Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.074
Filter
1.
Dis Model Mech ; 17(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38770680

ABSTRACT

Absence of dystrophin results in muscular weakness, chronic inflammation and cardiomyopathy in Duchenne muscular dystrophy (DMD). Pharmacological corticosteroids are the DMD standard of care; however, they have harsh side effects and unclear molecular benefits. It is uncertain whether signaling by physiological corticosteroids and their receptors plays a modifying role in the natural etiology of DMD. Here, we knocked out the glucocorticoid receptor (GR, encoded by Nr3c1) specifically in myofibers and cardiomyocytes within wild-type and mdx52 mice to dissect its role in muscular dystrophy. Double-knockout mice showed significantly worse phenotypes than mdx52 littermate controls in measures of grip strength, hang time, inflammatory pathology and gene expression. In the heart, GR deletion acted additively with dystrophin loss to exacerbate cardiomyopathy, resulting in enlarged hearts, pathological gene expression and systolic dysfunction, consistent with imbalanced mineralocorticoid signaling. The results show that physiological GR functions provide a protective role during muscular dystrophy, directly contrasting its degenerative role in other disease states. These data provide new insights into corticosteroids in disease pathophysiology and establish a new model to investigate cell-autonomous roles of nuclear receptors and mechanisms of pharmacological corticosteroids.


Subject(s)
Dystrophin , Mice, Inbred mdx , Mice, Knockout , Receptors, Glucocorticoid , Animals , Receptors, Glucocorticoid/metabolism , Dystrophin/metabolism , Dystrophin/genetics , Dystrophin/deficiency , Myocardium/pathology , Myocardium/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Mice , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Mice, Inbred C57BL , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/metabolism , Phenotype , Systole/drug effects
2.
Sci Rep ; 14(1): 12038, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802475

ABSTRACT

Hypertrophic cardiomyopathy (HCM) remains the most common cardiomyopathy in humans and cats with few preclinical pharmacologic interventional studies. Small-molecule sarcomere inhibitors are promising novel therapeutics for the management of obstructive HCM (oHCM) patients and have shown efficacy in left ventricular outflow tract obstruction (LVOTO) relief. The objective of this study was to explore the 6-, 24-, and 48-hour (h) pharmacodynamic effects of the cardiac myosin inhibitor, CK-586, in six purpose-bred cats with naturally occurring oHCM. A blinded, randomized, five-treatment group, crossover preclinical trial was conducted to assess the pharmacodynamic effects of CK-586 in this oHCM model. Dose assessments and select echocardiographic variables were assessed five times over a 48-h period. Treatment with oral CK-586 safely ameliorated LVOTO in oHCM cats. CK-586 treatment dose-dependently eliminated obstruction (reduced LVOTOmaxPG), increased measures of systolic chamber size (LVIDs Sx), and decreased select measures of heart function (LV FS% and LV EF%) in the absence of impact on heart rate. At all tested doses, a single oral CK-586 dose resulted in improved or resolved LVOTO with well-tolerated, dose-dependent, reductions in LV systolic function. The results from this study pave the way for the potential use of CK-586 in both the veterinary and human clinical setting.


Subject(s)
Cardiac Myosins , Cardiomyopathy, Hypertrophic , Animals , Cats , Cardiomyopathy, Hypertrophic/drug therapy , Cardiac Myosins/metabolism , Cat Diseases/drug therapy , Male , Female , Ventricular Outflow Obstruction/drug therapy , Systole/drug effects , Echocardiography , Cross-Over Studies
3.
Sci Rep ; 11(1): 24097, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916560

ABSTRACT

Levosimendan exerts positive inotropic and vasodilatory effects. Currently, its effects on right heart function remain uncertain. This systematic review and meta-analysis is intended to illustrate the impacts of levosimendan on systolic function of the right heart in patients with heart dysfunction. We systematically searched electronic databases (PubMed, the Cochrane Library, Embase and Web of Science) up to November 30, 2020, and filtered eligible studies that reported the impacts of levosimendan on right heart function. Of these, only studies whose patients suffered from heart dysfunction or pulmonary hypertension were included. Additionally, patients were divided into two groups (given levosimendan or not) in the initial research. Then, RevMan5.3 was used to conduct further analysis. A total of 8 studies comprising 390 patients were included. The results showed that after 24 h of levosimendan, patients' right ventricular fractional area change [3.17, 95% CI (2.03, 4.32), P < 0.00001], tricuspid annular plane systolic excursion [1.26, 95% CI (0.35, 2.16), P = 0.007] and tricuspid annular peak systolic velocity [0.86, 95% CI (0.41, 1.32), P = 0.0002] were significantly increased compared to the control group. And there is an increasing trend of cardiac output in levosimendan group [1.06, 95% CI (- 0.16, 2.29), P = 0.09 ] .Furthermore, patients' systolic pulmonary arterial pressure [- 5.57, 95% CI (- 7.60, - 3.54), P < 0.00001] and mean pulmonary arterial pressure [- 1.01, 95% CI (- 1.64, - 0.37), P = 0.002] were both significantly decreased, whereas changes in pulmonary vascular resistance [- 55.88, 95% CI (- 206.57, 94.82), P = 0.47] were not significant. Our study shows that in patients with heart dysfunction, levosimendan improves systolic function of the right heart and decreases the pressure of the pulmonary artery.


