - Min Chai
- Department of Critical Care Medicine, Ezhou Central Hospital, Ezhou, China
| | - Maojun Zhao
- Emergency Department, The First People’s Hospital of Guiyang, Guiyang, Guizhou, China
| | - Hong Li
- IID, NIEHS, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics, ESCBL, NIEHS, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
113
|
Harrison TG, Shukalek CB, Hemmelgarn BR, Zarnke KB, Ronksley PE, Iragorri N, Graham MM, James MT. Association of NT-proBNP and BNP With Future Clinical Outcomes in Patients With ESKD: A Systematic Review and Meta-analysis. Am J Kidney Dis 2020; 76:233-247. [PMID: 32387090 DOI: 10.1053/j.ajkd.2019.12.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022]
Abstract
RATIONALE & OBJECTIVE Use of brain natriuretic peptide (BNP) and N-terminal pro-BNP (NT-proBNP) for cardiovascular (CV) risk assessment in patients with end-stage kidney disease (ESKD) remains unclear. We examined the associations between different threshold elevations of these peptide levels and clinical outcomes in patients with ESKD. STUDY DESIGN Systematic review and meta-analysis. SETTING & STUDY POPULATIONS We searched MEDLINE and EMBASE (through September 2019) for observational studies of adults with ESKD (estimated glomerular filtration rate≤15mL/min/1.73m2 or receiving maintenance dialysis). SELECTION CRITERIA FOR STUDIES Studies that reported NT-proBNP or BNP levels and future CV events, CV mortality, or all-cause mortality. DATA EXTRACTION Cohort characteristics and measures of risk associated with study-specified peptide thresholds. ANALYTICAL APPROACH Hazard ratios (HRs) for clinical outcomes associated with different NT-proBNP and BNP ranges were categorized into common thresholds and pooled using random-effects meta-analysis. RESULTS We identified 61 studies for inclusion in our review (19,688 people). 49 provided sufficient detail for inclusion in meta-analysis. Pooled unadjusted HRs for CV mortality were progressively greater for greater thresholds of NT-proBNP, from 1.45 (95% CI, 0.91-2.32) for levels>2,000pg/mL to 5.95 (95% CI, 4.23-8.37) for levels>15,000pg/mL. Risk for all-cause mortality was significantly higher at all NT-proBNP thresholds ranging from> 1,000 to> 20,000pg/mL (HR range, 1.53-4.00). BNP levels>550pg/mL were associated with increased risk for CV mortality (HR, 2.54; 95% CI, 1.49-4.33), while the risks for all-cause mortality were 2.04 (95% CI, 0.82-5.12) at BNP levels>100pg/mL and 2.97 (95% CI, 2.21-3.98) at BNP levels>550pg/mL. Adjusted analyses demonstrated similarly greater risks for CV and all-cause mortality with greater NT-proBNP concentrations. LIMITATIONS Incomplete outcome reporting and risk for outcome reporting bias. Estimation of risk for CV events for specific thresholds of both peptides were limited by poor precision. CONCLUSIONS ESKD-specific NT-proBNP and BNP level thresholds of elevation are associated with increased risk for CV and all-cause mortality. This information may help guide interpretation of NT-proBNP and BNP levels in patients with ESKD.
Collapse
Affiliation(s)
- Tyrone G Harrison
- Departments of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Caley B Shukalek
- Departments of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Brenda R Hemmelgarn
- Departments of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kelly B Zarnke
- Departments of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul E Ronksley
- Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicolas Iragorri
- Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michelle M Graham
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Matthew T James
- Departments of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
114
|
Vlachou M, Kamperidis V, Giannakoulas G, Karamitsos T, Vlachaki E, Karvounis H. Biochemical and imaging markers in patients with thalassaemia. Hellenic J Cardiol 2020; 62:4-12. [PMID: 32387594 DOI: 10.1016/j.hjc.2020.04.012] [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: 12/21/2019] [Revised: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 11/15/2022] Open
Abstract
Beta-thalassaemia is a genetic disease with different clinical aspects, which can lead to heart failure with a multifactorial mechanism. Over the last years, growing interest has been reported for biomarkers that may help in the diagnosis, staging and prognosis of heart disease at an early stage, in patients with beta-thalassaemia. This review will highlight the current clinical value of cardiac biomarkers in patients with beta-thalassaemia and the ongoing research for a possible expanded future use.
Collapse
Affiliation(s)
- Maria Vlachou
- 1(st) Cardiology Department, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Vasileios Kamperidis
- 1(st) Cardiology Department, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece.
| | - George Giannakoulas
- 1(st) Cardiology Department, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Theodoros Karamitsos
- 1(st) Cardiology Department, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Efthymia Vlachaki
- Thalassaemia Unit, Ippokratio University Hospital, Thessaloniki, Greece
| | - Haralambos Karvounis
- 1(st) Cardiology Department, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
115
|
Ostróżka-Cieślik A, Dolińska B. The Role of Hormones and Trophic Factors as Components of Preservation Solutions in Protection of Renal Function before Transplantation: A Review of the Literature. Molecules 2020; 25:E2185. [PMID: 32392782 PMCID: PMC7248710 DOI: 10.3390/molecules25092185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Transplantation is currently a routine method for treating end-stage organ failure. In recent years, there has been some progress in the development of an optimal composition of organ preservation solutions, improving the vital functions of the organ and allowing to extend its storage period until implantation into the recipient. Optimizations are mostly based on commercial solutions, routinely used to store grafts intended for transplantation. The paper reviews hormones with a potential nephroprotective effect, which were used to modify the composition of renal perfusion and preservation solutions. Their effectiveness as ingredients of preservation solutions was analysed based on a literature review. Hormones and trophic factors are innovative preservation solution supplements. They have a pleiotropic effect and affect normal renal function. The expression of receptors for melatonin, prolactin, thyrotropin, corticotropin, prostaglandin E1 and trophic factors was confirmed in the kidneys, which suggests that they are a promising therapeutic target for renal IR (ischemia-reperfusion) injury. They can have anti-inflammatory, antioxidant and anti-apoptotic effects, limiting IR injury.
Collapse
Affiliation(s)
- Aneta Ostróżka-Cieślik
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
| | - Barbara Dolińska
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
- “Biochefa” Pharmaceutical Research and Production Plant, Kasztanowa 3, 41-200 Sosnowiec, Poland
| |
Collapse
|
116
|
Wehland M, Simonsen U, Buus NH, Krüger M, Grimm D. An evaluation of the fixed-dose combination sacubitril/valsartan for the treatment of arterial hypertension. Expert Opin Pharmacother 2020; 21:1133-1143. [PMID: 32133873 DOI: 10.1080/14656566.2020.1735356] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Essential hypertension is a significant risk factor for cardiovascular disease, renal disease, and mortality with increasing prevalence. Despite the availability of various antihypertensive agents, hypertension is still poorly controlled. Therefore, new chemical compounds with antihypertensive efficacy need to be developed. The dual angiotensin II receptor-neprilysin inhibitor LCZ696 is a single molecule synthesized by co-crystallization of valsartan and the neprilysin inhibitor prodrug sacubitril (1:1 molar ratio). AREAS COVERED This review includes an overview of hypertension and the current pharmacotherapy. The authors summarize the LCZ696 drug chemistry, pharmacodynamics, pharmacokinetics, metabolism, randomized control trials (RCTs), and safety concerns. Databases searched included PubMed, Google Scholar, Embase, and ClinicalTrials.gov. EXPERT OPINION LCZ696 is effective in hypertension treatment. Short-term RCTs have shown that the highest doses of LCZ696 (200 and 400 mg [q.d.]) were more effective at lowering office and ambulatory blood pressure than angiotensin II receptor blockers (ARB) alone while having a similar tolerability profile. The effects of LCZ696 on hypertensive organ damage are only sparsely investigated and so far no studies have established the impact of LCZ696 on cardiovascular event rates. Future studies should focus on the comparison of LCZ696 and combination therapies already in use such as ARB and calcium channel blockers.
Collapse
Affiliation(s)
- Markus Wehland
- Department of Biomedicine, Aarhus University , Aarhus C, Denmark.,Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg , Magdeburg, Germany
| | - Ulf Simonsen
- Department of Biomedicine, Aarhus University , Aarhus C, Denmark
| | - Niels Henrik Buus
- Department of Biomedicine, Aarhus University , Aarhus C, Denmark.,Department of Renal Medicine, Aarhus University Hospital , Aarhus, Denmark
| | - Marcus Krüger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg , Magdeburg, Germany
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University , Aarhus C, Denmark.,Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg , Magdeburg, Germany.,Department of Microgravity and Translational Regenerative Medicine, Faculty of Medicine and Mechanical Engineering, Otto von Guericke University , Magdeburg, Germany
| |
Collapse
|
117
|
Abstract
Acute heart failure (AHF) is a syndrome defined as the new onset (de novo heart failure (HF)) or worsening (acutely decompensated heart failure (ADHF)) of symptoms and signs of HF, mostly related to systemic congestion. In the presence of an underlying structural or functional cardiac dysfunction (whether chronic in ADHF or undiagnosed in de novo HF), one or more precipitating factors can induce AHF, although sometimes de novo HF can result directly from the onset of a new cardiac dysfunction, most frequently an acute coronary syndrome. Despite leading to similar clinical presentations, the underlying cardiac disease and precipitating factors may vary greatly and, therefore, the pathophysiology of AHF is highly heterogeneous. Left ventricular diastolic or systolic dysfunction results in increased preload and afterload, which in turn lead to pulmonary congestion. Fluid retention and redistribution result in systemic congestion, eventually causing organ dysfunction due to hypoperfusion. Current treatment of AHF is mostly symptomatic, centred on decongestive drugs, at best tailored according to the initial haemodynamic status with little regard to the underlying pathophysiological particularities. As a consequence, AHF is still associated with high mortality and hospital readmission rates. There is an unmet need for increased individualization of in-hospital management, including treatments targeting the causative factors, and continuation of treatment after hospital discharge to improve long-term outcomes.
