Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
1.
Naunyn Schmiedebergs Arch Pharmacol ; 395(10): 1257-1268, 2022 10.
Article in English | MEDLINE | ID: mdl-35798982

ABSTRACT

6-Nitrodopamine (6-ND) is an endogenous modulator of the contractility in the rat isolated epididymal vas deferens (RIEVD) and considered to be the main peripheral mediator of the emission process. Use of selective and unselective ß-adrenergic receptor antagonists has been associated with ejaculatory failure. Here, the effects of selective ß1- and ß1/ß2-adrenergic receptor antagonists on RIEVD contractions induced by 6-ND, dopamine, noradrenaline, adrenaline, and electric-field stimulation (EFS) were investigated. The selective ß1-adrenergic receptor antagonists atenolol (0.1 and 1 µï»¿M), betaxolol (1 µï»¿M), and metoprolol (1 µï»¿M) and the unselective ß1/ß2-adrenergic receptor antagonists propranolol (1 and 10 µï»¿M) and pindolol (10 µï»¿M) caused significant rightward shifts of the concentration-response curve to 6-ND (pA2 6.41, 6.91, 6.75, 6.47, and 5.74; for atenolol, betaxolol, metoprolol, propranolol, and pindolol), but had no effect on dopamine-, noradrenaline-, and adrenaline-induced contractions. The effects of selective ß1- and ß1/ß2-adrenergic receptor antagonists at a higher concentration (atenolol 1 µï»¿M, betaxolol 1 µï»¿M, metoprolol 1 µï»¿M, propranolol 10 µï»¿M, and pindolol 10 µï»¿M) also reduced the EFS-induced RIEVD contractions in control, but not in RIEVD obtained from L-NAME-treated animals. The selective ß1-adrenoceptor agonist RO-363, the selective ß2-adrenoceptor agonist salbutamol, and the selective ß3-adrenoceptor agonist mirabegron, up to 300 µï»¿M, had no effect on the RIEVD tone. The results demonstrate that ß1- and ß1-/ß2-adrenoceptor receptor antagonists act as 6-ND receptor antagonists in RIEVD, further confirming the main role of 6-ND in the RIEVD contractility.


Subject(s)
Propranolol , Vas Deferens , Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Atenolol/pharmacology , Betaxolol/pharmacology , Dopamine/analogs & derivatives , Epinephrine/pharmacology , Male , Metoprolol/pharmacology , Norepinephrine/pharmacology , Pindolol/pharmacology , Propranolol/pharmacology , Rats
2.
Sci Rep ; 12(1): 8852, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35614155

ABSTRACT

Renewal of the myocardium by preexisting cardiomyocytes is a powerful strategy for restoring the architecture and function of hearts injured by myocardial infarction. To advance this strategy, we show that combining two clinically approved drugs, but neither alone, muscularizes the heart through cardiomyocyte proliferation. Specifically, in adult murine cardiomyocytes, metoprolol, a cardioselective ß1-adrenergic receptor blocker, when given with triiodothyronine (T3, a thyroid hormone) accentuates the ability of T3 to stimulate ERK1/2 phosphorylation and proliferative signaling by inhibiting expression of the nuclear phospho-ERK1/2-specific phosphatase, dual-specificity phosphatase-5. While short-duration metoprolol plus T3 therapy generates new heart muscle in healthy mice, in mice with myocardial infarction-induced left ventricular dysfunction and pathological remodeling, it remuscularizes the heart, restores contractile function and reverses chamber dilatation; outcomes that are enduring. If the beneficial effects of metoprolol plus T3 are replicated in humans, this therapeutic strategy has the potential to definitively address ischemic heart failure.


Subject(s)
Myocardial Infarction , Ventricular Dysfunction, Left , Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Animals , Metoprolol/pharmacology , Metoprolol/therapeutic use , Mice , Myocardial Infarction/pathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Triiodothyronine/metabolism , Triiodothyronine/pharmacology , Ventricular Dysfunction, Left/pathology , Ventricular Remodeling
3.
Crit Care Med ; 50(9): e707-e718, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35234431

ABSTRACT

OBJECTIVES: Although cardiovascular benefits of ß 1 -adrenergic receptor blockade have been described in sepsis, little is known about its impact on the adaptive immune response, specifically CD4 T cells. Herein, we study the effects of ß 1 -adrenergic receptor modulation on CD4 T-cell function in a murine model of sepsis. DESIGN: Experimental study. SETTING: University laboratory. SUBJECTS: C57BL/6 mice. INTERVENTIONS: High-grade sepsis was induced by cecal ligation and puncture in wild-type mice (ß 1+/+ ) with or without esmolol (a selective ß 1 -adrenergic receptor blocker) or in ß 1 -adrenergic receptor knockout mice (ß 1-/- ). At 18 hours after surgery, echocardiography was performed with blood and spleen collected to analyze lymphocyte function. MEASUREMENTS AND MAIN RESULTS: At 18 hours, ß 1+/+ cecal ligation and puncture mice exhibited characteristics of high-grade sepsis and three surrogate markers of immunosuppression, namely decreased splenic CD4 T cells, reduced CD4 T-cell proliferation, and increased regulatory T lymphocyte cell proportions. Pharmacologic and genetic ß 1 -adrenergic receptor blockade reversed the impact of sepsis on CD4 T and regulatory T lymphocyte proportions and maintained CD4 T-cell proliferative capacity. ß 1 -adrenergic receptor blocked cecal ligation and puncture mice also exhibited a global decrease in both pro- and anti-inflammatory mediators and improved in vivo cardiovascular efficiency with maintained cardiac power index despite the expected decrease in heart rate. CONCLUSIONS: ß 1 -adrenergic receptor activation enhances regulatory T lymphocyte inhibitory function and thus contributes to sepsis-induced immunosuppression. This can be attenuated by ß 1 -adrenergic receptor blockade, suggesting a potential immunoregulatory role for this therapy in the management of sepsis.