Subject(s)
Cardiotonic Agents , Simendan/administration & dosage , Simendan/pharmacology , Vasodilator Agents , Ventricular Dysfunction, Right/drug therapy , Aged , Cardiac Output/drug effects , Female , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Systole/drug effects , Vascular Resistance/drug effects , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right/drug effects
4.
Nutrients ; 13(12)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34959763

ABSTRACT

The role of magnesium in blood pressure has been studied among hypertensive patients; however, there is a dearth of studies exploring the role of magnesium in hypertensive crises. The primary objective of this study was to evaluate the relationship between serum magnesium and blood pressure in patients with hypertensive crises. This was a single-center, retrospective, chart review, cross-sectional study of patients with hypertensive crises. Patients were included if they were eighteen years of age or older, with an international classification disease ninth revision (ICD-9) code of 401.9 (hypertensive crises: emergency or urgency) and a documented magnesium level on their electronic medical record. The primary outcome of the study was the correlation between serum magnesium and blood pressure (systolic blood pressure and diastolic blood pressure) in patients with hypertensive crises. Two hundred and ninety-three patients were included in the study. The primary outcome result showed that serum magnesium was positively correlated with systolic blood pressure (r = 0.143, p = 0.014), but not diastolic blood pressure. Conclusion: This study found a significant positive association between magnesium and systolic blood pressure, but not diastolic blood pressure, among patients with hypertensive crises. This positive association of serum magnesium with systolic blood pressure was maintained after adjusting for covariates. This study's findings suggest a potential role of magnesium in blood pressure among patients with hypertensive crises.


Subject(s)
Blood Pressure/drug effects , Hypertension/blood , Magnesium/blood , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Diastole/drug effects , Female , Humans , Male , Middle Aged , Retrospective Studies , Systole/drug effects , Young Adult
5.
Bioengineered ; 12(2): 9367-9376, 2021 12.
Article in English | MEDLINE | ID: mdl-34787054

ABSTRACT

Cardiac dysfunction is a common complication of sepsis, and is attributed to severe inflammatory responses. Ferroptosis is reported to be involved in sepsis-induced cardiac inflammation. Therefore, we speculated that ferrostatin-1 (Fer-1), a ferroptosis inhibitor, improves cardiac dysfunction caused by sepsis. An intraperitoneal injection of lipopolysaccharide (LPS) was performed to induce a rat cardiac dysfunction model. Echocardiography, cardiac histopathology, biochemical and western blot results were analyzed. Twelve hours after the LPS injection, LPS-treated rats exhibited deteriorating cardiac systolic function, increased levels of cardiac injury markers and levels of ferroptosis markers prostaglandin endoperoxide synthase 2 (PTGS2). Additionally, LPS increased iron deposition in the myocardium, with downregulating ferroportin (FPN, SLC40A1) and transferrin receptor (TfR)expression, and upregulating ferritin light chain (FTL) and ferritin heavy chain (FTH1) expression. Meanwhile, LPS also increased lipid peroxidation in the rat heart by decreasing the expression of glutathione peroxidase 4 (GPX4). Moreover, the expression of inflammatory cytokines, such as tumor necrosis-alpha (TNF-α), interleukin-1 (IL-1ß), and interleukin-6 (IL-6), and inflammatory cell infiltration were also increased following LPS challenge. Finally, the abovementioned adverse effects of LPS were relieved by Fer-1 except for TfR expression. Mechanistically, Fer-1 significantly reduced the levels of toll-like receptor 4 (TLR4), phospho-nuclear factor kappa B (NF-κB), and phospho-inhibitor of kappa Bα (IκBα) in LPS-treated rats. In summary, these findings imply that Fer-1 improved sepsis-induced cardiac dysfunction at least partially via the TLR4/NF-κB signaling pathway.


Subject(s)
Cyclohexylamines/pharmacology , Heart/physiopathology , Phenylenediamines/pharmacology , Animals , Electrocardiography , Heart/diagnostic imaging , Heart/drug effects , Inflammation/complications , Inflammation/pathology , Inflammation/physiopathology , Lipopolysaccharides , Male , Myocardium/pathology , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Rats, Wistar , Sepsis/complications , Sepsis/physiopathology , Survival Analysis , Systole/drug effects , Toll-Like Receptor 4/metabolism
6.
BMC Cancer ; 21(1): 1223, 2021 Nov 13.
Article in English | MEDLINE | ID: mdl-34774000