Collapse
Affiliation(s)
- Mattia Arrigo
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Mariell Jessup
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wilfried Mullens
- Ziekenhuis Oost Limburg, Genk, Belgium
- University of Hasselt, Hasselt, Belgium
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Department of Medicine and Cardiology, University of Cape Town, Cape Town, South Africa
| | - Alexandre Mebazaa
- Université de Paris, MASCOT, Inserm, Paris, France.
- Department of Anesthesia, Burn and Critical Care Medicine, AP-HP, Hôpital Lariboisière, Paris, France.
| |
Collapse
|
118
|
cGMP signalling in cardiomyocyte microdomains. Biochem Soc Trans 2020; 47:1327-1339. [PMID: 31652306 DOI: 10.1042/bst20190225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
3',5'-Cyclic guanosine monophosphate (cGMP) is one of the major second messengers critically involved in the regulation of cardiac electrophysiology, hypertrophy, and contractility. Recent molecular and cellular studies have significantly advanced our understanding of the cGMP signalling cascade, its local microdomain-specific regulation and its role in protecting the heart from pathological stress. Here, we summarise recent findings on cardiac cGMP microdomain regulation and discuss their potential clinical significance.
Collapse
|
119
|
Polónia J, Gonçalves FR. The historical evolution of knowledge of the involvement of neurohormonal systems in the pathophysiology and treatment of heart failure. Rev Port Cardiol 2020; 38:883-895. [PMID: 32063405 DOI: 10.1016/j.repc.2019.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/22/2019] [Accepted: 05/26/2019] [Indexed: 01/29/2023] Open
Abstract
Our knowledge of the pathophysiology of heart failure (HF) underwent profound changes during the 1980s. Once thought to be of exclusively structural origin, HF began to be seen as the consequence of hormonal imbalance. A number of seminal studies were published in that decade focusing on the impact of neurohormonal activation in HF. Presently, eight neurohormonal systems are known to have a key role in HF development: four stimulate vasoconstriction and sodium/water retention (the sympathetic nervous system, the renin-angiotensin-aldosterone system [RAAS], endothelin, and the vasopressin-arginine system), while the other four stimulate vasodilation and natriuresis (the prostaglandin system, nitric oxide, the dopaminergic system, and the natriuretic peptide system [NPS]). These systems are strongly interconnected and are subject to intricate regulation, functioning together in a delicate homeostasis. Disruption of this homeostasis is characteristic of HF. This review explores the historical development of knowledge on the impact of the neurohormonal systems on HF pathophysiology, from the first studies to current understanding. In addition, the therapeutic potential of each of these systems is discussed, and currently used neurohormonal antagonists are characterized. Special emphasis is given to the latest drug approved for use in HF with reduced ejection fraction, sacubitril/valsartan. This drug combines two different molecules, acting on two different systems (RAAS and NPS) simultaneously.
Collapse
Affiliation(s)
- Jorge Polónia
- Departamento de Medicina da Faculdade de Medicina da Universidade do Porto, Porto, Portugal.
| | | |
Collapse
|
120
|
Parsanathan R, Jain SK. Novel Invasive and Noninvasive Cardiac-Specific Biomarkers in Obesity and Cardiovascular Diseases. Metab Syndr Relat Disord 2020; 18:10-30. [PMID: 31618136 PMCID: PMC7041332 DOI: 10.1089/met.2019.0073] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of fatality and disability worldwide regardless of gender. Obesity has reached epidemic proportions in population across different regions. According to epidemiological studies, CVD risk markers in childhood obesity are one of the significant risk factors for adulthood CVD, but have received disproportionally little attention. This review has examined the evidence for the presence of traditional cardiac biomarkers (nonspecific; lactate dehydrogenase, alanine aminotransferase, aspartate aminotransferase, creatine kinase, myoglobulin, glycogen phosphorylase isoenzyme BB, myosin light chains, ST2, and ischemia-modified albumin) and novel emerging cardiac-specific biomarkers (cardiac troponins, natriuretic peptides, heart-type fatty acid-binding protein, and miRNAs). Besides, noninvasive anatomical and electrophysiological markers (carotid intima-media thickness, coronary artery calcification, and heart rate variability) in CVDs and obesity are also discussed. Modifiable and nonmodifiable risk factors associated with metabolic syndrome in the progression of CVD, such as obesity, diabetes, hypertension, dyslipidemia, oxidative stress, inflammation, and adipocytokines are also outlined. These underlying prognostic risk factors predict the onset of future microvascular and macrovascular complications. The understanding of invasive and noninvasive cardiac-specific biomarkers and the risk factors may yield valuable insights into the pathophysiology and prevention of CVD in a high-risk obese population at an early stage.
Collapse
Affiliation(s)
- Rajesh Parsanathan
- Department of Pediatrics and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Sushil K. Jain
- Department of Pediatrics and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| |
Collapse
|
121
|
Grubić Rotkvić P, Cigrovski Berković M, Rotkvić L, Bulj N. Prevention of cardiac allograft vasculopathy - A new possible indication for SGLT-2 inhibitors? Med Hypotheses 2020; 137:109594. [PMID: 32006921 DOI: 10.1016/j.mehy.2020.109594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/15/2020] [Accepted: 01/22/2020] [Indexed: 10/25/2022]
Abstract
One of the main risk factors influencing patient survival after heart transplantation is cardiac allograft vasculopathy, the leading cause of death after the first year of transplantation. It is an entity of multifactorial origin including both humoral and cellular alloimmune responses as well as immunologic-independent factors such as graft injury, ischaemia-reperfusion injury, oxidative stress, cytomegalovirus infection, hyperlipidaemia, diabetes mellitus and hypertension. A fundamental characteristic of cardiac allograft vasculopathy is vascular remodelling, initially driven by the injury and apoptosis of endothelial cells, then by the migration of smooth muscle cells leading to intimal thickening and ultimately allograft vessel occlusion. Since cardiac allograft vasculopathy occurs within the first year of transplantation, prevention strategies should be implemented early. The disease could be partially prevented with overall cardiovascular risk reduction, mainly by controlling diabetes, hyperlipidemia and hypertension that can be related to the recipient but also induced or augmented by immunosuppressive drugs used. Current therapeutic options are only partially effective in postponing the development of vascular lesions. Diabetes is an important issue in the management of patients following cardiac transplantation. Although it is highly prevalent among heart transplant recipients (23% at 1 year increasing to 37% at 5 years after the procedure), no specific therapeutic protocols have been recommended yet. Sodium-glucose cotransporter-2 (SGLT-2) inhibitors are a novel class of antidiabetic drugs that produce glycosuric and natriuretic effects by inhibiting glucose and sodium reabsorption from the renal proximal tubules and have already shown benefits in cardiovascular outcome trials. Our hypothesis is that SGLT-2 inhibitors could prevent or delay the development of cardiac allograft vasculopathy targeting various mechanisms underpinning its pathogenesis due to their antidiabetic, antihypertensive, anti-inflammatory, antifibrotic, antioxidative and antiapoptotic effects, as well as through amelioration of endothelial dysfunction, ischaemia-reperfusion injury and modification of neurohumoral system. All the segments of the proposed theory that could interfere with evolution of vasculopathy are discussed separately within the main text. The implications for the science if the hypothesis were to be confirmed are as follows: prolongation of lifespan in heart transplant patients with diabetes, reduction of polypragmasia in posttransplant patients while targeting several mechanisms with one drug, and the possibility of spreading the indications even to patients without diabetes.