Subject(s)
Sepsis , T-Lymphocytes, Regulatory , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Disease Models, Animal , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Sepsis/drug therapy
4.
Cardiovasc Toxicol ; 22(2): 99-107, 2022 02.
Article in English | MEDLINE | ID: mdl-34800264

ABSTRACT

Cardiomyocyte senescence is involved in the pathological mechanism of cardiac diseases. Metoprolol is a ß1 receptor blocker used for the treatment of hypertension. Recent studies show that Metoprolol can protect cardiomyocytes against ischemia injury. The present study aims to investigate the protective effects of Metoprolol against arginine vasopressin (AVP)-induced cellular senescence in cultured cardiomyocytes. The cell proliferation assay and cytotoxicity lactate dehydrogenase assay showed that the highest tolerated dosage of Metoprolol in H9C2 cardiomyocytes was optimized as 10 µM. The enzyme-linked immunosorbent assay showed that Metoprolol significantly ameliorated the elevated level of the DNA oxidation product 8-hydroxy-2 deoxyguanosine. Metoprolol also decreased the percentage of senescence-associated ß-galactosidase positive cells and improved the telomerase activity under AVP exposure. Moreover, treatment with Metoprolol ameliorated the decreased intracellular nicotinamide phosphoribosyltransferase activity, nicotinamide adenine dinucleotide/nicotinamide adenine dinucleotide phosphate (NAD+/NADPH) ratio, and Sirtuin1 activity in cardiomyocytes by AVP. Finally, Metoprolol was able to downregulate the AVP-induced expression of acetylated p53 and p21. Taken together, our data reveal that Metoprolol protected the cardiomyocytes from AVP-induced senescence.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Arginine Vasopressin/toxicity , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Metoprolol/pharmacology , Myocytes, Cardiac/drug effects , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Acetylation , Animals , Cell Line , Cytokines/metabolism , DNA Damage/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NAD/metabolism , NADP/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidative Stress/drug effects , Protein Processing, Post-Translational , Rats , Signal Transduction , Telomerase/metabolism
5.
J Am Coll Cardiol ; 78(25): 2505-2517, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34915981

ABSTRACT

BACKGROUND: The use of ß-adrenergic receptor blocking agents in symptomatic patients with obstructive hypertrophic cardiomyopathy (HCM) rests on clinical experience and observational cohort studies. OBJECTIVES: This study aimed to investigate the effects of metoprolol on left ventricular outflow tract (LVOT) obstruction, symptoms, and exercise capacity in patients with obstructive HCM. METHODS: This double-blind, placebo-controlled, randomized crossover trial enrolled 29 patients with obstructive HCM and New York Heart Association (NYHA) functional class II or higher symptoms from May 2018 to September 2020. Patients received metoprolol or placebo for 2 consecutive 2-week periods in random order. The effect parameters were LVOT gradients, NYHA functional class, Canadian Cardiovascular Society (CCS) angina class, Kansas City Cardiomyopathy Questionnaire Overall Summary Score (KCCQ-OSS), and cardiopulmonary exercise testing. RESULTS: Compared with placebo, the LVOT gradient during metoprolol was lower at rest (25 mm Hg [interquartile range (IQR): 15-58 mm Hg] vs 72 mm Hg [IQR: 28-87 mm Hg]; P = 0.007), at peak exercise (28 mm Hg [IQR: 18-40 mm Hg] vs 62 mm Hg [IQR: 31-113 mm Hg]; P < 0.001), and postexercise (45 mm Hg [IQR: 24-100 mm Hg] vs 115 mm Hg [IQR: 55-171 mm Hg]; P < 0.0001). During metoprolol treatment, 14% of patients were in NYHA functional class III or higher compared with 38% of patients receiving placebo (P < 0.01). Similarly, no patients were in CCS class III or higher during metoprolol treatment compared with 10% during placebo treatment (P < 0.01). These findings were confirmed by higher KCCQ-OSS during metoprolol treatment (76.2 ± 16.2 vs 73.8 ± 19.5; P = 0.039). Measures of exercise capacity, peak oxygen consumption, and N-terminal pro-B-type natriuretic peptide did not differ between the study arms. CONCLUSIONS: Compared with placebo, metoprolol reduced LVOT obstruction at rest and during exercise, provided symptom relief, and improved quality of life in patients with obstructive HCM. Maximum exercise capacity remained unchanged. (The Effect of Metoprolol in Patients with Hypertrophic Obstructive Cardiomyopathy [TEMPO]; NCT03532802).