ABSTRACT

BACKGROUND: To investigate risk factors for orally administered 5-aminolevulinic acid (ALA)-induced hypotension for bladder cancer patients receiving photodynamic diagnosis (PDD)-assisted transurethral resection of bladder tumor (TURBT). METHODS: Patients were categorized into two groups intraoperatively: a hypotensive group (minimum systolic blood pressure (SBP) ≤80 mmHg) and a non-hypotensive group (minimum SBP > 80 mmHg). We examined differences between the hypotensive group and non-hypotensive groups to identify clinical risk of ALA-induced hypotension using multivariate logistic regression analysis and decision tree analysis. RESULTS: Among 282 cases with ALA-PDD-assisted TURBT from three institutions who were screened, 245 patients were included in the final analysis. In total, 156 patients (63.7%) showed any grade of hypotension during ALA-PDD-assisted TURBT. General anesthesia and spinal anesthesia were induced intraoperatively in 113 patients (46.1%) and 132 patients (53.9%), respectively. Median SBP at baseline (before taking ALA) and at the beginning of anesthesia was 127 mmHg (range, 69-186 mmHg) and 124 mmHg (range, 69-186 mmHg), respectively. Median minimum SBP during ALA-PDD-assisted TURBT was 75 mmHg (range, 43-140 mmHg). Multivariate logistic regression analysis revealed that history of hypertension (odds ratio (OR) 7.568, p < 0.05) and general anesthesia (OR 14.435, p < 0.05) as significantly associated with an increased risk of hypotension incidence. Use of calcium antagonist showed significant negative associations with hypotension (OR 0.183, p < 0.05). Decision tree analysis showed presence of general anesthesia, age ≥ 74 years and American Society of Anesthesiologists physical status (ASA-PS) ≥2 as the most important discriminators. CONCLUSIONS: General anesthesia and hypertension were independent risk factors related to ALA-induced hypotension. In contrast, use of calcium antagonists was identified as a factor associated with reduced risk of ALA-induced hypotension.


Subject(s)
Aminolevulinic Acid/adverse effects , Cystectomy/methods , Hypotension/chemically induced , Photosensitizing Agents/adverse effects , Urinary Bladder Neoplasms/surgery , Aged , Aged, 80 and over , Anesthesia, General/adverse effects , Anesthesia, Spinal , Calcium Channel Blockers/therapeutic use , Decision Trees , Female , Humans , Hypotension/epidemiology , Incidence , Male , Middle Aged , Regression Analysis , Retrospective Studies , Risk Factors , Systole/drug effects , Urinary Bladder Neoplasms/diagnosis
7.
Oxid Med Cell Longev ; 2021: 1257283, 2021.
Article in English | MEDLINE | ID: mdl-34630844

ABSTRACT

Heart failure (HF) is a clinical syndrome caused by impairment of ventricular filling, ejection of blood, or both and is categorized as HF with reduced ejection fraction (HFrEF) or HF with preserved ejection fraction (HFpEF) based on left ventricular function. Cardiac fibrosis contributes to left ventricular dysfunction and leads to the development of HF. Ivabradine, an If current selective specific inhibitor, has been shown to improve the prognosis of patients with HF. However, the effects of ivabradine on cardiac function and fibrosis in HFpEF and HFrEF and the underlying mechanism remain unclear. In the present study, we utilized mouse models to mimic HFpEF and HFrEF and evaluated the therapeutic effects of ivabradine. By treating mice with different doses (10 mg/kg/d and 20 mg/kg/d) of ivabradine for 4 or 8 weeks, we found that a high dose of ivabradine improved cardiac diastolic function in HFpEF mice and ameliorated cardiac diastolic and systolic function and ventricular tachycardia incidence in HFrEF mice. Moreover, ivabradine significantly reduced the activation of cardiac fibroblasts and myocardial fibrosis in mice. Mechanistically, microRNA-133a, which was upregulated by ivabradine, targeted connective tissue growth factor and collagen 1 in cardiac fibroblasts and might contribute to the protective role of ivabradine. Together, our work utilized mouse models to study HFpEF and HFrEF, demonstrated the protective role of ivabradine in HFpEF and HFrEF, and elucidated the potential underlying mechanism, which provides an effective strategy for related diseases.


Subject(s)
Cardiotonic Agents/administration & dosage , Heart Failure/drug therapy , Heart Failure/metabolism , Ivabradine/administration & dosage , MicroRNAs/metabolism , Stroke Volume/drug effects , Up-Regulation/drug effects , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/metabolism , Animals , Animals, Newborn , Cells, Cultured , Diastole/drug effects , Disease Models, Animal , Fibroblasts/metabolism , Heart Ventricles/cytology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Systole/drug effects , Transfection , Treatment Outcome
8.
Int J Mol Sci ; 22(18)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34576081