Collapse
Affiliation(s)
- Petra Grubić Rotkvić
- Department of Cardiology, University Hospital "Sveti Duh", Sveti Duh 64, 10 000 Zagreb, Croatia.
| | - Maja Cigrovski Berković
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia; Department for Medicine of Sports and Exercise, Faculty of Kinesiology, University of Zagreb, Zagreb, Croatia
| | - Luka Rotkvić
- Department of Cardiology, Magdalena Clinic for Cardiovascular Disease, Krapinske Toplice, Croatia
| | - Nikola Bulj
- Department of Cardiology, University Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
122
|
Kiuchi MG, Ho JK, Nolde JM, Gavidia LML, Carnagarin R, Matthews VB, Schlaich MP. Sympathetic Activation in Hypertensive Chronic Kidney Disease - A Stimulus for Cardiac Arrhythmias and Sudden Cardiac Death? Front Physiol 2020; 10:1546. [PMID: 32009970 PMCID: PMC6974800 DOI: 10.3389/fphys.2019.01546] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Studies have revealed a robust and independent correlation between chronic kidney disease (CKD) and cardiovascular (CV) events, including death, heart failure, and myocardial infarction. Recent clinical trials extend this range of adverse CV events, including malignant ventricular arrhythmias and sudden cardiac death (SCD). Moreover, other studies point out that cardiac structural and electrophysiological changes are a common occurrence in this population. These processes are likely contributors to the heightened hazard of arrhythmias in CKD population and may be useful indicators to detect patients who are at a higher SCD risk. Sympathetic overactivity is associated with increased CV risk, specifically in the population with CKD, and it is a central feature of the hypertensive state, occurring early in its clinical course. Sympathetic hyperactivity is already evident at the earliest clinical stage of CKD and is directly related to the progression of renal failure, being most pronounced in those with end-stage renal disease. Sympathetic efferent and afferent neural activity in kidney failure is a crucial facilitator for the perpetuation and evolvement of the disease. Here, we will revisit the role of the feedback loop of the sympathetic neural cycle in the context of CKD and how it may aggravate several of the risk factors responsible for causing SCD. Targeting the overactive sympathetic nervous system therapeutically, either pharmacologically or with newly available device-based approaches, may prove to be a pivotal intervention to curb the substantial burden of cardiac arrhythmias and SCD in the high-risk population of patients with CKD.
Collapse
Affiliation(s)
- Márcio Galindo Kiuchi
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Jan K Ho
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Janis Marc Nolde
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Leslie Marisol Lugo Gavidia
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia.,Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, WA, Australia.,Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
123
|
Savitha MN, Suvilesh KN, Siddesha JM, Milan Gowda MD, Choudhury M, Velmurugan D, Umashankar M, Vishwanath BS. Combinatorial inhibition of Angiotensin converting enzyme, Neutral endopeptidase and Aminopeptidase N by N-methylated peptides alleviates blood pressure and fibrosis in rat model of dexamethasone-induced hypertension. Peptides 2020; 123:170180. [PMID: 31715212 DOI: 10.1016/j.peptides.2019.170180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 01/07/2023]
Abstract
Angiotensin converting enzyme (ACE), neutral endopeptidase (NEP) and aminopeptidase N (APN) are responsible for generation of vasoactive peptides that regulates vasoconstriction, vasodilation and natriuresis, which altogether regulate blood pressure. Cumulative inhibition of ACE, NEP and APN effectively blocks the progression of respective pathways. In this study, N-methylated peptide inhibitors F-N(Me)H-L, V-N(Me)F-R and R-N(Me)V-Y were synthesized against ACE, NEP and APN respectively, using their respective physiological substrates. F-N(Me)H-L inhibited ACE activity with an IC50 of 83 nmol/L, V-N(Me)F-R inhibited NEP activity with an IC50 of 1.173 μmol/L and R-N(Me)V-Y inhibited APN activity with an IC50 of 3.94 nmol/L respectively. Further, the anti-hypertensive effect of N-methylated peptides was evaluated using rat model of dexamethasone-induced hypertension. Individual peptides and their cocktail treatment were started from day 6 of the study period and blood pressure was measured on every alternate day during 15 day study. Administration of F-N(Me)H-L (138 ± 3 mmHg) and cocktail of all the three peptides at a dose of 100 mg/kg significantly reduced systolic blood pressure (SBP) compared to dexamethasone group (SBP of Groups-dexamethasone; (167 ± 5 mmHg), F-N(Me)H-L (138 ± 3 mmHg), and Cocktail (122 ± 3 mmHg). Anti-hypertensive, anti-hypertrophic and anti-fibrotic effects of N-methylated peptides and cocktail was further reflected by the decreased levels of circulating Ang II and increased ANP levels in sera of hypertensive rats along with decrease in collagen deposition in heart and kidney. Though, ACE inhibition is adequate to reduce SBP, targeting NEP and APN along with ACE is beneficial in tackling hypertension and associated fibrosis of heart.
Collapse
Affiliation(s)
- Mysuru Natarajan Savitha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India
| | - Kanve Nagaraj Suvilesh
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India
| | - Jalahalli Mariswamy Siddesha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India; Division of Biochemistry, JSS Academy of Higher education and Research, Mysuru 570015
| | - M D Milan Gowda
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India
| | - Manisha Choudhury
- Department of Crystallography and Biophysics, University of Madras, Chennai, 600005, India
| | - Devadasan Velmurugan
- Department of Crystallography and Biophysics, University of Madras, Chennai, 600005, India
| | - Muddegowda Umashankar
- Department of Chemistry, Karnataka State Open University, Mukthagangotri, Mysuru, 570006, India
| | | |
Collapse
|
124
|
Mottelson MN, Lundsgaard CC, Møller S. Mechanisms in fluid retention - towards a mutual concept. Clin Physiol Funct Imaging 2019; 40:67-75. [PMID: 31823451 DOI: 10.1111/cpf.12615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 12/04/2019] [Indexed: 12/12/2022]
Abstract
Fluid retention is a common and challenging condition in daily clinical practice. The normal fluid homoeostasis in the human body is based on accurately counter-balanced physiological mechanisms. When compromised fluid retention occurs and is seen in pathophysiologically different conditions such as liver cirrhosis, heart and kidney failure, and in preeclampsia. These conditions may share pathophysiological mechanisms such as functional arterial underfilling, which seems to be a mutual element in cirrhosis, cardiac failure, cardiorenal and hepatorenal syndromes, and in pregnancy. However, there are also distinct differences and it is still unclear whether kidney dysfunction or arterial underfilling is the initiating factor of fluid retention or if they happen simultaneously. This review focuses on similarities and differences in water retaining conditions and points to areas where important knowledge is still needed.
Collapse
Affiliation(s)
- Mathis N Mottelson
- Department of Clinical Physiology and Nuclear Medicine, Centre of Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.,Department of Internal Medicine, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Christoffer C Lundsgaard
- Department of Clinical Physiology and Nuclear Medicine, Centre of Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Søren Møller
- Department of Clinical Physiology and Nuclear Medicine, Centre of Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
125
|
Ying W, Zhao D, Ouyang P, Subramanya V, Vaidya D, Ndumele CE, Guallar E, Sharma K, Shah SJ, Kass DA, Hoogeveen RC, Lima JA, Heckbert SR, deFilippi CR, Post WS, Michos ED. Associations Between the Cyclic Guanosine Monophosphate Pathway and Cardiovascular Risk Factors: MESA. J Am Heart Assoc 2019; 8:e013149. [PMID: 31838972 PMCID: PMC6951064 DOI: 10.1161/jaha.119.013149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background cGMP mediates numerous cardioprotective functions and is a potential therapeutic target for cardiovascular disease. Preclinical studies suggest that plasma cGMP is reflective of natriuretic peptide stimulation. Epidemiologic associations between cGMP and natriuretic peptide, as well as cardiovascular disease risk factors, are unknown. Methods and Results We measured plasma cGMP in 542 men and 496 women free of cardiovascular disease and heart failure in MESA (Multi‐Ethnic Study of Atherosclerosis). Cross‐sectional associations of N‐terminal pro‐B type natriuretic peptide, sex hormones, and cardiovascular disease/heart failure risk factors with log(cGMP) were analyzed using multivariable linear regression models. Mean (SD) cGMP was 4.7 (2.6) pmol/mL, with no difference between the sexes. After adjusting for cardiovascular risk factors, N‐terminal pro‐B type natriuretic peptide was significantly positively associated with cGMP (P<0.05). Higher blood pressure and lower estimated glomerular filtration rate were associated with higher cGMP (P<0.05). Triglyceride levels, total/high‐density lipoprotein cholesterol ratio, presence of diabetes mellitus, and the homeostatic model assessment of insulin resistance were inversely associated with cGMP (P<0.05). Among women, free testosterone and dehydroepiandrosterone were inversely associated with cGMP, while sex hormone binding globulin was positively associated (P<0.05). Conclusions In a community‐cohort, plasma cGMP was associated with natriuretic peptide signaling. Higher blood pressure and greater renal dysfunction were positively associated with cGMP, while adverse metabolic risk factors were inversely associated. Increased androgenicity in postmenopausal women was inversely associated with cGMP. These novel associations further our understanding of the role of cGMP in a general population.