Subject(s)
Adrenergic beta-1 Receptor Antagonists/therapeutic use , Cardiomyopathy, Hypertrophic/drug therapy , Metoprolol/therapeutic use , Ventricular Outflow Obstruction/drug therapy , Adrenergic beta-1 Receptor Antagonists/pharmacology , Aged , Cardiomyopathy, Hypertrophic/complications , Cross-Over Studies , Double-Blind Method , Exercise Tolerance/drug effects , Female , Humans , Male , Metoprolol/pharmacology , Middle Aged , Ventricular Outflow Obstruction/etiology
6.
Biomed Pharmacother ; 142: 112069, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34470728

ABSTRACT

ß-blockers are commonly prescribed to treat multiple cardiovascular (CV) diseases, but, frequently, adverse drug reactions and intolerance limit their use in clinical practice. Interindividual variability in response to ß-blockers may be explained by genetic differences. In fact, pharmacogenetic interactions for some of these drugs have been widely studied, such as metoprolol. But studies that explore genetic variants affecting bisoprolol response are inconclusive, limited or confusing because of mixed results with other ß-Blockers, different genetic polymorphisms observed, endpoint studied etc. Because of this, we performed a systematic review in order to find relevant genetic variants affecting bisoprolol response. We have found genetic polymorphism in several genes, but most of the studies focused in ADRB variants. The ADRB1 Arg389Gly (rs1801253) was the most studied genetic polymorphism and it seems to influence the response to bisoprolol, although studies are inconclusive. Even, we performed a meta-analysis about its influence on systolic/diastolic blood pressure in patients treated with bisoprolol, but this did not show statistically significant results. In conclusion, many genetic polymorphisms have been assessed about their influence on patients´ response to bisoprolol and the ADRB1 Arg389Gly (rs1801253) seems the most relevant genetic polymorphism in this regard but results have not been confirmed with a meta-analysis. Our results support the need of further studies about the impact of genetic variants on bisoprolol response, considering different genetic polymorphisms and conducting single and multiple SNPs analysis, including other clinical parameters related to bisoprolol response in a multivariate study.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Bisoprolol/pharmacology , Pharmacogenetics , Blood Pressure/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Humans , Polymorphism, Single Nucleotide , Receptors, Adrenergic, beta-1/genetics , Treatment Outcome
7.
Pharmacol Rep ; 73(4): 1179-1187, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34117630

ABSTRACT

BACKGROUND: Evidence indicates that Gα12, Gα13, and its downstream effectors, RhoA and Rac1, regulate neuronal morphology affected by stress. This study was aimed at investigating whether repeated stress influences the expression of proteins related to the Gα12/13 intracellular signaling pathway in selected brain regions sensitive to the effects of stress. Furthermore, the therapeutic impact of ß(1)adrenergic receptors (ß1AR) blockade was assessed. METHODS: Restraint stress (RS) model in mice (2 h/14 days) was used to assess prolonged stress effects on the mRNA expression of Gα12, Gα13, RhoA, Rac1 in the prefrontal cortex (PFC), hippocampus (HIP) and amygdala (AMY). In a separate study, applying RS model in rats (3-4 h/1 day or 14 days), we evaluated stress effects on the expression of Gα12, Gα11, Gαq, RhoA, RhoB, RhoC, Rac1/2/3 in the HIP. Betaxolol (BET), a selective ß1AR antagonist, was introduced (5 mg/kg/p.o./8-14 days) in the rat RS model to assess the role of ß1AR in stress effects. RT-qPCR and Western Blot were used for mRNA and protein assessments, respectively. RESULTS: Chronic RS decreased mRNA expression of Gα12 and increased mRNA for Rac1 in the PFC of mice. In the mice AMY, decreased mRNA expression of Gα12, Gα13 and RhoA was observed. Fourteen days of RS exposure increased RhoA protein level in the rats' HIP in the manner dependent on ß1AR activity. CONCLUSIONS: Together, these results suggest that repeated RS affects the expression of genes and proteins known to be engaged in neural plasticity, providing potential targets for further studies aimed at unraveling the molecular mechanisms of stress-related neuropsychiatric diseases.


Subject(s)
Brain/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Neurons/metabolism , Restraint, Physical/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Brain/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction/drug effects
8.
Circulation ; 143(25): 2431-2440, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33993702

ABSTRACT

BACKGROUND: Adjuvant breast cancer therapy containing anthracyclines with or without anti-human epidermal growth factor receptor-2 antibodies and radiotherapy is associated with cancer treatment-related cardiac dysfunction. In the PRADA trial (Prevention of Cardiac Dysfunction During Adjuvant Breast Cancer Therapy), concomitant treatment with the angiotensin receptor blocker candesartan attenuated the reduction in left ventricular ejection fraction (LVEF) in women receiving treatment for breast cancer, whereas the ß-blocker metoprolol attenuated the increase in cardiac troponins. This study aimed to assess the long-term effects of candesartan and metoprolol or their combination to prevent a reduction in cardiac function and myocardial injury. METHODS: In this 2×2 factorial, randomized, placebo-controlled, double-blind, single-center trial, patients with early breast cancer were assigned to concomitant treatment with candesartan cilexetil, metoprolol succinate, or matching placebos. Target doses were 32 and 100 mg, respectively. Study drugs were discontinued after adjuvant therapy. All 120 validly randomized patients were included in the intention-to-treat analysis. The primary outcome measure was change in LVEF assessed by cardiovascular magnetic resonance imaging from baseline to extended follow-up. Secondary outcome measures included changes in left ventricular volumes, echocardiographic peak global longitudinal strain, and circulating cardiac troponin concentrations. RESULTS: A small decline in LVEF but no significant between-group differences were observed from baseline to extended follow-up, at a median of 23 months (interquartile range, 21 to 28 months) after randomization (candesartan, 1.7% [95% CI, 0.5 to 2.8]; no candesartan, 1.8% [95% CI, 0.6 to 3.0]; metoprolol, 1.6% [95% CI, 0.4 to 2.7]; no metoprolol, 1.9% [95% CI, 0.7 to 3.0]). Candesartan treatment during adjuvant therapy was associated with a significant reduction in left ventricular end-diastolic volume compared with the noncandesartan group (P=0.021) and attenuated decline in global longitudinal strain (P=0.046) at 2 years. No between-group differences in change in cardiac troponin I and T concentrations were observed. CONCLUSIONS: Anthracycline-containing adjuvant therapy for early breast cancer was associated with a decline in LVEF during extended follow-up. Candesartan during adjuvant therapy did not prevent reduction in LVEF at 2 years, but was associated with modest reduction in left ventricular end-diastolic volume and preserved global longitudinal strain. These results suggest that a broadly administered cardioprotective approach may not be required in most patients with early breast cancer without preexisting cardiovascular disease. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT01434134.


