Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 784
Filter
1.
Environ Toxicol ; 39(6): 3425-3433, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38450887

ABSTRACT

Recent reports indicate a potential oncogenic role of antihypertensive drugs in common cancers. However, it remains uncertain whether this phenomenon influences the risk of glioblastoma multiforme (GBM). This study aimed to assess the potential causal effects of blood pressure (BP) and antihypertensive drugs on GBM. Genome-wide association study (GWAS) summary statistics for systolic blood pressure (SBP), diastolic blood pressure (DBP), and GBM in Europeans were downloaded. To represent the effects of antihypertensive drugs, we utilized single nucleotide polymorphisms (SNPs) associated with SBP/DBP adjacent to the coding regions of different antihypertensive drugs as instrumental variables to model five antihypertensive drugs, including angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, calcium channel blockers, ß-receptor blockers (BBs), and thiazide diuretics. Positive control studies were performed using GWAS data in chronic heart failure. The primary method for causality estimation was the inverse-variance-weighted method. Mendelian randomization analysis showed that BBs with the ß1-adrenergic receptor (ADRB1) as a therapeutic target could significantly reduce the risk of GBM by mediating DBP (OR = 0.431, 95% CI: 0.267-0.697, p < .001) and that they could also significantly reduce the risk of GBM by mediating SBP (OR = 0.595, 95% CI: 0.422-0.837, p = .003). Sensitivity analysis and colocalization analysis reinforced the robustness of these findings. Finally, the low expression of the ADRB1 gene in malignant gliomas was found by GBM data from TCGA and single-cell RNA sequencing, which most likely contributed to the poor prognosis of GBM patients. In summary, our study provides preliminary evidence of some causal relationship between ADRB1-targeted BBs and glioblastoma development. However, more studies are needed to validate these findings and further reveal the complex relationship between BP and GBM.


Subject(s)
Antihypertensive Agents , Genome-Wide Association Study , Glioblastoma , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Receptors, Adrenergic, beta-1 , Glioblastoma/genetics , Glioblastoma/drug therapy , Humans , Antihypertensive Agents/therapeutic use , Receptors, Adrenergic, beta-1/genetics , Quantitative Trait Loci , Blood Pressure/drug effects , Sequence Analysis, RNA , Single-Cell Analysis , Adrenergic beta-Antagonists/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/drug therapy
2.
Wiad Lek ; 77(1): 105-113, 2024.
Article in English | MEDLINE | ID: mdl-38431814

ABSTRACT

OBJECTIVE: Aim: To analyze the role of cytokines in the progression of heart failure (HF) in patients with concomitant pathology of the thyroid gland. PATIENTS AND METHODS: Materials and Methods: The systematization of literature data on the role of cytokines in the progression of HF in patients with concomitant thyroid pathology (TP) was carried out. The results of our own research were presented. CONCLUSION: Conclusions: The final chapter in the history of the role of cytokines in the progression of HF has not yet been written. Further studies, including genetic ones, are necessary. The patients with HF have higher levels of TNFß and IL-6, and a lower concentration of IL-4, compared to the control group. Patients with a fatal outcome of the disease, in contrast to those who survived for two years, have an increased level of TNFß. In patients with concomitant TP, who had repeated hospitalization, a lower level was registered, compared to that under conditions of a more favorable course of heart failure. Concentrations of cytokines in the blood of patients with HF are associated with gene polymorphisms of the ß-adrenoreceptor system: the C-allele of the Gly389A polymorphism of the ß1-adrenoceptor gene leads to a decrease in the risk of increasing TNFα; IL-1α increases in the presence of the A-allele of the Ser49Gly polymorphism of this gene. In patients with HF and concomitant thyroid pathology, the risk of IL-6 growth increases in homozygous (C) patients for the Ser275 polymorphism of the ß3 subunit of the G-protein.