Collapse
Affiliation(s)
- Wendy Ying
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Di Zhao
- Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore MD
| | - Pamela Ouyang
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Vinita Subramanya
- Department of Epidemiology Emory University Rollins School of Public Health Atlanta GA
| | - Dhananjay Vaidya
- Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore MD.,Division of General Internal Medicine Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Chiadi E Ndumele
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD.,Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore MD
| | - Eliseo Guallar
- Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore MD
| | - Kavita Sharma
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Sanjiv J Shah
- Division of Cardiology Department of Medicine Northwestern University Feinberg School of Medicine Chicago IL
| | - David A Kass
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Ron C Hoogeveen
- Division of Atherosclerosis & Vascular Medicine Department of Medicine Baylor College of Medicine Houston TX
| | - Joao A Lima
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology University of Washington Seattle WA
| | | | - Wendy S Post
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD.,Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore MD
| | - Erin D Michos
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD.,Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore MD
| |
Collapse
|
126
|
The historical evolution of knowledge of the involvement of neurohormonal systems in the pathophysiology and treatment of heart failure. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.repce.2020.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
127
|
Feygina EE, Katrukha AG, Semenov AG. Neutral Endopeptidase (Neprilysin) in Therapy and Diagnostics: Yin and Yang. BIOCHEMISTRY (MOSCOW) 2019; 84:1346-1358. [PMID: 31760922 DOI: 10.1134/s0006297919110105] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neprilysin (NEP) is a zinc-dependent metalloproteinase that exists in organisms in both transmembrane and soluble forms. NEP substrates are involved in regulating the cardiovascular and nervous systems. In this review, we discuss some of the biochemical characteristics and physiological functions of this enzyme with special emphasis on the use of NEP as a therapeutic target. The history and various physiological aspects of applying NEP inhibitors for treating heart failure and attempts to increase NEP activity when treating Alzheimer's disease using gene and cell therapies are described. Another important issue discussed is the role of NEP as a potential marker for predicting the risk of cardiovascular disease complications. The diagnostic and prognostic performance of soluble NEP in various types of heart failure is analyzed and presented. We also discuss the methods and approaches for measuring NEP activity for prognosis and diagnosis, as well as a possible new role of natriuretic peptides (NEP substrates) in cardiovascular diagnostics.
Collapse
Affiliation(s)
- E E Feygina
- HyTest Ltd., Turku, 20520, Finland. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - A G Katrukha
- HyTest Ltd., Turku, 20520, Finland.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - A G Semenov
- HyTest Ltd., Turku, 20520, Finland.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
128
|
Roxana ME, Georgică TÃ, Ionuț D, Gianina M, Cristina F. Atrial and Brain Natriuretic Peptides- Benefits and Limits of their use in Cardiovascular Diseases. Curr Cardiol Rev 2019; 15:283-290. [PMID: 30914031 PMCID: PMC8142359 DOI: 10.2174/1573403x15666190326150550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023] Open
Abstract
Natriuretic peptides, produced by cardiac myocytes, are regulators of the intravascular volume and blood pressure, and also exhibit neuroendocrine, metabolic and growth controlling effects. In heart failure, their synthesis increases exponentially as part of the neuroendocrine activation, but their beneficial effects are diminished. The paper reviews relevant data about their role as diagnosis and prognosis markers in heart failure, the hemodynamic and clinical benefits of their use as therapy in heart failure, together with the main adverse effects. Peptides non-specifically increase in extracardiac pathology and the literature reveals the mechanisms of increase, significance and threshold values to exclude cardiac dysfunction.
Collapse
Affiliation(s)
- Mustafa Edme Roxana
- Department of Internal Medicine-Cardiology, Craiova University of Medicine and Pharmacy, Craiova 200349, Romania
| | - TÃrtea Georgică
- Department of Physiology, Craiova University of Medicine and Pharmacy, Craiova 200349, Romania
| | - Donoiu Ionuț
- Department of Internal Medicine-Cardiology, Craiova University of Medicine and Pharmacy, Craiova 200349, Romania
| | - Moise Gianina
- County Clinical Emergency Hospital of Craiova, Craiova 200349, Romania
| | - Florescu Cristina
- Department of Internal Medicine-Cardiology, Craiova University of Medicine and Pharmacy, Craiova 200349, Romania
| |
Collapse
|
129
|
Lu YY, Cheng CC, Wu HJ, Lin YK, Chen YC, Chen SA, Chen YJ. Effects of ANP on pulmonary vein electrophysiology, Ca 2+ homeostasis and adrenergic arrhythmogenesis via PKA. Clin Exp Pharmacol Physiol 2019; 47:247-254. [PMID: 31637746 DOI: 10.1111/1440-1681.13199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/28/2022]
Abstract
Atrial fibrillation (AF) is the most common form of arrhythmia and increases the risk of stroke and heart failure (HF). Pulmonary veins (PVs) are important sources of triggers that generate AF, and calcium (Ca2+ ) overload participates in PV arrhythmogenesis. Neurohormonal activation is an important cause of AF. Higher atrial natriuretic peptide (ANP) level predicts paroxysmal AF occurrence in HF patients. However, it is not clear if ANP directly modulates electrophysiological characteristics and Ca2+ homeostasis in the PVs. Conventional microelectrodes, whole-cell patch-clamp, and the Fluo-3 fluorimetric ratio technique were performed using isolated rabbit PV preparations or single isolated PV cardiomyocytes before and after ANP administration. We found that ANP (1, 10, and 100 nmol/L) concentration-dependently decreased spontaneous activity in PV preparations. ANP (100 nmol/L) decreased isoproterenol (1 μmol/L)-induced PV spontaneous activity and burst firing. AP811 (100 nmol/L, NPR-C agonist), H89 (1μmol/L, PKA inhibitor) decreased isoproterenol-induced PV spontaneous activity or burst firing, but successive administration of ANP had no further effect on PV activity. KT5823 (1 μmol/L, PKG inhibitor) decreased isoproterenol-induced PV spontaneous activity but did not change isoproterenol-induced PV burst firing, whereas successive administration of ANP did not change isoproterenol-induced PV burst firing. ANP decreased intracellular Ca2+ transient and sarcoplasmic reticulum Ca2+ content in single PV cardiomyocytes. ANP decreased the late sodium current, L-type Ca2+ current, but did not change nickel-sensitive Na+ -Ca2+ exchanger current in single PV cardiomyocytes. In conclusion, ANP directly regulates PV electrophysiological characteristics and Ca2+ homeostasis and attenuates isoproterenol-induced arrhythmogenesis through NPR-C/cAMP/PKA signal pathway.
Collapse
Affiliation(s)
- Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan.,School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | | | - Hui-Jun Wu
- Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
130
|
Ahmed A, Gulino A, Amayo S, Arancio W, Florena AM, Belmonte B, Jurjus A, Leone A, Miletich I. Natriuretic peptide system expression in murine and human submandibular salivary glands: a study of the spatial localisation of ANB, BNP, CNP and their receptors. J Mol Histol 2019; 51:3-13. [PMID: 31722080 DOI: 10.1007/s10735-019-09849-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/18/2019] [Indexed: 11/30/2022]
Abstract
The natriuretic peptide (NP) system comprises of three ligands, the Atrial Natriuretic Peptide (ANP), Brain Natriuretic peptide (BNP) and C-type Natriuretic peptide (CNP), and three natriuretic peptide receptors, NPRA, NPRB and NPRC. Here we present a comprehensive study of the natriuretic peptide system in healthy murine and human submandibular salivary glands (SMGs). We show CNP is the dominant NP in mouse and human SMG and is expressed together with NP receptors in ducts, autonomic nerves and the microvasculature of the gland, suggesting CNP autocrine signalling may take place in some of these glandular structures. These data suggest the NP system may control salivary gland function during homeostasis through the regulation of electrolyte re-absorption, neural stimulation and/or blood vessel wall contraction/relaxation. We also show abnormal expression of NPRA in the stroma of a subset of human SMGs resected from patients diagnosed with oral squamous cell carcinoma (OSCC) of non-salivary gland origin. This finding warrants further research to investigate a possible correlation between early OSCC invasion and NPRA overexpression.
Collapse
Affiliation(s)
- Araz Ahmed
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Hospital, Floor 27 Guy's Tower, London, SE1 9RT, UK
| | - Alessandro Gulino
- Dipartimento Di Promozione Della Salute, Materno-Infantile, Medicina Interna E Specialistica Di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Simita Amayo
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Hospital, Floor 27 Guy's Tower, London, SE1 9RT, UK
| | - Walter Arancio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Ada Maria Florena
- Dipartimento Di Promozione Della Salute, Materno-Infantile, Medicina Interna E Specialistica Di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Beatrice Belmonte
- Dipartimento Di Promozione Della Salute, Materno-Infantile, Medicina Interna E Specialistica Di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Beirut, Lebanon
| | - Angelo Leone
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Hospital, Floor 27 Guy's Tower, London, SE1 9RT, UK. .,Department of Biomedicine, Neuroscience and Advanced Diagnostic, Bi.N.D, School of Medicine, Institute of Anatomy and Histology, University of Palermo, Palermo, Italy.
| | - Isabelle Miletich
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Hospital, Floor 27 Guy's Tower, London, SE1 9RT, UK.
| |
Collapse
|
131
|
Fu S, Chang Z, Luo L, Deng J. Therapeutic Progress and Knowledge Basis on the Natriuretic Peptide System in Heart Failure. Curr Top Med Chem 2019; 19:1850-1866. [PMID: 31448711 DOI: 10.2174/1568026619666190826163536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/25/2019] [Accepted: 07/25/2019] [Indexed: 01/26/2023]
Abstract
Notwithstanding substantial improvements in diagnosis and treatment, Heart Failure (HF) remains a major disease burden with high prevalence and poor outcomes worldwide. Natriuretic Peptides (NPs) modulate whole cardiovascular system and exhibit multiple cardio-protective effects, including the counteraction of the Renin-Angiotensin-Aldosterone System (RAAS) and Sympathetic Nervous System (SNS), promotion of vasodilatation and natriuresis, and inhibition of hypertrophy and fibrosis. Novel pharmacological therapies based on NPs may achieve a valuable shift in managing patients with HF from inhibiting RAAS and SNS to a reversal of neurohormonal imbalance. Enhancing NP bioavailability through exogenous NP administration and inhibiting Neutral Endopeptidase (NEP) denotes valuable therapeutic strategies for HF. On the one hand, NEP-resistant NPs may be more specific as therapeutic choices in patients with HF. On the other hand, NEP Inhibitors (NEPIs) combined with RAAS inhibitors have proved to exert beneficial effects and reduce adverse events in patients with HF. Highly effective and potentially safe Angiotensin Receptor Blocker Neprilysin Inhibitors (ARNIs) have been developed after the failure of NEPIs and Vasopeptidase Inhibitors (VPIs) due to lacking efficacy and safety. Therapeutic progress and knowledge basis on the NP system in HF are summarized in the current review.