Subject(s)
Benzimidazoles/therapeutic use , Biphenyl Compounds/therapeutic use , Breast Neoplasms/therapy , Chemoradiotherapy, Adjuvant/adverse effects , Heart Diseases/prevention & control , Metoprolol/therapeutic use , Tetrazoles/therapeutic use , Ventricular Function, Left/drug effects , Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Adult , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Benzimidazoles/pharmacology , Biphenyl Compounds/pharmacology , Breast Neoplasms/diagnostic imaging , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cross-Over Studies , Double-Blind Method , Female , Follow-Up Studies , Heart Diseases/chemically induced , Heart Diseases/diagnostic imaging , Humans , Metoprolol/pharmacology , Middle Aged , Stroke Volume/drug effects , Stroke Volume/physiology , Tetrazoles/pharmacology , Ventricular Function, Left/physiology
9.
Am J Physiol Heart Circ Physiol ; 320(6): H2295-H2304, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33861148

ABSTRACT

Extreme preterm birth conveys an elevated risk of heart failure by young adulthood. Smaller biventricular chamber size, diastolic dysfunction, and pulmonary hypertension may contribute to reduced ventricular-vascular coupling. However, how hemodynamic manipulations may affect right ventricular (RV) function and coupling remains unknown. As a pilot study, 4D flow MRI was used to assess the effect of afterload reduction and heart rate reduction on cardiac hemodynamics and function. Young adults born premature were administered sildenafil (a pulmonary vasodilator) and metoprolol (a ß blocker) on separate days, and MRI with 4D flow completed before and after each drug administration. Endpoints include cardiac index (CI), direct flow fractions, and ventricular kinetic energy including E/A wave kinetic energy ratio. Sildenafil resulted in a median CI increase of 0.24 L/min/m2 (P = 0.02), mediated through both an increase in heart rate (HR) and stroke volume. Although RV ejection fraction improved only modestly, there was a significant increase (4% of end diastolic volume) in RV direct flow fraction (P = 0.04), consistent with hemodynamic improvement. Metoprolol administration resulted in a 5-beats/min median decrease in HR (P = 0.01), a 0.37 L/min/m2 median decrease in CI (P = 0.04), and a reduction in time-averaged kinetic energy (KE) in both ventricles (P < 0.01), despite increased RV diastolic E/A KE ratio (P = 0.04). Despite reduced right atrial workload, metoprolol significantly depressed overall cardiac systolic function. Sildenafil, however, increased CI and improved RV function, as quantified by the direct flow fraction. The preterm heart appears dependent on HR but sensitive to RV afterload manipulations.NEW & NOTEWORTHY We assessed the effect of right ventricular afterload reduction with sildenafil and heart rate reduction with metoprolol on cardiac hemodynamics and function in young adults born premature using 4D flow MRI. Metoprolol depressed cardiac function, whereas sildenafil improved cardiac function including right ventricular direct flow fraction by 4D flow, consistent with hemodynamic improvement. This suggests that the preterm heart is dependent on heart rate and sensitive to right ventricular afterload changes.


Subject(s)
Sildenafil Citrate/pharmacology , Vasodilator Agents/pharmacology , Ventricular Function, Right/drug effects , Adrenergic beta-1 Receptor Antagonists/pharmacology , Adult , Female , Follow-Up Studies , Heart Rate , Hemodynamics , Humans , Imaging, Three-Dimensional , Infant, Extremely Premature , Infant, Newborn , Infant, Premature , Infant, Very Low Birth Weight , Magnetic Resonance Imaging , Magnetic Resonance Imaging, Cine , Male , Metoprolol/pharmacology , Pilot Projects , Stroke Volume
10.
Glycoconj J ; 38(2): 261-271, 2021 04.
Article in English | MEDLINE | ID: mdl-33687640