Subject(s)
Heart Failure , Thyroid Gland , Humans , Cytokines/genetics , Interleukin-6/genetics , Receptors, Adrenergic, beta-1/genetics , Polymorphism, Genetic , Heart Failure/genetics
4.
Medicina (Kaunas) ; 59(12)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38138160

ABSTRACT

Background and Objectives: Tachycardia is a common cardiovascular disease. Drugs blocking ß1-adrenergic receptors (ADRB1) are used in the therapy of arrhythmogenic heart diseases. Disease-related polymorphisms can be observed within the ADRB1 gene. The two most important are Ser49Gly and Arg389Gly, and they influence the treatment efficacy. The family of the cytochrome P450 system consists of the isoenzyme CYP2D6 (Debrisoquine 4-hydroxylase), which is involved in phase I metabolism of almost 25% of clinically important drugs, including antiarrhythmic drugs. A study was conducted to detect the ADRB1 and CYP2D6 gene polymorphisms. Materials and Methods: The material for the test was whole blood from 30 patients with ventricular and supraventricular tachycardia and 20 controls. The samples were obtained from the Department of Pediatric Cardiology. The first to be made was the extraction of DNA using a GeneMATRIX Quick Blood DNA Purification Kit from EURx. The selected ADRB1 and CYP2D6 gene polymorphisms were detected by high-resolution melting polymerase chain reaction (HRM-PCR) analysis. Results: Based on the analysis of melt profile data for each PCR product, the identification of polymorphisms was carried out. Heterozygotes and homozygotes were found in the examined alleles. Conclusions: The frequency of the Arg389Gly polymorphism differs statistically significantly between the control group and patients with supraventricular and ventricular arrhythmias, as well as between these two groups of patients. Moreover, the Arg389Gly polymorphism was statistically more prevalent in the group of girls with SVT arrhythmia compared to girls with VT. A few carriers of homozygous and heterozygous systems of the S49G polymorphism were detected among patients with arrhythmias, as well as control group. The percentage of individuals carrying the CYP2D6 4 allele as either homozygous or heterozygous was observed in the study and control groups. The high prevalence of the CYP2D6*4 allele carriers in both groups prompts the optimization of beta-1 blocker therapy.


Subject(s)
Adrenergic beta-Antagonists , Cytochrome P-450 CYP2D6 , Child , Female , Humans , Adrenergic beta-Antagonists/therapeutic use , Arrhythmias, Cardiac/genetics , Cytochrome P-450 CYP2D6/genetics , DNA , Polymorphism, Genetic/genetics , Receptors, Adrenergic, beta-1/genetics
5.
Biomed Pharmacother ; 169: 115869, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37952358

ABSTRACT

Betablockers (BBs) are prescribed for ischaemia in patients with acute coronary syndrome (ACS). In Spain, bisoprolol and carvedilol are the most prescribed BBs, but patients often had to discontinue them due to adverse effects. Single nucleotide polymorphisms (SNPs) in ADRB1, ADRB2 and CYP2D6 genes have strong evidence of pharmacogenetic association with BBs in heart failure or hypertension, but the evidence in ACS is limited. Therefore, our study focuses on investigating how these genes influence the response to BBs in ACS patients. We analysed the association between SNPs in ADRB1 Gly389Arg (rs1801253) and Ser49Gly (rs1801252), ADRB2 Gly16Arg (rs1042713) and Glu27Gln (rs1042714), and CYP2D* 6 (*2- rs1080985, *4- rs3892097, *10 - rs1065852) and the occurrence of bradycardia/hypotension events during one year of follow-up. We performed an observational study and included 285 ACS-PCI-stent patients. A first analysis including patients treated with bisoprolol and a second analysis including patients treated with other BBs were performed. We found that the presence of the G allele (Glu) of the ADRB2 gene (rs1042714; Glu27Gln) conferred a protective effect against hypotension-induced by BBs; OR (CI 95%) = 0,14 (0,03-0,60), p < 0.01. The ADRB2 (rs1042713; Gly16Arg) GG genotype could also prevent hypotensive events; OR (CI 95%) = 0.49 (0.28-0.88), p = 0015. SNPs in ADRB1 and CYP2D6 * 2, CYP2D6 * 4 weren´t associated with primary events. The effect of CYP2D6 * 10 does not seem to be relevant for the response to BBs. According to our findings, SNPs in ADRB2 (rs1042713, rs1042714) could potentially affect the response and tolerance to BBs in ACS-patients. Further studies are necessary to clarify the impact of ADRB2 polymorphisms.