ABSTRACT

Pulmonary hypertension (PH) is a progressive cardiovascular disorder in which local vascular inflammation leads to increased pulmonary vascular remodeling and ultimately to right heart failure. The HDAC inhibitor butyrate, a product of microbial fermentation, is protective in inflammatory intestinal diseases, but little is known regarding its effect on extraintestinal diseases, such as PH. In this study, we tested the hypothesis that butyrate is protective in a Sprague-Dawley (SD) rat model of hypoxic PH. Treatment with butyrate (220 mg/kg intake) prevented hypoxia-induced right ventricular hypertrophy (RVH), hypoxia-induced increases in right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, and permeability. A reversal effect of butyrate (2200 mg/kg intake) was observed on elevated RVH. Butyrate treatment also increased the acetylation of histone H3, 25-34 kDa, and 34-50 kDa proteins in the total lung lysates of butyrate-treated animals. In addition, butyrate decreased hypoxia-induced accumulation of alveolar (mostly CD68+) and interstitial (CD68+ and CD163+) lung macrophages. Analysis of cytokine profiles in lung tissue lysates showed a hypoxia-induced upregulation of TIMP-1, CINC-1, and Fractalkine and downregulation of soluble ICAM (sICAM). The expression of Fractalkine and VEGFα, but not CINC-1, TIMP-1, and sICAM was downregulated by butyrate. In rat microvascular endothelial cells (RMVEC), butyrate (1 mM, 2 and 24 h) exhibited a protective effect against TNFα- and LPS-induced barrier disruption. Butyrate (1 mM, 24 h) also upregulated tight junctional proteins (occludin, cingulin, claudin-1) and increased the acetylation of histone H3 but not α-tubulin. These findings provide evidence of the protective effect of butyrate on hypoxic PH and suggest its potential use as a complementary treatment for PH and other cardiovascular diseases.


Subject(s)
Butyrates/pharmacology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Lung/physiopathology , Pneumonia/physiopathology , Vascular Remodeling/drug effects , Acetylation/drug effects , Animals , Blood Pressure/drug effects , Cytokines/metabolism , Endothelial Cells/drug effects , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/physiopathology , Lung/blood supply , Lung/drug effects , Macrophages/drug effects , Macrophages/pathology , Microvessels/pathology , Pneumonia/complications , Rats, Sprague-Dawley , Systole/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , Up-Regulation/drug effects
9.
Hypertension ; 78(4): 946-954, 2021 09.
Article in English | MEDLINE | ID: mdl-34365808

ABSTRACT

Hypertension is a potent cardiovascular risk factor with deleterious end-organ effects and is especially prevalent among patients with chronic kidney disease. The SPRINT (Systolic Blood Pressure Intervention Trial) enrolled patients at an elevated cardiac risk including patients with mild to moderate chronic kidney disease and found that an intensive systolic blood pressure goal of <120 mm Hg significantly reduced the rates of adverse cardiovascular events and all-cause mortality and nonsignificantly reduced the rates of probable dementia; these results were consistent whether one had chronic kidney disease or not. However, results of intensive blood pressure therapy on chronic kidney disease progression were inconclusive, and there was an increased risk of incident chronic kidney disease and acute kidney injury, but the declines in kidney function appear to be hemodynamically driven and reversible. Overall, an intensive blood pressure target is effective in reducing cardiovascular disease and all-cause mortality and may reduce the risk of probable dementia in patients with mild to moderate chronic kidney disease. More studies are needed to determine its long-term effects on kidney function.


Subject(s)
Hypertension/drug therapy , Renal Insufficiency, Chronic/complications , Aged , Aged, 80 and over , Cardiovascular Diseases/etiology , Female , Glomerular Filtration Rate , Humans , Hypertension/etiology , Male , Middle Aged , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Systole/drug effects , Systole/physiology
11.
Int J Mol Sci ; 22(11)2021 May 21.
Article in English | MEDLINE | ID: mdl-34063987

ABSTRACT

The effects of the selective sodium-glucose cotransporter 2 (SGLT2) inhibitor empagliflozin in low dose on cardiac function were investigated in normoglycemic rats. Cardiac parameters were measured by intracardiac catheterization 30 min after intravenous application of empagliflozin to healthy animals. Empagliflozin increased the ventricular systolic pressure, mean pressure, and the max dP/dt (p < 0.05). Similarly, treatment with empagliflozin (1 mg/kg, p.o.) for one week increased the cardiac output, stroke volume, and fractional shortening (p < 0.05). Myocardial infarction (MI) was induced by ligation of the left coronary artery. On day 7 post MI, empagliflozin (1 mg/kg, p.o.) improved the systolic heart function as shown by the global longitudinal strain (-21.0 ± 1.1% vs. -16.6 ± 0.7% in vehicle; p < 0.05). In peri-infarct tissues, empagliflozin decreased the protein expression of matrix metalloproteinase 9 (MMP9) and favorably regulated the cardiac transporters sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) and sodium hydrogen exchanger 1 (NHE1). In H9c2 cardiac cells, empagliflozin decreased the MMP2,9 activity and prevented apoptosis. Empagliflozin did not alter the arterial stiffness, blood pressure, markers of fibrosis, and necroptosis. Altogether, short-term treatment with low-dose empagliflozin increased the cardiac contractility in normoglycemic rats and improved the systolic heart function in the early phase after MI. These effects are attributed to a down-regulation of MMP9 and NHE1, and an up-regulation of SERCA2a. This study is of clinical importance because it suggests that a low-dose treatment option with empagliflozin may improve cardiovascular outcomes post-MI. Down-regulation of MMPs could be relevant to many remodeling processes including cancer disease.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Matrix Metalloproteinase 9/metabolism , Myocardial Infarction/drug therapy , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Hydrogen Exchanger 1/metabolism , Systole/drug effects , Animals , Cells, Cultured , Down-Regulation/drug effects , Fibrosis/drug therapy , Fibrosis/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardial Infarction/metabolism , Myocardium/metabolism , Rats , Rats, Wistar , Up-Regulation/drug effects , Ventricular Function, Left , Ventricular Remodeling/drug effects
12.
Sci Rep ; 11(1): 13056, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158524