ABSTRACT

Amongst a wide range of biological macromolecules, saccharides exhibit the potential to be specifically recognized by cell-surface receptors and hence can be utilized as ligands in targeted drug delivery. The current study aims to use saccharides viz. Galactose, Pectin and Chitosan to improve targeting of Atenolol by oxalyl chloride mediated grafting. Conjugates were engineered by grafting Atenolol, a cardiovascular agent with the modified saccharide units. The conjugates were characterized by FTIR, DSC and 1H NMR study. Drug release analysis and cellular uptake study was carried out using H9c2 cell lines which represent that concentration of drug in cells treated with all atenolol-saccharide conjugates is enhanced by almost two-folds in comparison with cells treated with atenolol solution. Thus cell line study confers the evidence of selective cardiac delivery. No significant cytotoxicity was observed in case of all synthesized conjugates in the Brine shrimp lethality bioassay. Possible binding of the developed conjugates with the GLUT-4 receptors was assessed by in silico analysis using homology model developed by Swiss Model server. Hence it was concluded that the application of these conjugates with saccharides in selective cardiovascular drug delivery can be a promising approach to increase bioavailability, minimize drug loss by degradation and prevent harmful side effects by increasing specific cell targeting.


Subject(s)
Atenolol/chemistry , Atenolol/pharmacology , Drug Carriers/chemistry , Adrenergic beta-1 Receptor Antagonists/chemistry , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Artemia/drug effects , Atenolol/metabolism , Atenolol/toxicity , Cell Line , Chitosan/chemistry , Computer Simulation , Drug Carriers/pharmacology , Drug Carriers/toxicity , Drug Evaluation, Preclinical , Galactose/chemistry , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/metabolism , Lethal Dose 50 , Magnetic Resonance Spectroscopy , Myocardium/cytology , Pectins/chemistry , Rats , Spectroscopy, Fourier Transform Infrared
11.
Eur J Appl Physiol ; 121(5): 1499-1511, 2021 May.
Article in English | MEDLINE | ID: mdl-33646423

ABSTRACT

PURPOSE: Habitual endurance exercise results in increased erythropoiesis, which is primarily controlled by erythropoietin (EPO), yet studies demonstrating upregulation of EPO via a single bout of endurance exercise have been equivocal. This study compares the acute EPO response to 30 min of high versus 90 min of moderate-intensity endurance exercise and whether that response can be upregulated via selective adrenergic receptor blockade. METHODS: Using a counterbalanced, cross-over design, fifteen participants (age 28 ± 8) completed two bouts of running (30-min, high intensity vs 90-min, moderate intensity) matched for overall training stress. A separate cohort of fourteen participants (age 31 ± 6) completed three bouts of 30-min high-intensity cycling after ingesting the preferential ß1-adrenergic receptor (AR) antagonist bisoprolol, the non-preferential ß1 + ß2 antagonist nadolol or placebo. Venous blood was collected before, during, and after exercise, and serum EPO levels were determined by ELISA. RESULTS: No detectable EPO response was observed during or after high intensity running, however, in the moderate-intensity trial EPO was significantly elevated at both during-exercise timepoints (+ 6.8% ± 2.3% at 15 min and + 8.7% ± 2.2% at 60 min). No significant change in EPO was observed post-cycling or between the trials involving ßAR blockade. CONCLUSION: Neither training mode (running or cycling), nor beta-blockade significantly influenced the EPO response to 30 min of high-intensity exercise, however, 90 min of moderate-intensity running elevated EPO during exercise, returning to baseline immediately post-exercise. Identifying the optimal mode, duration and intensity required to evoke an EPO response to exercise may help tailor exercise prescriptions designed to maximize EPO response for both performance and clinical applications.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Bisoprolol/pharmacology , Erythropoietin/metabolism , Nadolol/pharmacology , Physical Endurance/physiology , Adult , Bicycling/physiology , Cross-Over Studies , Female , Humans , Male , Running/physiology , Up-Regulation
12.
Nat Commun ; 12(1): 105, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397973

ABSTRACT

Environmental triggers have important functions in multiple sclerosis (MS) susceptibility, phenotype, and trajectory. Exposure to early life trauma (ELT) has been associated with higher relapse rates in MS patients; however, the underlying mechanisms are not well-defined. Here we show ELT induces mechanistic and phenotypical alterations during experimental autoimmune encephalitis (EAE). ELT sustains downregulation of immune cell adrenergic receptors, which can be attributed to chronic norepinephrine circulation. ELT-subjected mice exhibit interferon-ß resistance and neurodegeneration driven by lymphotoxin and CXCR2 involvement. These phenotypic changes are observed in control EAE mice treated with ß1 adrenergic receptor antagonist. Conversely, ß1 adrenergic receptor agonist treatment to ELT mice abrogates phenotype changes via restoration of immune cell ß1 adrenergic receptor function. Our results indicate that ELT alters EAE phenotype via downregulation of ß1 adrenergic signaling in immune cells. These results have implications for the effect of environmental factors in provoking disease heterogeneity and might enable prediction of long-term outcomes in MS.