Collapse
Affiliation(s)
- Shihui Fu
- Department of Geriatric Cardiology, National Clinical Research Center of Geriatrics Disease, Beijing Key Laboratory of Precision Medicine for Chronic Heart Failure, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhenyu Chang
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Leiming Luo
- Department of Geriatric Cardiology, National Clinical Research Center of Geriatrics Disease, Beijing Key Laboratory of Precision Medicine for Chronic Heart Failure, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Juelin Deng
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
132
|
Lugnier C, Meyer A, Charloux A, Andrès E, Gény B, Talha S. The Endocrine Function of the Heart: Physiology and Involvements of Natriuretic Peptides and Cyclic Nucleotide Phosphodiesterases in Heart Failure. J Clin Med 2019; 8:jcm8101746. [PMID: 31640161 PMCID: PMC6832599 DOI: 10.3390/jcm8101746] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/09/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022] Open
Abstract
Besides pumping, the heart participates in hydro-sodium homeostasis and systemic blood pressure regulation through its endocrine function mainly represented by the large family of natriuretic peptides (NPs), including essentially atrial natriuretic (ANP) and brain natriuretic peptides (BNP). Under normal conditions, these peptides are synthesized in response to atrial cardiomyocyte stretch, increase natriuresis, diuresis, and vascular permeability through binding of the second intracellular messenger’s guanosine 3′,5′-cyclic monophosphate (cGMP) to specific receptors. During heart failure (HF), the beneficial effects of the enhanced cardiac hormones secretion are reduced, in connection with renal resistance to NP. In addition, there is a BNP paradox characterized by a physiological inefficiency of the BNP forms assayed by current methods. In this context, it appears interesting to improve the efficiency of the cardiac natriuretic system by inhibiting cyclic nucleotide phosphodiesterases, responsible for the degradation of cGMP. Recent data support such a therapeutic approach which can improve the quality of life and the prognosis of patients with HF.
Collapse
Affiliation(s)
- Claire Lugnier
- Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France.
| | - Alain Meyer
- Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France.
- Department of Physiology and Functional Explorations, New Civil Hospital, University Hospitals of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France.
| | - Anne Charloux
- Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France.
- Department of Physiology and Functional Explorations, New Civil Hospital, University Hospitals of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France.
| | - Emmanuel Andrès
- Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France.
- Department of Internal Medicine and Metabolic Diseases, Medical Clinic B, Civil Hospital, University Hospitals of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France.
| | - Bernard Gény
- Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France.
- Department of Physiology and Functional Explorations, New Civil Hospital, University Hospitals of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France.
| | - Samy Talha
- Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France.
- Department of Physiology and Functional Explorations, New Civil Hospital, University Hospitals of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France.
| |
Collapse
|
133
|
Zhang Y, Zhang Y, Li X. Roles of small-dose recombinant human brain natriuretic peptide without bolus in Chinese older patients with septic cardiac dysfunction. Eur Geriatr Med 2019; 10:721-726. [PMID: 34652711 DOI: 10.1007/s41999-019-00229-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/01/2019] [Indexed: 01/25/2023]
Abstract
PURPOSE Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. As one of the most common organs is affected by sepsis, cardiac dysfunction increases adverse prognosis. This study was designed to analyze the roles of small-dose recombinant human brain natriuretic peptide without bolus in Chinese older patients with septic cardiac dysfunction. METHODS This study recruited 250 Chinese older patients with sepsis cardiac dysfunction in intensive care unit. Participants were randomly allocated into the control group (n = 125) and recombinant human brain natriuretic peptide group (n = 125). The control group received early goal-directed therapy, and the recombinant human brain natriuretic peptide group received recombinant human brain natriuretic peptide therapy in addition to early goal-directed therapy. RESULTS There was no significant difference in medical histories, infection types, failing organs, mortality within 28 days [37 (29.6%) vs. 34 (27.2%)], intensive care unit stay [27 (21.6) vs. 22 (17.6)] and hospital stay [41 (32.8%) vs. 37 (29.6%)] between the control and recombinant human brain natriuretic peptide groups (P > 0.05 for all). Lengths of intensive care unit [16 (13-22) days] and hospital stay [25 (22-30) days] in the recombinant human brain natriuretic peptide group were significantly shorter than in the control group [21 (14-23) days; 27 (23-30) days; P < 0.05 for all]. There was no significant difference in mean daily Sequential Organ Failure Assessment score between the two groups (P > 0.05 for all). Cardiovascular and respiratory scores (P < 0.05 for all) but not other organ scores (P > 0.05 for all) were significantly lower in the recombinant human brain natriuretic peptide group than in the control group. CONCLUSIONS Small-dose recombinant human brain natriuretic peptide was unable to lower the mortality and improve the liver, renal, and coagulation functions, but able to shorten the length of hospital stay and improve the cardiovascular and respiratory functions in Chinese older patients with septic cardiac dysfunction.
Collapse
Affiliation(s)
- Yongyang Zhang
- Emergency Department, Jining No. 1 People's Hospital, Jining, 272011, China
| | - Ying Zhang
- Department of Surgery, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Xuling Li
- Department of Pharmacy, Jining No. 1 People's Hospital, Jining, 272011, China.
| |
Collapse
|
134
|
Geng Q, Li S, Wang Z, Ren Y. Efficacy and safety of combined neprilysin and RAS inhibition in heart failure: A meta-analysis of randomized controlled trials. Int J Cardiol 2019; 293:159-164. [DOI: 10.1016/j.ijcard.2019.05.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/19/2019] [Accepted: 05/21/2019] [Indexed: 12/28/2022]
|
135
|
Hui D, Naberhuis J, Dibaj S, Naqvi M, Liu D, Bruera E. Association Between Plasma Brain Natriuretic Peptide and Overall Survival in Patients With Advanced Cancer: Preliminary Findings. J Pain Symptom Manage 2019; 58:465-471. [PMID: 31128228 PMCID: PMC6708736 DOI: 10.1016/j.jpainsymman.2019.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/12/2019] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
CONTEXT Atrial and brain natriuretic peptides (ANP and BNP) are established diagnostic and prognostic markers in heart failure, but their utility in patients with advanced cancer is unclear. OBJECTIVES Our objective was to examine the association between plasma natriuretic peptides and survival in patients with advanced cancer without clinical evidence of heart failure. METHODS This exploratory analysis of a multicenter, randomized clinical trial of cancer patients receiving hospice care assessed the association between elevated plasma ANP, BNP, or Pro-BNP (cutoffs of >77, 100, and 900 pg/mL, respectively) and overall survival. Time-to-event analyses, including multivariate Cox regression, were conducted. RESULTS Among 97 patients, the mean age was 67.2 years and the overall survival was 16 days (95% CI, 13-23 days). ANP, BNP, and Pro-BNP were elevated in 29 of 36 (81%), nine of 23 (39%), and 32 of 38 (84%) patients, respectively. Elevated ANP, BNP, or Pro-BNP was associated with worse survival (median 14 vs. 21 days; P = 0.02). BNP or Pro-BNP was inversely associated with overall survival (hazard ratio = 2.27; 95% CI, 1.29-3.97) in univariate Cox regression analysis, and remained significant in multivariate Cox regression analysis (hazard ratio = 3.09; 95% CI, 1.40-6.84) after adjusting for treatment group and known prognostic variables such as performance status, albumin, creatinine, delirium, dyspnea, and anorexia. Elevated ANP alone was not significantly associated with survival (P = 0.17). CONCLUSION Our preliminary findings suggest that BNP or Pro-BNP may be a novel objective prognostic marker in cancer patients without heart failure. Further research is needed to confirm these findings.