ABSTRACT

There has been interest in the use of nonintubated techniques for video-assisted thoracoscopic surgery (VATS) in both awake and sedated patients. The authors' centre developed a nonintubated technique with spontaneous ventilation for use in a patient under general anaesthesia using a phrenic nerve block. This treatment was compared with a case-matched control group. The authors believe that this technique is beneficial for optimizing anaesthesia for patients undergoing VATS. The patients were randomly allocated (1:1) to the phrenic nerve block (PNB) group and the control group. Both groups of patients received a laryngeal mask airway (LMA) that was inserted after anaesthetic induction, which permitted spontaneous ventilation and local anaesthesia in the forms of a paravertebral nerve block, a PNB and a vagal nerve block. However, the patients in the PNB group underwent procedures with 2% lidocaine, whereas saline was used in the control group. The primary outcome included the propofol doses. Secondary outcomes included the number of propofol boluses, systolic blood pressure (SBP), pH values of arterial blood gas and lactate (LAC), length of LMA pulled out, length of hospital stay (length of time from the operation to the time of discharge) and complications after 1 month. Intraoperatively, there were increases in lactate (F = 12.31, P = 0.001) in the PNB group. There was less propofol (49.20 ± 8.73 vs. 57.20 ± 4.12, P = 0.000), fewer propofol boluses (P = 0.002), a lower pH of arterial blood gas (F = 7.98, P = 0.006) and shorter hospital stays (4.10 ± 1.39 vs. 5.40 ± 1.22, P = 0.000) in the PNB group. There were no statistically significant differences in the length of the LMA pulled out, SBP or complications after 1 month between the groups. PNB optimizes the anaesthesia of nonintubated VATS.


Subject(s)
Nerve Block , Phrenic Nerve/surgery , Thoracic Surgery, Video-Assisted , Blood Pressure/drug effects , Double-Blind Method , Female , Humans , Hydrogen-Ion Concentration , Lactic Acid/blood , Male , Middle Aged , Postoperative Complications/etiology , Propofol/pharmacology , Systole/drug effects , Treatment Outcome
13.
High Blood Press Cardiovasc Prev ; 28(4): 383-391, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33950510

ABSTRACT

INTRODUCTION: Hypertensive mediated heart disease is the consequence of anatomical and functional changes in cardiovascular system. The benefits on left ventricular (LV) diastolic impairment and remodeling of hypertension treatment are well established. AIM: To evaluate LV structure, systolic and diastolic function of treated hypertensive patients on a medium to long term follow-up. METHODS: Prospectively observational cohort study. Hypertensive patients over 18 years, ultrasound evaluation of LV structure and diastolic and systolic function, follow-up at least once a year. Diastolic function assessed following recommendations of the American Society of Echocardiography and the European Association of Cardiovascular Imaging. RESULTS: 285 patients, mean follow up of 1731 ± 952 days. Sample mean age 56.3 ± 12.5 years, 166 patients (58.3%) were males. Baseline blood pressure 147.8 ± 19/86.8 ± 11 mm Hg, 5 years blood pressure 134.4 ± 15.7/79 ± 9 mm Hg (p < 0.005 SBP and p < 0.01 DBP). Baseline fixed dose combinations 115 patients (40.4%), follow-up 53.1% (p < 0.05). LV remodeling was detected in 88 patients (30.9%) vs. 30.1% at 5 years (p = NS). The frequency of an E/e' ratio > 14 was reduced from 38 patients (13.3%) to 3.6% (p < 0.001), e' septal velocity < 7 cm/sec or e' lateral velocity < 10 cm/sec was reduced from 38.6% (110 patients) to 19.3% (p < 0.001). Baseline normal diastolic function was detected in 85.6% (244 patients) and 94% at the end of the follow-up (p < 0.02). CONCLUSIONS: In this observational cohort followed by a mean of 5 years, the main benefit of hypertension treatment was the prevention or regression of diastolic dysfunction.