Subject(s)
Down-Regulation , Interferon-beta/metabolism , Multiple Sclerosis/complications , Nerve Degeneration/complications , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction , Stress, Psychological/complications , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Biomarkers/metabolism , Brain/immunology , Brain/pathology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Golgi Apparatus/metabolism , Male , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Nerve Degeneration/blood , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Norepinephrine/blood , Phenotype , Severity of Illness Index , Up-Regulation/drug effects
13.
Clin Sci (Lond) ; 135(2): 327-346, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33480422

ABSTRACT

A high salt intake exacerbates insulin resistance, evoking hypertension due to systemic perivascular inflammation, oxidative-nitrosative stress and endothelial dysfunction. Angiotensin-converting enzyme inhibitor (ACEi) and angiotensin receptor blockers (ARBs) have been shown to abolish inflammation and redox stress but only partially restore endothelial function in mesenteric vessels. We investigated whether sympatho-adrenal overactivation evokes coronary vascular dysfunction when a high salt intake is combined with insulin resistance in male Goto-Kakizaki (GK) and Wistar rats treated with two different classes of ß-blocker or vehicle, utilising synchrotron-based microangiography in vivo. Further, we examined if chronic carvedilol (CAR) treatment preserves nitric oxide (NO)-mediated coronary dilation more than metoprolol (MET). A high salt diet (6% NaCl w/w) exacerbated coronary microvessel endothelial dysfunction and NO-resistance in vehicle-treated GK rats while Wistar rats showed modest impairment. Microvascular dysfunction was associated with elevated expression of myocardial endothelin, inducible NO synthase (NOS) protein and 3-nitrotyrosine (3-NT). Both CAR and MET reduced basal coronary perfusion but restored microvessel endothelium-dependent and -independent dilation indicating a role for sympatho-adrenal overactivation in vehicle-treated rats. While MET treatment reduced myocardial nitrates, only MET treatment completely restored microvessel dilation to dobutamine (DOB) stimulation in the absence of NO and prostanoids (combined inhibition), indicating that MET restored the coronary flow reserve attributable to endothelium-derived hyperpolarisation (EDH). In conclusion, sympatho-adrenal overactivation caused by high salt intake and insulin resistance evoked coronary microvessel endothelial dysfunction and diminished NO sensitivity, which were restored by MET and CAR treatment in spite of ongoing inflammation and oxidative-nitrosative stress presumably caused by uninhibited renin-angiotensin-aldosterone system (RAAS) overactivation.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carvedilol/pharmacology , Endothelium, Vascular/drug effects , Insulin Resistance , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Coronary Angiography , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Hypertension/physiopathology , Male , Metoprolol/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Wistar , Sodium Chloride, Dietary/administration & dosage
14.
Cells ; 11(1)2021 12 24.
Article in English | MEDLINE | ID: mdl-35011613

ABSTRACT

Hyperglycemia-induced impairment of the blood-retinal barrier represents the main pathological event in diabetic retinopathy that is elicited by a reduced cellular response to an accumulation of reactive oxygen species (ROS) and increased inflammation. The purpose of the study was to evaluate whether the selective ß1-adrenoreceptor (ß1-AR) antagonist metoprolol could modulate the inflammatory response to hyperglycemic conditions. For this purpose, human retinal endothelial cells (HREC) were treated with normal (5 mM) or high glucose (25 mM, HG) in the presence of metoprolol (10 µM), epinephrine (1 µM), or both compounds. Metoprolol prevented both the HG-induced reduction of cell viability (MTT assays) and the modulation of the angiogenic potential of HREC (tube formation assays) reducing the TNF-α, IL-1ß, and VEGF mRNA levels (qRT-PCR). Moreover, metoprolol prevented the increase in phospho-ERK1/2, phospho-cPLA2, COX2, and protein levels (Western blot) as well as counteracting the translocation of ERK1/2 and cPLA2 (high-content screening). Metoprolol reduced ROS accumulation in HG-stimulated HREC by activating the anti-oxidative cellular response mediated by the Keap1/Nrf2/HO-1 pathway. In conclusion, metoprolol exerted a dual effect on HG-stimulated HREC, decreasing the activation of the pro-inflammatory ERK1/2/cPLA2/COX2 axis, and counteracting ROS accumulation by activating the Keap1/Nrf2/HO-1 pathway.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Endothelial Cells/pathology , Glucose/toxicity , Metoprolol/pharmacology , Microvessels/pathology , Retina/pathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epinephrine/pharmacology , Heme Oxygenase-1/metabolism , Humans , Interleukin-1beta/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/metabolism , Neovascularization, Physiologic/drug effects , Phospholipases A2, Cytosolic/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
J Pharmacol Exp Ther ; 376(1): 127-135, 2021 01.
Article in English | MEDLINE | ID: mdl-33100271