Subject(s)
Acute Coronary Syndrome , Hypotension , Percutaneous Coronary Intervention , Humans , Cytochrome P-450 CYP2D6/genetics , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/genetics , Bisoprolol/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Genotype , Polymorphism, Single Nucleotide/genetics , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics
6.
Wiad Lek ; 76(9): 2054-2060, 2023.
Article in English | MEDLINE | ID: mdl-37898944

ABSTRACT

OBJECTIVE: The aim: To study the association of left ventricular hypertrophy (LVH) and polymorphisms rs1801253 and rs1801252 of the ADRB1 gene with the risk of sudden cardiac death (SCD). PATIENTS AND METHODS: Materials and methods: The study included 179 patients which underwent clinical investigation, echocardiography, elektrokardiography. The examined were divided into groups with a low (110 people) and high risk (69 people) of SCD. The distribution of allelic polymorphisms was investigated with polymerase chain reaction (PCR). RESULTS: Results: All patients of group with high-risk cardiovascular mortality showed a decrease in heart rate variability (RV) due to an increase in sympathetic activity (p=0.013). Also, in the group of patients with LVH, predictors of sudden cardiac death and arrhythmogenic substrate, were observed. The variability of the allele C1165G rs1801253 of the ADRB1 gene was associated with an increased risk (2.55-fold increase) of SCD and LVH. Also, the associations of polymorphic locus A145G (rs1801252) of the ADRB1 gene proved the presence of a permanent difference for the "risky" allele A in patients with a high risk of SCD. CONCLUSION: Conclusions: It was set the probable association of alleles rs1801253 (C1165G) and rs1801252 (A145G) ADRB1 at the patients with a high risk of SCD compared to the control group.


Subject(s)
Hypertension , Hypertrophy, Left Ventricular , Humans , Alleles , Hypertrophy, Left Ventricular/genetics , Hypertension/complications , Hypertension/genetics , Polymorphism, Genetic , Death, Sudden, Cardiac/etiology , Risk Factors , Receptors, Adrenergic, beta-1/genetics
7.
Cancer Discov ; 13(12): OF12, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37772780

ABSTRACT

The ß1-adrenergic receptor is a T-cell immune checkpoint in chronic infection and in cancer.


Subject(s)
Adrenergic Agents , T-Cell Exhaustion , Humans , Signal Transduction , CD8-Positive T-Lymphocytes , Receptors, Adrenergic, beta-1/genetics
8.
Life Sci ; 322: 121644, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37004731

ABSTRACT

Altered sensitivity to the chronotropic and inotropic effects of catecholamines and reduction in ß1/ß2-adrenoceptor (ß1/ß2-AR) ratio were reported in failing and in senescent human heart, as well as in isolated atria and ventricle of rats submitted to stress. This was due to downregulation of ß1-AR with or without up-regulation of ß2-AR. AIMS: To investigate the stress-induced behavior of ß1-AR in the heart of mice expressing a non-functional ß2-AR subtype. The guiding hypothesis is that the absence of ß2-AR signaling will not affect the behavior of ß1-AR during stress and that those are independent processes. MATERIALS AND METHODS: The chronotropic and inotropic responses to ß-AR agonists in isolated atria of stressed mice expressing a non-functional ß2-AR were analyzed. The mRNA and protein expressions of ß1- and ß2-AR were also determined. KEY FINDINGS: No deaths were observed in mice under stress protocol. Atria of stressed mice displayed reduced sensitivity to isoprenaline compared to the controls, an effect that was abolished by the ß2- and ß1-AR antagonists 50 nM ICI118,551 and 300 nM CGP20712A, respectively. Sensitivity and maximum response to the ß-agonists dobutamine and salbutamol were not altered by stress or ICI118,551. The responses to dobutamine and salbutamol were prevented by CGP20712A. The expression of ß1-AR was reduced at protein levels. SIGNIFICANCE: Collectively, our data provide evidence that the cardiac ß2-AR is not essential for survival in a stressful situation and that the stress-induced reduction of ß1-AR expression was independent of the ß2-AR presence.