Collapse
Affiliation(s)
- David Hui
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Jane Naberhuis
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Seyedeh Dibaj
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mujtaba Naqvi
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Diane Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eduardo Bruera
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
136
|
Normalizing Plasma Renin Activity in Experimental Dilated Cardiomyopathy: Effects on Edema, Cachexia, and Survival. Int J Mol Sci 2019; 20:ijms20163886. [PMID: 31404946 PMCID: PMC6720926 DOI: 10.3390/ijms20163886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022] Open
Abstract
Heart failure (HF) patients frequently have elevated plasma renin activity. We examined the significance of elevated plasma renin activity in a translationally-relevant model of dilated cardiomyopathy (DCM), which replicates the progressive stages (A–D) of human HF. Female mice with DCM and elevated plasma renin activity concentrations were treated with a direct renin inhibitor (aliskiren) in a randomized, blinded fashion beginning at Stage B HF. By comparison to controls, aliskiren treatment normalized pathologically elevated plasma renin activity (p < 0.001) and neprilysin levels (p < 0.001), but did not significantly alter pathological changes in plasma aldosterone, angiotensin II, atrial natriuretic peptide, or corin levels. Aliskiren improved cardiac systolic function (ejection fraction, p < 0.05; cardiac output, p < 0.01) and significantly reduced the longitudinal development of edema (extracellular water, p < 0.0001), retarding the transition from Stage B to Stage C HF. The normalization of elevated plasma renin activity reduced the loss of body fat and lean mass (cachexia/sarcopenia), p < 0.001) and prolonged survival (p < 0.05). In summary, the normalization of plasma renin activity retards the progression of experimental HF by improving cardiac systolic function, reducing the development of systemic edema, cachexia/sarcopenia, and mortality. These data suggest that targeting pathologically elevated plasma renin activity may be beneficial in appropriately selected HF patients.
Collapse
|
137
|
Oh A, Okazaki R, Sam F, Valero-Muñoz M. Heart Failure With Preserved Ejection Fraction and Adipose Tissue: A Story of Two Tales. Front Cardiovasc Med 2019; 6:110. [PMID: 31428620 PMCID: PMC6687767 DOI: 10.3389/fcvm.2019.00110] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by signs and symptoms of heart failure in the presence of a normal left ventricular ejection fraction. Although it accounts for up to 50% of all clinical presentations of heart failure, there are no evidence-based therapies for HFpEF to reduce morbidity and mortality. Additionally there is a lack of mechanistic understanding about the pathogenesis of HFpEF. HFpEF is associated with many comorbidities (such as obesity, hypertension, type 2 diabetes, atrial fibrillation, etc.) and is coupled with both cardiac and extra-cardiac abnormalities. Large outcome trials and registries reveal that being obese is a major risk factor for HFpEF. There is increasing focus on investigating the link between obesity and HFpEF, and the role that the adipose tissue and the heart, and the circulating milieu play in development and pathogenesis of HFpEF. This review discusses features of the obese-HFpEF phenotype and highlights proposed mechanisms implicated in the inter-tissue communication between adipose tissue and the heart in obesity-associated HFpEF.
Collapse
Affiliation(s)
- Albin Oh
- Evans Department of Medicine, Boston Medical Center, Boston, MA, United States
| | - Ross Okazaki
- Boston University School of Medicine, Boston, MA, United States
| | - Flora Sam
- Evans Department of Medicine, Boston Medical Center, Boston, MA, United States
- Boston University School of Medicine, Boston, MA, United States
- Section of Cardiovascular Medicine, Boston Medical Center, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Maria Valero-Muñoz
- Boston University School of Medicine, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
138
|
Okamoto R, Ali Y, Hashizume R, Suzuki N, Ito M. BNP as a Major Player in the Heart-Kidney Connection. Int J Mol Sci 2019; 20:ijms20143581. [PMID: 31336656 PMCID: PMC6678680 DOI: 10.3390/ijms20143581] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Brain natriuretic peptide (BNP) is an important biomarker for patients with heart failure, hypertension and cardiac hypertrophy. Although it is known that BNP levels are relatively higher in patients with chronic kidney disease and no heart disease, the mechanism remains unknown. Here, we review the functions and the roles of BNP in the heart-kidney interaction. In addition, we discuss the relevant molecular mechanisms that suggest BNP is protective against chronic kidney diseases and heart failure, especially in terms of the counterparts of the renin-angiotensin-aldosterone system (RAAS). The renal medulla has been reported to express depressor substances. The extract of the papillary tips from kidneys may induce the expression and secretion of BNP from cardiomyocytes. A better understanding of these processes will help accelerate pharmacological treatments for heart-kidney disease.
Collapse
Affiliation(s)
- Ryuji Okamoto
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan.
| | - Yusuf Ali
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Ryotaro Hashizume
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Noboru Suzuki
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Masaaki Ito
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| |
Collapse
|
139
|
Cannone V, Cabassi A, Volpi R, Burnett JC. Atrial Natriuretic Peptide: A Molecular Target of Novel Therapeutic Approaches to Cardio-Metabolic Disease. Int J Mol Sci 2019; 20:E3265. [PMID: 31269783 PMCID: PMC6651335 DOI: 10.3390/ijms20133265] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 12/11/2022] Open
Abstract
Atrial natriuretic peptide (ANP) is a cardiac hormone with pleiotropic cardiovascular and metabolic properties including vasodilation, natriuresis and suppression of the renin-angiotensin-aldosterone system. Moreover, ANP induces lipolysis, lipid oxidation, adipocyte browning and ameliorates insulin sensitivity. Studies on ANP genetic variants revealed that subjects with higher ANP plasma levels have lower cardio-metabolic risk. In vivo and in humans, augmenting the ANP pathway has been shown to exert cardiovascular therapeutic actions while ameliorating the metabolic profile. MANP is a novel designer ANP-based peptide with greater and more sustained biological actions than ANP in animal models. Recent studies also demonstrated that MANP lowers blood pressure and inhibits aldosterone in hypertensive subjects whereas cardiometabolic properties of MANP are currently tested in an on-going clinical study in hypertension and metabolic syndrome. Evidence from in vitro, in vivo and in human studies support the concept that ANP and related pathway represent an optimal target for a comprehensive approach to cardiometabolic disease.
Collapse
Affiliation(s)
- Valentina Cannone
- Cardiorenal Research Laboratory, Circulatory Failure Division, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
- Division of Clinical Medicine, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Aderville Cabassi
- Division of Clinical Medicine, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Riccardo Volpi
- Division of Clinical Medicine, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - John C Burnett
- Cardiorenal Research Laboratory, Circulatory Failure Division, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
140
|
Chang RL, Nithiyanantham S, Huang CY, Pai PY, Chang TT, Hu LC, Chen RJ, VijayaPadma V, Kuo WW, Huang CY. Synergistic cardiac pathological hypertrophy induced by high-salt diet in IGF-IIRα cardiac-specific transgenic rats. PLoS One 2019; 14:e0216285. [PMID: 31211784 PMCID: PMC6581245 DOI: 10.1371/journal.pone.0216285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 04/17/2019] [Indexed: 11/18/2022] Open
Abstract
Stress-induced cardiac hypertrophy leads to heart failure. Our previous studies demonstrate that insulin-like growth factor-II receptor (IGF-IIR) signaling is pivotal to hypertrophy regulation. In this study, we show a novel IGF-IIR alternative spliced transcript, IGF-IIRα (150 kDa) play a key role in high-salt induced hypertrophy mechanisms. Cardiac overexpression of IGF-IIRα and high-salt diet influenced cardiac dysfunction by increasing pathophysiological changes with up-regulation of hypertrophy markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). We found that, cardiac hypertrophy under high-salt conditions were amplified in the presence of IGF-IIRα overexpression. Importantly, high-salt induced angiotensin II type I receptor (AT1R) up regulation mediated IGF-IIR expressions via upstream mitogen activated protein kinase (MAPK)/silent mating type information regulation 2 homolog 1 (SIRT1)/heat shock factor 1 (HSF1) pathway. Further, G-coupled receptors (Gαq) activated calcineurin/nuclear factor of activated T-cells, cytoplasmic 3 (NFATc3)/protein kinase C (PKC) signaling was significantly up regulated under high-salt conditions. All these effects were observed to be dramatically over-regulated in IGF-IIRα transgenic rats fed with a high-salt diet. Altogether, from the findings, we demonstrate that IGF-IIRα plays a crucial role during high-salt conditions leading to synergistic cardiac hypertrophy.
Collapse
Affiliation(s)
- Ruey-Lin Chang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | | | - Chih-Yang Huang
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Pei-Ying Pai
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Tung-Ti Chang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lai-Chin Hu
- Department of Internal Medicine, Division of Cardiology, Armed Forces Taichung General Hospital, Taichung, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - V. VijayaPadma
- Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- College of Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
141
|
Tanase DM, Radu S, Al Shurbaji S, Baroi GL, Florida Costea C, Turliuc MD, Ouatu A, Floria M. Natriuretic Peptides in Heart Failure with Preserved Left Ventricular Ejection Fraction: From Molecular Evidences to Clinical Implications. Int J Mol Sci 2019; 20:E2629. [PMID: 31142058 PMCID: PMC6600439 DOI: 10.3390/ijms20112629] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
The incidence of heart failure with preserved ejection fraction (HFpEF) is increasing and its challenging diagnosis and management combines clinical, imagistic and biological data. Natriuretic peptides (NPs) are hormones secreted in response to myocardial stretch that, by increasing cyclic guanosine monophosphate (cGMP), counteract myocardial fibrosis and hypertrophy, increase natriuresis and determine vasodilatation. While their role in HFpEF is controversial, most authors focused on b-type natriuretic peptides (BNPs) and agreed that patients may show lower levels. In this setting, newer molecules with an increased specificity, such as middle-region pro-atrial natriuretic peptide (MR-proANP), emerged as promising markers. Augmenting NP levels, either by NP analogs or breakdown inhibition, could offer a new therapeutic target in HFpEF (already approved in their reduced EF counterparts) by increasing the deficient cGMP levels found in patients. Importantly, these peptides also retain their prognostic value. This narrative review focuses on NPs' physiology, diagnosis, therapeutic and prognostic implication in HFpEF.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700111 Iasi, Romania.