Subject(s)
Antihypertensive Agents/therapeutic use , Heart Diseases/etiology , Hypertension/complications , Hypertension/drug therapy , Adult , Aged , Blood Pressure/drug effects , Diastole/drug effects , Female , Follow-Up Studies , Heart Diseases/physiopathology , Heart Diseases/prevention & control , Humans , Hypertension/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Male , Middle Aged , Prospective Studies , Systole/drug effects , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control
14.
Eur J Pharmacol ; 904: 174170, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33984298

ABSTRACT

Diastolic dysfunction is a major feature of hypertrophic cardiomyopathy (HCM). Data from patient tissue and animal models associate increased Ca2+ sensitivity of myofilaments with altered Na+ and Ca2+ ion homeostasis in cardiomyocytes with diastolic dysfunction. In this study, we tested the acute effects of ouabain on ventricular myocytes of an HCM mouse model. The effects of ouabain on contractility and Ca2+ transients were tested in intact adult mouse ventricular myocytes (AMVMs) of Mybpc3-targeted knock-in (KI) and wild-type (WT) mice. Concentration-response assessment of contractile function revealed low sensitivity of AMVMs to ouabain (10 µM) compared to literature data on human cardiomyocytes (100 nM). Three hundred µM ouabain increased contraction amplitude (WT ~1.8-fold; KI ~1.5-fold) and diastolic intracellular Ca2+ in both WT and KI (+12-18%), but further decreased diastolic sarcomere length in KI cardiomyocytes (-5%). Western Blot analysis of whole heart protein extracts revealed 50% lower amounts of Na+/K+ ATPase (NKA) in KI than in WT. Ouabain worsened the diastolic phenotype of KI cardiomyocytes at concentrations which did not impair WT diastolic function. Ouabain led to an elevation of intracellular Ca2+, which was poorly tolerated in KI showing already high cytosolic Ca2+ at baseline due to increased myofilament Ca2+ sensitivity. Lower amounts of NKA in KI could amplify the need to exchange excessive intracellular Na+ for Ca2+ and thereby explain the general tendency to higher diastolic Ca2+ in KI.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Diastole/drug effects , Enzyme Inhibitors/toxicity , Myocytes, Cardiac/drug effects , Ouabain/toxicity , Sarcomeres/drug effects , Animals , Calcium/metabolism , Carrier Proteins/genetics , Disease Models, Animal , Gene Knock-In Techniques , Mice , Myocardial Contraction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Systole/drug effects
15.
J Cardiovasc Pharmacol ; 77(6): 805-813, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34001722

ABSTRACT

ABSTRACT: Levosimendan, a calcium sensitizer, exerts inotropic action through improving left ventricular ejection fraction. We noticed that only few clinical studies are published in which the effects of levosimendan on cardiac function are studied by echocardiography. When screening the literature (PubMed, Embase, and CENTRAL, from inception to August 2020), we found 29 randomized controlled trials on levosimendan containing echocardiographic data. We included those studies, describing a total of 574 heart failure patients, in our meta-analysis and extracted 14 ultrasonic parameters, pooling the effect estimates using a random-effect model. Our analysis of the diastolic parameters of the left ventricle shows that levosimendan reduce the early/late transmitral diastolic peak flow velocity ratio [standardized mean difference (SMD) -0.45 to 95% confidence interval (CI) (-0.87 to -0.03), P = 0.037] and E/e' (e': mitral annulus peak early diastolic wave velocity using tissue-doppler imaging) [SMD -0.59, 95% CI (-0.8 to -0.39), P < 0.001]. As it regards the systolic parameters of the right ventricle, levosimendan increased tricuspid annular plane systolic excursion [SMD 0.62, 95% CI (0.28 to 0.95), P < 0.001] and tricuspid annular peak systolic velocity [SMD 0.75, 95% CI (0.35 to 1.16), P < 0.001], and reduced systolic pulmonary artery pressure [SMD -1.02, 95% CI (-1.32, -0.73), P < 0.001]. As it regards the diastolic parameters of the right ventricle, levosimendan was associated with the decrease of Aa (peak late diastolic tricuspid annular velocity using tissue-doppler imaging) [SMD -0.38, 95% CI (-0.76 to 0), P = 0.047] and increase of Ea (peak early diastolic tricuspid annular velocity using tissue-doppler imaging) [SMD 1.03, 95% CI (0.63 to 1.42), P < 0.001] and Ea/Aa [SMD 0.86, 95% CI (0.18 to 1.54), P = 0.013]. We show that levosimendan is associated with an amelioration in the diastolic and systolic functions of both ventricles in heart failure patients.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Simendan/pharmacology , Diastole/drug effects , Echocardiography , Heart Failure/physiopathology , Humans , Randomized Controlled Trials as Topic , Stroke Volume/drug effects , Systole/drug effects , Ventricular Function, Left/drug effects , Ventricular Function, Right/drug effects
16.
Bioengineered ; 12(1): 1826-1837, 2021 12.
Article in English | MEDLINE | ID: mdl-33975512