ABSTRACT

The practice of prescribing ß-blockers to lower blood pressure and mitigate perioperative cardiovascular events has been questioned because of reports of an increased risk of stroke. The benefit of ß-blocker therapy primarily relies on preventing activation of cardiac ß1-adrenergic receptors (ARs). However, we reported that ß1ARs also mediate vasodilator responses of rat cerebral arteries (CAs), implying that ß-blockers may impair cerebral blood flow under some conditions. Here, we defined the impact of metoprolol (MET), a widely prescribed ß1AR-selective antagonist, on adrenergic-elicited diameter responses of rat CAs ex vivo and in vivo. MET (1-10 µmol/l) prevented ß1AR-mediated increases in diameter elicited by dobutamine in cannulated rat CAs. The ß1AR-mediated dilation elicited by the endogenous adrenergic agonist norepinephrine (NE) was reversed to a sustained constriction by MET. Acute oral administration of MET (30 mg/kg) to rats in doses that attenuated resting heart rate and dobutamine-induced tachycardia also blunted ß1AR-mediated dilation of CAs. In the same animals, NE-induced dilation of CAs was reversed to sustained constriction. Administration of MET for 2 weeks in drinking water (2 mg/ml) or subcutaneously (15 mg/kg per day) also resulted in NE-induced constriction of CAs in vivo. Thus, doses of MET that protect the heart from adrenergic stimulation also prevent ß1AR-mediated dilation of CAs and favor anomalous adrenergic constriction. Our findings raise the possibility that the increased risk of ischemic stroke in patients on ß-blockers relates in part to adrenergic dysregulation of cerebrovascular tone. SIGNIFICANCE STATEMENT: ß-Blocker therapy using second-generation, cardioselective ß-blockers is associated with an increased risk of stroke, but the responsible mechanisms are unclear. Here, we report that either acute or chronic systemic administration of a cardioselective ß-blocker, metoprolol, mitigates adrenergic stimulation of the heart as an intended beneficial action. However, metoprolol concomitantly eliminates vasodilator responses to adrenergic stimuli of rat cerebral arteries in vivo as a potential cause of dysregulated cerebral blood flow predisposing to ischemic stroke.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Cardiotonic Agents/pharmacology , Cerebral Arteries/drug effects , Metoprolol/pharmacology , Receptors, Adrenergic, beta-1/metabolism , Vasodilation , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Adrenergic beta-1 Receptor Antagonists/adverse effects , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/adverse effects , Cerebral Arteries/physiology , Dobutamine/pharmacology , Heart Rate/drug effects , Male , Metoprolol/administration & dosage , Metoprolol/adverse effects , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley
16.
Clin Radiol ; 76(3): 236.e21-236.e25, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33298312

ABSTRACT

AIM: To evaluate the use and safety of intravenous (IV) metoprolol in a cohort of patients undergoing coronary computed tomographic angiography (CCTA) at a university hospital, and in particular, to establish if the minimum dose required to achieve the target heart rate (HR) in a given patient can be predicted from the baseline HR. MATERIALS AND METHODS: Patients undergoing CCTA at a tertiary centre between January 2015 and May 2018, with baseline HR ≥60 bpm requiring IV metoprolol, were identified retrospectively from the database. Patients with a contraindication to beta-blockade or an indication for CCTA other than coronary disease were excluded. HR at baseline and at the time of scanning were recorded, together with the total dose of IV metoprolol administered. RESULTS: Of 625 patients identified, 330 (52.8%) achieved HR ≤60 with IV metoprolol. Patients who achieved target HR had lower baseline HR. They received a lower radiation exposure due to tight prospective gating and a lower tube voltage. The lower quartile dose of metoprolol administered was 5 mg for patients with baseline HR <65 beats per minute (bpm), but 10 mg for HR 65-74 bpm, and ≥20 mg for higher HRs. There were no cases of symptomatic bradycardia/hypotension. CONCLUSION: Patients with a resting HR of ≥60 bpm can reasonably be given an initial minimum dose of 5-20 mg metoprolol IV before CCTA, with additional doses as required.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Computed Tomography Angiography/methods , Coronary Angiography/methods , Coronary Artery Disease/diagnostic imaging , Heart Rate/drug effects , Metoprolol/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Retrospective Studies , Time
17.
Surgery ; 169(4): 885-893, 2021 04.
Article in English | MEDLINE | ID: mdl-33303271

ABSTRACT

BACKGROUND: Open abdominal surgery is frequently related to excessive inflammation and a compromised intestinal barrier, leading to poor clinical outcomes. The administration of beta-1 blocker has been shown to effectively reduce inflammation and preserve intestinal barrier function in patients with sepsis, shock, or other critical illnesses. The underlying mechanism of these effects may be associated with the autonomic nervous system's activation via cholecystokinin receptors. This study aimed to investigate the effect of beta-1 blocker on systemic and local inflammatory responses and the intestinal barrier function in the context of open abdominal surgery. METHODS: A rat model of open abdominal surgery was induced through peritoneal air exposure for 3 hours and treated via gavage with the beta-1 blocker, metoprolol, or saline. Cholecystokinin-receptor antagonists were administered before the metoprolol treatment. Peritoneal lavage fluid, serum, and tissues were collected 24 hours after surgery to determine systemic and local inflammation and intestinal integrity. RESULTS: The intervention with metoprolol significantly reduced serum tumor necrosis factor-alpha and interleukin-6 (P < .05) and peritoneal interleukin-6 (P < .01) compared with those of animals treated with saline. The intestinal myeloperoxidase indicating the influx of neutrophils was also significantly prevented by the administration of metoprolol (P < .05). Above all, this intervention resulted in a significant decrease in serum D-lactate and intestinal fatty acid-binding protein, intestinal permeability, bacterial translocation, and Chiu's score for intestinal mucosa injury (P < .05). However, the anti-inflammatory and intestinal integrity protective effects of metoprolol were prevented by the blockage of cholecystokinin receptors (P < .05). CONCLUSION: Our data indicate that beta-1 blocker reduces systemic and local inflammatory responses and preserves intestinal barrier function after open abdominal surgery through a mechanism that depends on cholecystokinin receptors. Clinically, these findings imply that perioperative intervention with a beta-1 blocker may be an effective new therapy to enhance recovery after open abdominal surgery.