- Internal Medicine Clinic, "Sf. Spiridon" County Clinical Emergency Hospital Iasi, 700115 Iasi, Romania.
| | - Smaranda Radu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700111 Iasi, Romania.
- Cardiology Clinic, "Prof. Dr. George I.M. Georgescu" Institute of Cardiovascular Diseases, 700503 Iasi, Romania.
| | - Sinziana Al Shurbaji
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700111 Iasi, Romania.
- Institute of Gastroenterology and Hepatology, 700115 Iasi, Romania.
| | - Genoveva Livia Baroi
- Department of Surgery, "Grigore T. Popa" University of Medicine and Pharmacy, 700111 Iasi, Romania.
- Vascular Surgery Clinic, "Sf. Spiridon" County Clinical Emergency Hospital Iasi, 700115 Iasi, Romania.
| | - Claudia Florida Costea
- Department of Ophthalmology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania.
- 2nd Ophthalmology Clinic, "Prof. Dr. Nicolae Oblu" Emergency Clinical Hospital, 700115 Iași, Romania.
| | - Mihaela Dana Turliuc
- Department of Neurosurgery, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
- 2nd Neurosurgery Clinic, "Prof. Dr. Nicolae Oblu" Emergency Clinical Hospital, 700115 Iași, Romania.
| | - Anca Ouatu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700111 Iasi, Romania.
- Internal Medicine Clinic, "Sf. Spiridon" County Clinical Emergency Hospital Iasi, 700115 Iasi, Romania.
| | - Mariana Floria
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700111 Iasi, Romania.
- Internal Medicine Clinic, "Sf. Spiridon" County Clinical Emergency Hospital Iasi, 700115 Iasi, Romania.
| |
Collapse
|
142
|
Dizaye K, Ali RH. Effects of neprilysin-renin inhibition in comparison with neprilysin-angiotensin inhibition on the neurohumoral changes in rats with heart failure. BMC Pharmacol Toxicol 2019; 20:23. [PMID: 31053170 PMCID: PMC6500024 DOI: 10.1186/s40360-019-0304-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Background The activation of neurohumoral compensatory mechanisms is a common physiological phenomenon in heart failure in order to make up for a failing heart, which will usually have a deteriorating effect on overall health condition. Many medications, such as neprilysin and angiotensin inhibitors, have recently been introduced to remediate neurohumoral changes. This study was conducted to evaluate the efficacy of the sacubitril-aliskiren combination versus the sacubitril-ramipril combination in the treatment of neurohumoral changes in rats with experimentally induced heart failure. Method Thirty Wister rats were randomly assigned into five groups each of six rats, the first group was the control group. Intraperitoneal isoprenaline injections of 5 mg/kg/day for 1 week were used to induce experimental models of heart failure in rats of the rest of experimental groups. The second group served as a positive control. Rats in the third, fourth, and fifth groups received oral daily dose of sacubitril 30 mg/kg/day, sacubitril-aliskiren 30,10 mg/kg/day, and sacubitril-ramipril 30/10 mg/kg/day respectively, for 2 weeks. Results Induction of heart failure in rats has significantly increased circulating NT-proBNP (980 ± 116.71 pg/ml), MMP9 (15.85 ± 0.57 ng/ml), troponin-I (3.09 ± 0.147 ng/ml), CK-MB (31.55 ± 1.69 ng/ml), renin (736 ± 45.8 pg/ml), urea (52.1 ± 1.57 mg/dl), and creatinine (0.92 ± 0.04 mg/dl). Significant decreases in glomerular filtration rate (7.031 ± 1.6 ml/hr./kg), urine flow (0.2761 ± 0.06 ml/h/kg), total solute excretion (0.11 ± 0.03 meq/m), and mean blood pressure (83.5 ± 2.6 mm hg) were seen in rats with heart failure. Rats treated with sacubitril combined with aliskiren or ramipril showed a statistically significant reduction of NT-proBNP, MMP9, troponin serum urea, and serum creatinine. Sacubitril-aliskiren or sacubitril-ramipril administration produced a significant increase in renin plasma level, total solute excretion, urine flow, and glomerular filtration rate. Conclusion Sacubitril in combination with aliskiren or with ramipril effectively reduced plasma cardiac biomarkers, such as CK-MB, MMP9, and NT-proBNP, in rats with heart failure. Both combinations showed significant remediation of renal function through increasing GFR, urine flow, and total solute excretion, as well as reducing plasma level of renin. Net results revealed that the sacubitril-aliskiren combination has similar remediating effects on neurohumoral changes compared to the sacubitril-ramipril combination.
Collapse
Affiliation(s)
- Kawa Dizaye
- College of Medicine, Hawler Medical University, Minara village A05, Erbil, Iraq.
| | - Rojgar H Ali
- College of pharmacy, Hawler Medical University, Erbil, Iraq
| |
Collapse
|
143
|
Sacubitril/valsartan: A practical guide. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.repce.2018.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
144
|
Lin P, Li H, Yu T, Liu Y. The Effect of Angiotensin-Converting Enzyme Gene Polymorphisms on the Clinical Efficacy of Perindopril Prescribed for Acute Myocardial Infarction in Chinese Han Patients. Genet Test Mol Biomarkers 2019; 23:316-324. [PMID: 30942616 DOI: 10.1089/gtmb.2018.0232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective: Perindopril is an angiotensin-converting enzyme (ACE) inhibitor that is commonly used in the treatment of Chinese Han patients with acute myocardial infarction (AMI). However, there have been few studies on whether polymorphisms of the ACE gene affect the efficacy of perindopril or the prognosis of AMI patients. The purpose of this study was to analyze the relationship among the ACE rs121912703 (C>T), rs767880620 (C>A), and rs397514689 (C>T) gene polymorphisms and the prognosis of AMI patients and the clinical efficacy of perindopril in the treatment of AMI. Methods: The ACE genotypes at the rs121912703, rs767880620, and rs397514689 loci in 225 AMI patients treated with perindopril were determined by polymerase chain reaction/Sanger sequencing. Differences in cardiac structure, functional indicators, hemodynamic parameters, and related laboratory indicators were detected before and after treatment. Results: After administration of perindopril, improved ventricular remodeling in AMI patients with wild-type ACE was better than in patients with the ACE rs121912703, rs767880620, and rs397514689 minor variant alleles. The patients harboring wild-type ACE had lower systolic blood pressure and diastolic blood pressure than the patients harboring the minor variant alleles (p < 0.01). The contents of serum ACE and Ang II (angiotensin II) in AMI patients carrying the wild-type ACE alleles were lower than those of patients harboring any of the minor variant alleles (p < 0.01). The 3-year survival time of AMI patients carrying the wild-type ACE alleles was markedly greater compared with AMI patients carrying the mutant genes (p < 0.01). Conclusion: Mutations at the ACE rs121912703, rs767880620, and rs397514689 loci affect the efficacy of perindopril on ventricular remodeling and hemodynamics in Chinese Han AMI patients. The 3-year survival of AMI patients harboring the variant alleles is less than that of the patients harboring the wild-type gene.
Collapse
Affiliation(s)
- Peng Lin
- 1 Department of Intensive Care Unit and Yuhuangding Hospital Affiliated to Qingdao University, Yantai, P.R. China
| | - Haiyong Li
- 2 Department of Emergency, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, P.R. China
| | - Tianhua Yu
- 3 Department of Gynecology and Obstetrics, Penglai Traditional Chinese Medicine Hospital, Yantai, P.R. China
| | - Yuanyuan Liu
- 2 Department of Emergency, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, P.R. China
| |
Collapse
|
145
|
Fauconnier C, Roy T, Gillerot G, Roy C, Pouleur AC, Gruson D. FGF23: Clinical usefulness and analytical evolution. Clin Biochem 2019; 66:1-12. [PMID: 30853324 DOI: 10.1016/j.clinbiochem.2019.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/05/2019] [Accepted: 03/05/2019] [Indexed: 12/16/2022]
Abstract
Fibroblast Growth Factor 23 (FGF23) is a key hormone for the regulation of phosphate homeostasis. Over the past decades, FGF23 was the subject of intense research in the fields of nephrology and the cardiology. It presents a remarkable correlation with well-established biomarkers of cardiovascular disorders in both chronic kidney disease (CKD) and heart failure (HF) patients. The interest of FGF23 lies in its early-onset in the primary course of CKD as well as in the incremental prognosis information it conveys in both CKD and HF. Different types of assays of FGF-23 testing exist, those targeting the intact form (iFGF23), the other one detecting terminal fragments (cFGF23). The issue is still pending which assay suits best for clinical use. Recently, the implementation of this biomarker on multianalyzer platforms, on which other markers of phospho-calcic balance are set up, allows a rapid turn-around-time and a potential financial gain. However, despite the good analytical performances of the automated methods, there is a poor harmonization between assays. The introduction of an international certified reference material should standardize the measurement and improve the harmonization of results from different laboratories. A deeper understanding of physio-pathological mechanisms and processing of FGF-23 should reinforce its clinical indications and might also identify new therapeutic targets for the treatment of CKD and HF.