ABSTRACT

Ivabradine (Iva), a heart rate reducing agent that specifically inhibits the pacemaker I(f) ionic current, has been demonstrated to be cardioprotective in many cardiovascular diseases. Autophagy is an evolutionarily conserved metabolic process that regulates cardiac homeostasis. This study is aimed to explore whether autophagy is functionally involved in the cardioprotective effect of Iva in a rat model of myocardial infarction (MI). We observed that Iva treatment (po, 10 mg/kg/day) showed significant recovery on the hemodynamics parameters in MI rats, including left ventricular systolic pressure, left ventricular end diastolic pressure, and maximal ascending/descending rate of left ventricular pressure. Also, Iva treatment dramatically decreased infarct size, inhibited myocardial apoptosis, and reduced the levels of pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6 in MI rats. Moreover, Iva treatment enhanced autophagy and inhibited PI3K/AKT/mTOR/p70S6K pathway in MI rats. Simultaneously, we observed that autophagy enhancer rapamycin (ip, 10 mg/kg/day) showed similar cardioprotective effects with Iva. Furthermore, we observed that addition of autophagy inhibitor 3-methyladenine (ip, 10 mg/kg/day) counteracted the therapeutic effect of Iva, addressing that Iva attenuated post-MI cardiac injury by enhancing autophagy. In summary, these findings demonstrated that Iva attenuated MI in rats by enhancing autophagy, and PI3K/AKT/mTOR/p70S6K pathway might be involved in the process. Autophagy activation by Iva may be a potential therapeutic strategy for the treatment of MI.


Subject(s)
Autophagy , Cardiotonic Agents/therapeutic use , Ivabradine/therapeutic use , Myocardial Infarction/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiotonic Agents/pharmacology , Cytokines/metabolism , Diastole/drug effects , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Inflammation Mediators/metabolism , Ivabradine/pharmacology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats, Sprague-Dawley , Signal Transduction , Systole/drug effects
17.
Am J Physiol Heart Circ Physiol ; 321(1): H175-H184, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34018850

ABSTRACT

Inorganic nitrite is a source of nitric oxide (NO) and is considered as a potential therapy in settings where endogenous NO bioactivity is reduced and left ventricular (LV) function impaired. However, the effects of nitrite on human cardiac contractile function, and the extent to which these are direct or indirect, are unclear. We studied 40 patients undergoing diagnostic cardiac catheterization who had normal LV systolic function and were not found to have obstructive coronary disease. They received either an intracoronary sodium nitrite infusion (8.7-26 µmol/min, n = 20) or an intravenous sodium nitrite infusion (50 µg/kg/min, n = 20). LV pressure-volume relations were recorded. The primary end point was LV end-diastolic pressure (LVEDP). Secondary end points included indices of LV systolic and diastolic function. Intracoronary nitrite infusion induced a significant reduction in LVEDP, LV end-diastolic pressure-volume relationship (EDPVR), and the time to LV end-systole (LVEST) but had no significant effect on LV systolic function or systemic hemodynamics. Intravenous nitrite infusion induced greater effects, with significant decreases in LVEDP, EDPVR, LVEST, LV dP/dtmin, tau, and mean arterial pressure. Inorganic nitrite has modest direct effects on human LV diastolic function, independent of LV loading conditions and without affecting LV systolic properties. However, the systemic administration of nitrite has larger effects on LV diastolic function, which are related to reduction in both preload and afterload. These contractile effects of inorganic nitrite may indicate a favorable profile for conditions characterized by LV diastolic dysfunction.NEW & NOTEWORTHY This is the first study to assess the direct and indirect effects of inorganic nitrite on invasive measures of left ventricular function in humans in vivo. Inorganic nitrite has a modest direct myocardial effect, improving diastolic function. Systemic administration of nitrite has larger effects related to alterations in cardiac preload and afterload. The changes induced by nitrite appear favorable for potential use in conditions characterized by LV diastolic dysfunction.


Subject(s)
Myocardial Contraction/drug effects , Sodium Nitrite/administration & dosage , Systole/drug effects , Ventricular Function, Left/drug effects , Aged , Female , Hemodynamics/drug effects , Humans , Male , Middle Aged , Stroke Volume/drug effects
18.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802928

ABSTRACT

Blood platelets' adenosine receptors (AR) are considered to be a new target for the anti-platelet therapy. This idea is based on in vitro studies which show that signaling mediated by these receptors leads to a decreased platelet response to activating stimuli. In vivo evidence for the antithrombotic activity of AR agonists published to date were limited, however, to the usage of relatively high doses given in bolus. The present study was aimed at verifying if these substances used in lower doses in combination with inhibitors of P2Y12 could serve as components of dual anti-platelet therapy. We have found that a selective A2A agonist 2-hexynyl-5'-N-ethylcarboxamidoadenosine (HE-NECA) improved the anti-thrombotic properties of either cangrelor or prasugrel in the model of ferric chloride-induced experimental thrombosis in mice. Importantly, HE-NECA was effective not only when applied in bolus as other AR agonists in the up-to-date published studies, but also when given chronically. In vitro thrombus formation under flow conditions revealed that HE-NECA enhanced the ability of P2Y12 inhibitors to decrease fibrinogen content in thrombi, possibly resulting in their lower stability. Adenosine receptor agonists possess a certain hypotensive effect and an ability to increase the blood-brain barrier permeability. Therefore, the effects of anti-thrombotic doses of HE-NECA on blood pressure and the blood-brain barrier permeability in mice were tested. HE-NECA applied in bolus caused a significant hypotension in mice, but the effect was much lower when the substance was given in doses corresponding to that obtained by chronic administration. At the same time, no significant effect of HE-NECA was observed on the blood-brain barrier. We conclude that chronic administration of the A2A agonist can be considered a potential component of a dual antithrombotic therapy. However, due to the hypotensive effect of the substances, dosage and administration must be elaborated to minimize the side-effects. The total number of animals used in the experiments was 146.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Adenosine-5'-(N-ethylcarboxamide)/analogs & derivatives , Antithrombins/pharmacology , Fibrinogen/metabolism , Prasugrel Hydrochloride/pharmacology , Purinergic P1 Receptor Agonists/pharmacology , Thrombosis/metabolism , Adenosine Monophosphate/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Adult , Animals , Blood Pressure/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Chlorides , Diastole/drug effects , Female , Ferric Compounds , Humans , Laser-Doppler Flowmetry , Male , Mice, Inbred C57BL , Permeability/drug effects , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Systole/drug effects
19.
Biochem Biophys Res Commun ; 556: 207-214, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33848935