Subject(s)
Abdomen/surgery , Adrenergic beta-1 Receptor Antagonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Intestinal Mucosa/drug effects , Postoperative Care , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Cytokines/blood , Cytokines/metabolism , Digestive System Surgical Procedures/adverse effects , Disease Models, Animal , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Metoprolol/pharmacology , Permeability , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Rats
18.
J Clin Invest ; 131(3)2021 02 01.
Article in English | MEDLINE | ID: mdl-33301422

ABSTRACT

Propranolol, a pleiotropic ß-adrenergic blocker, has been anecdotally reported to reduce cerebral cavernous malformations (CCMs) in humans. However, propranolol has not been rigorously evaluated in animal models, nor has its mechanism of action in CCM been defined. We report that propranolol or its S(-) enantiomer dramatically reduced embryonic venous cavernomas in ccm2 mosaic zebrafish, whereas R-(+)-propranolol, lacking ß antagonism, had no effect. Silencing of the ß1, but not ß2, adrenergic receptor mimicked the beneficial effects of propranolol in a zebrafish CCM model, as did the ß1-selective antagonist metoprolol. Thus, propranolol ameliorated cavernous malformations by ß1 adrenergic antagonism in zebrafish. Oral propranolol significantly reduced lesion burden in 2 chronic murine models of the exceptionally aggressive Pdcd10/Ccm3 form of CCM. Propranolol or other ß1-selective antagonists may be beneficial in CCM disease.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/adverse effects , Hemangioma, Cavernous, Central Nervous System , Propranolol/pharmacology , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Female , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Hemangioma, Cavernous, Central Nervous System/chemically induced , Hemangioma, Cavernous, Central Nervous System/drug therapy , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
19.
Cell Stress Chaperones ; 25(6): 993-1012, 2020 11.
Article in English | MEDLINE | ID: mdl-32779001

ABSTRACT

The adoptive transfer of donor-derived virus-specific T cells (VSTs) is an effective treatment for infections following allogeneic hematopoietic cell transplantation. Acute exercise mobilizes effector lymphocytes and VSTs to the circulation and augments the ex vivo manufacture of VSTs. This study determined if ß2 adrenergic receptor (AR) signaling precipitated the VST response to acute exercise. Healthy participants (n = 12) completed 30 min of steady-state cycling exercise after ingesting a placebo, a ß1 + 2 AR antagonist (nadolol) or a ß1 AR antagonist (bisoprolol). Circulating VSTs to cytomegalovirus (CMV), Epstein-Barr virus (EBV), and adenovirus (AdV) antigens were enumerated before and after exercise, and peripheral blood mononuclear cells were cultured with viral peptides for 8 days to expand multi-VSTs. Compared with placebo, nadolol blunted the exercise-induced mobilization of CMV-VSTs (Δ VSTs/100,000 CD3+ T cells = 93 ± 104 vs. 22 ± 91 for placebo and nadolol, respectively; p = 0.036), while bisoprolol did not, despite both drugs evoking similar reductions in exercising heart rate and blood pressure. Circulating AdV and EBV VSTs (VSTs/mL blood) only increased after exercise with placebo. Although not significant, nadolol partially mitigated exercise-induced increases in multi-VST expansion, particularly in participants that demonstrated an exercise-induced increase in VST expansion. We conclude that exercise-induced enhancements in VST mobilization and expansion are at least partially ß2 AR mediated, thus highlighting a role for the ß2 AR in targeted therapy for the augmentation of VST immune cell therapeutics in the allogeneic adoptive transfer setting. Moreover, long-term regular exercise may provide additional viral protection in the host through frequent ß2 AR-dependent mobilization and redistribution of VSTs cumulated with each bout of exercise.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Antiviral Agents/pharmacology , Cell- and Tissue-Based Therapy , Exercise , T-Lymphocytes/immunology , Viruses/immunology , Adult , Blood Pressure/drug effects , Catecholamines/blood , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Heart Rate/drug effects , Humans , Hydrocortisone/blood , Lactic Acid/blood , Male , Peptides/pharmacology , Phenotype , Receptors, Adrenergic, beta/metabolism , Species Specificity , T-Lymphocytes/drug effects , Young Adult
20.
Article in English | MEDLINE | ID: mdl-32598313

ABSTRACT

Background In previous studies, we have observed that glutamate antagonists injected within the nucleus accumbens septi (NAS) induced an anxiolytic-like effect in the elevated plus maze (EPM) test in rats. In the present study, the effect of Atenolol, a specific Beta Adreno-receptor antagonist in the EPM was studied in male rats bilaterally cannulated NAS. Methods Rats were divided into five groups that received either 1 µL injections of saline or atenolol in different doses (0.75, 1 or 2 µg/1 µL, n=15-16) 15 min before testing. Results Time Spent in the Open Arm was modified by treatment (F=4.563, p=0.006, df 3). This was increased by the lowest dose of atenolol (p<0.05), by the medium doses (p<0.001) and also by the highest dose (p<0.01). Time per Entry was modified by treatment (F=4.54, p=0.06, df 3). This parameter was increased by the lowest dose of atenolol (p<0.01), but not for the medium and higher doses. Conclusions We conclude that Atenolol beta receptor blockade in the accumbens lead to an anxiolytic-like effect related to an increase in the time spent in the open arm and in the time per entry, showing specific behavioral patterns.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Anti-Anxiety Agents/pharmacology , Atenolol/pharmacology , Nucleus Accumbens/metabolism , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Animals , Anti-Anxiety Agents/administration & dosage , Atenolol/administration & dosage , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Elevated Plus Maze Test , Male , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...