Collapse
Affiliation(s)
- Charlotte Fauconnier
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Tatiana Roy
- Department of Laboratory Medicine, Clinique Saint-Pierre Ottignies, Belgium
| | - Gaëlle Gillerot
- Nephrology Department, Clinique Saint-Pierre Ottignies, Belgium
| | - Clotilde Roy
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Anne-Catherine Pouleur
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Damien Gruson
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium; Pôle de recherche en endocrinologie, diabète et nutrition, Institut de recherche expérimentale et clinique, Cliniques universitaires Saint-Luc et Université catholique de Louvain, Bruxelles, Belgium.
| |
Collapse
|
146
|
Sun S, Kee HJ, Ryu Y, Choi SY, Kim GR, Kim HS, Kee SJ, Jeong MH. Gentisic acid prevents the transition from pressure overload-induced cardiac hypertrophy to heart failure. Sci Rep 2019; 9:3018. [PMID: 30816171 PMCID: PMC6395621 DOI: 10.1038/s41598-019-39423-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022] Open
Abstract
We previously reported that gentisic acid attenuates cardiac hypertrophy and fibrosis in transverse aortic constriction (TAC)-induced cardiac hypertrophy. Here, we examined whether gentisic acid prevents the development of heart failure. Heart failure was induced in mice via chronic TAC. Mice were administered the vehicle, gentisic acid (10 and 100 mg∙kg-1∙day-1), or bisoprolol (0.5 mg∙kg-1∙day-1) orally for 3 weeks, beginning 3 weeks after TAC. After oral administration of gentisic acid (2000 mg∙kg-1), no significant differences in organ weight, histology, or analyzed serum and hematological parameters were observed between female mice in the control and gentisic acid-treated groups. Gentisic acid administration inhibited cardiac dysfunction in a dose-dependent manner, and reduced cardiac hypertrophy and fibrosis, as was revealed via western blotting, quantitative real-time PCR, and Masson's trichrome staining. Gentisic acid dose-dependently reduced the expression of fibrosis marker genes, suppressed the renin-angiotensin-aldosterone system, and reduced lung size and pulmonary vascular remodeling. Our data indicate that gentisic acid prevents cardiac hypertrophy, fibrosis, cardiac dysfunction, and pulmonary pathology in TAC-induced heart failure. These findings suggest that supplementation with gentisic acid may provide an advantage in preventing the progression from cardiac hypertrophy to heart failure.
Collapse
Affiliation(s)
- Simei Sun
- Zhoushan Hospital, Zhejiang University School of Medicine, No 739, Dingshen Road Lincheng New District, Zhoushan Zhejiang, 316021, China.,Heart Research Center of Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, 61469, Republic of Korea. .,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.
| | - Yuhee Ryu
- Heart Research Center of Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Sin Young Choi
- Heart Research Center of Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Gwi Ran Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University, Medical School and Hospital, Gwangju, 61469, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, 61469, Republic of Korea. .,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.
| |
Collapse
|
147
|
Priddy M, Bock C. Acute Decompensated Heart Failure: A Pharmacotherapy Approach. AACN Adv Crit Care 2019; 29:233-239. [PMID: 30185488 DOI: 10.4037/aacnacc2018293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Mark Priddy
- Mark Priddy is a first-year pharmacy practice resident, Tampa General Hospital, PO Box 1289, Tampa, FL 33601-1289 . Czarina Bock is Clinical Pharmacist, Coronary Care Unit, Tampa General Hospital, Tampa, Florida
| | - Czarina Bock
- Mark Priddy is a first-year pharmacy practice resident, Tampa General Hospital, PO Box 1289, Tampa, FL 33601-1289 . Czarina Bock is Clinical Pharmacist, Coronary Care Unit, Tampa General Hospital, Tampa, Florida
| |
Collapse
|
148
|
Stevenson MJ, Uyeda KS, Harder NHO, Heffern MC. Metal-dependent hormone function: the emerging interdisciplinary field of metalloendocrinology. Metallomics 2019; 11:85-110. [PMID: 30270362 PMCID: PMC10249669 DOI: 10.1039/c8mt00221e] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
For over 100 years, there has been an incredible amount of knowledge amassed concerning hormones in the endocrine system and their central role in human health. Hormones represent a diverse group of biomolecules that are released by glands, communicate signals to their target tissue, and are regulated by feedback loops to maintain organism health. Many disease states, such as diabetes and reproductive disorders, stem from misregulation or dysfunction of hormones. Increasing research is illuminating the intricate roles of metal ions in the endocrine system where they may act advantageously in concert with hormones or deleteriously catalyze hormone-associated disease states. As the critical role of metal ions in the endocrine system becomes more apparent, it is increasingly important to untangle the complex mechanisms underlying the connections between inorganic biochemistry and hormone function to understand and control endocrinological phenomena. This tutorial review harmonizes the interdisciplinary fields of endocrinology and inorganic chemistry in the newly-termed field of "metalloendocrinology". We describe examples linking metals to both normal and aberrant hormone function with a focus on highlighting insight to molecular mechanisms. Hormone activities related to both essential metal micronutrients, such as copper, iron, zinc, and calcium, and disruptive nonessential metals, such as lead and cadmium are discussed.
Collapse
Affiliation(s)
- Michael J Stevenson
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
149
|
Sacubitril/valsartan: A practical guide. Rev Port Cardiol 2019; 38:309-313. [PMID: 30679005 DOI: 10.1016/j.repc.2018.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/09/2018] [Accepted: 10/21/2018] [Indexed: 02/08/2023] Open
Abstract
Renin-angiotensin-aldosterone system (RAAS) inhibitors are a cornerstone in the treatment of heart failure with reduced ejection fraction (HFrEF). Sacubitril/valsartan modulates the neurohormonal axis by inhibiting both angiotensin receptors and neprilysin, and improves neurohormonal balance more than blocking the RAAS alone. The PARADIGM-HF trial validated this new treatment option for patients with HFrEF. Sacubitril/valsartan was also more effective than enalapril in slowing disease progression by decreasing the risk of worsening heart failure requiring hospitalization or emergency admission and the need for intensified therapy, heart failure devices or cardiac transplantation. More than 70% of patients included in PARADIGM-HF were in NYHA class II, and overall, the results indicate that sacubitril/valsartan should be started in the earliest symptomatic stages of the disease. As PARADIGM-HF has excellent robustness for a cardiovascular trial, sacubitril/valsartan has been included as a new treatment option with a strong level of recommendation in the main international guidelines. This expert task force proposes a practical guide to the use of this new drug that has been endorsed by the Working Group on Heart Failure of the Portuguese Society of Cardiology.
Collapse
|
150
|
Vio CP, Salas D, Cespedes C, Diaz-Elizondo J, Mendez N, Alcayaga J, Iturriaga R. Imbalance in Renal Vasoactive Enzymes Induced by Mild Hypoxia: Angiotensin-Converting Enzyme Increases While Neutral Endopeptidase Decreases. Front Physiol 2018; 9:1791. [PMID: 30618804 PMCID: PMC6297360 DOI: 10.3389/fphys.2018.01791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Chronic hypoxia has been postulated as one of the mechanisms involved in salt-sensitive hypertension and chronic kidney disease (CKD). Kidneys have a critical role in the regulation of arterial blood pressure through vasoactive systems, such as the renin-angiotensin and the kallikrein-kinin systems, with the angiotensin-converting enzyme (ACE) and kallikrein being two of the main enzymes that produce angiotensin II and bradykinin, respectively. Neutral endopeptidase 24.11 or neprilysin is another enzyme that among its functions degrade vasoactive peptides including angiotensin II and bradykinin, and generate angiotensin 1-7. On the other hand, the kidneys are vulnerable to hypoxic injury due to the active electrolyte transportation that requires a high oxygen consumption; however, the oxygen supply is limited in the medullary regions for anatomical reasons. With the hypothesis that the chronic reduction of oxygen under normobaric conditions would impact renal vasoactive enzyme components and, therefore; alter the normal balance of the vasoactive systems, we exposed male Sprague-Dawley rats to normobaric hypoxia (10% O2) for 2 weeks. We then processed renal tissue to identify the expression and distribution of kallikrein, ACE and neutral endopeptidase 24.11 as well as markers of kidney damage. We found that chronic hypoxia produced focal damage in the kidney, mainly in the cortico-medullary region, and increased the expression of osteopontin. Moreover, we observed an increase of ACE protein in the brush border of proximal tubules at the outer medullary region, with increased mRNA levels. Kallikrein abundance did not change significantly with hypoxia, but a tendency toward reduction was observed at protein and mRNA levels. Neutral endopeptidase 24.11 was localized in proximal tubules, and was abundantly expressed under normoxic conditions, which markedly decreased both at protein and mRNA levels with chronic hypoxia. Taken together, our results suggest that chronic hypoxia produces focal kidney damage along with an imbalance of key components of the renal vasoactive system, which could be the initial steps for a long-term contribution to salt-sensitive hypertension and CKD.
Collapse
Affiliation(s)
- Carlos P Vio
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Daniela Salas
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Cespedes
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jessica Diaz-Elizondo
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia Mendez
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute of Anatomy, Histology, and Pathology, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Julio Alcayaga
- Laboratorio de Fisiología Celular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Department of Physiology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|