ABSTRACT

Bortezomib is a classical proteasome inhibitor and previous researches have reported its roles of anti-oxidation and anti-inflammatory functions in various diseases. However, the role of Bortezomib in myocardial ischemia reperfusion injury (MIRI) is unclear. Thus, our research seeks to reveal the protective effects of Bortezomib pretreatment in the mice model of MIRI. First, by the optimization of Bortezomib concentration and pretreatment timepoints, we found that 0.5 mg/kg Bortezomib pretreatment 2 h before MIRI significantly attenuated pathological damage and neutrophil infiltration. Then we found that pretreatment with Bortezomib obviously increased myocardial systolic function ((left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS)) and decreased infarct size, as well as serum Troponin T levels. Meanwhile, Bortezomib pretreatment also remarkably augmented oxidative stress related protein levels of Superoxide dismutase [Cu-Zn] (SOD1), Catalase (CAT) and Glutathione (GSH), while reactive oxygen species (ROS) contents and Malonaldehyde (MDA) protein level were significantly reduced. Mechanistically, Bortezomib pretreatment significantly promoted nuclear translocation of transcriptional factor nuclear factor erythroid 2-related factor 2(Nrf2) and Heme Oxygenase 1(HO-1) expression. Interestingly, co-treatment with ML-385, a new type and selective Nrf2 inhibitor, counteracted antioxidative effects induced by Bortezomib pretreatment. In conclusion, Bortezomib pretreatment mitigates MIRI by inhibiting oxidative damage which is regulated by Nrf2/HO-1 signaling pathway.


Subject(s)
Bortezomib/pharmacology , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Myocardial Reperfusion Injury/prevention & control , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Animals , Bortezomib/administration & dosage , Bortezomib/therapeutic use , Disease Models, Animal , Drug Administration Schedule , Heart/drug effects , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Systole/drug effects , Time Factors , Troponin T/blood , Ventricular Function/drug effects
20.
Tissue Cell ; 72: 101550, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33915356

ABSTRACT

Acute stress is a feature of our daily events that affects cardiovascular system and predisposes to hypertension. H2S is now considered as a vasorelaxant gasotransmitter although it was considered as a toxic agent. In present work we studied the effect of H2S releasing Na2S in acute stress induced hypertension and cardiac damage. Rats were divided into five groups: control, Na2S, acute stress, half dose of Na2S (6 mg/kg), and finally full dose of Na2S (12 mg/kg) to acute stressed rats. BP was measured then blood samples were taken for estimation of cortisol, cardiac enzymes markers, IL-6 and H2S. Finally, animals were sacrificed, hearts and thoracic aortae were excised for histological assessment, estimation of MDA, SOD and RNA extraction of CSE. Acute stress significantly elevated BP, cortisol, cardiac enzymes markers, IL-6, and tissue levels of MDA. It also, induced cardiac cell damage with congested B.V., extravasation of blood and decreased eNOs. Moreover, acute stress reduced H2S levels, RNA expression of CSE and SOD in cardiac tissues. Na2S significantly decreased BP, serum levels of cortisol, cardiac enzymes markers, IL-6, and tissue levels of MDA. Also, Na2S elevated serum H2S, RNA expression of CSE, SOD in cardiac tissue and increased eNOs activity.


Subject(s)
Hydrogen Sulfide/pharmacology , Hypertension/enzymology , Hypertension/etiology , Nitric Oxide Synthase Type III/metabolism , Protective Agents/pharmacology , Stress, Psychological/complications , Sulfides/pharmacology , Animals , Aorta, Thoracic/pathology , Biomarkers/metabolism , Blood Pressure/drug effects , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Hydrocortisone/metabolism , Hypertension/blood , Interleukin-6/blood , Interleukin-6/metabolism , Myocardium/enzymology , Oxidative Stress , Rats , Stress, Psychological/blood , Systole/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...