Subject(s)
Adrenergic beta-Agonists , Dobutamine , Humans , Mice , Rats , Animals , Dobutamine/pharmacology , Dobutamine/metabolism , Adrenergic beta-Agonists/pharmacology , Heart Atria/metabolism , Receptors, Adrenergic, beta-2/metabolism , Isoproterenol/pharmacology , Isoproterenol/metabolism , Albuterol/pharmacology , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/metabolism
9.
Eur J Pharmacol ; 948: 175700, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37001579

ABSTRACT

Downregulation of cell surface ß-adrenergic receptors (ß-AR) is an important adaptive response that prevents deleterious effects of receptor overstimulation. Various factors including reactive oxygen species cause ß-AR downregulation. In this study, we evaluated the effects of ligands of the peripheral benzodiazepine receptor (PBR), a key protein in regulating oxidative stress, on surface density of endogenous ß1-and ß2-ARs in highly differentiated cells such as human monocytes, which express both ß-AR subtypes. ß-AR expression in human monocytes was evaluated by flow cytometry, qPCR and western blotting. Monocyte treatment with ß-AR agonist isoproterenol did not change surface ß1-AR density while downregulating surface ß2-AR density. This effect was antagonized by the ß-blocker propranolol. An opposite response was observed with benzodiazepine diazepam that led to a time-dependent reduction in ß1-AR density. In particular, while no significant downregulation was observed after 3 h of treatment, only 63% of ß1-ARs were still present on the cell surface after 48 h of treatment with diazepam at 1 µM. Treatment with the PBR antagonist PK11195, but not with propranolol, antagonized the effects of diazepam. No change in ß1-AR-mRNA or protein levels was observed at any time after diazepam treatment. We also found that diazepam did not affect Gs-protein or ß-arrestin-2 recruitment for both ß-ARs in engineered fibroblasts, further suggesting that diazepam activity on ß1-AR density is mediated by PBR. Finally, no sex-related differences were found. Collectively, these results indicate that monocyte ß1-ARs are resistant to catecholamine-mediated downregulation and suggest that PBR plays an important role in regulating ß1-AR density.


Subject(s)
Monocytes , Propranolol , Humans , Monocytes/metabolism , Propranolol/pharmacology , Benzodiazepines , Diazepam/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism
11.
Gene ; 844: 146820, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-35987510

ABSTRACT

ß1-adrenergic receptors (ß1-AR) directly affect on intracardiac hemodynamic and the ability of the heart to tolerate physical activity by regulating its inotropic and chronotropic functions. Severe hypersympathicotonia, specific to coronary artery disease (CAD) and chronic heart failure (HF), leads to impaired functioning of ß1-AR. The aim of this research was to assess the expression level of the ß1-AR ADRB1 gene in the myocardium, to evaluate the ß-adrenergic reactivity of the membrane (ß-ARM) of erythrocytes, and to analyze the association of these parameters with myocardial contractile dysfunction in patients with a myocardial infarction (MI) in the past and without it. The study included 126 patients with chronic CAD. Among the patients, 55.6 % had a history of MI at least 6 months ago. The expression of the ADRB1 gene was assessed using real-time polymerase chain reaction. With this purpose, we isolated RNA from the right atrial appendage, which was excised when a heart-lung machine was connected during a planned coronary bypass surgery. ß-ARM was evaluated in 57 patients. This method is based on the fact of inhibition of hemolysis of erythrocytes, placed in a hyposmotic medium, in the presence of a ß-blocker. Within the whole sample of patients, the expression of the ADRB1 gene is comparable in different functional classes of HF. There was no linear correlation between the expression of the ADRB1 gene and left ventricle ejection fraction (LVEF). In patients with a history of MI, the expression of the ADRB1 gene was elevated when compared to a group of patients without MI (p = 0.017). Patients with a history of MI had higher values of ß-ARM than those without MI (p = 0.017). The reverse correlation between ß-ARM and LVEF (r = -0,570, p = 0,002) was revealed in the group of patients without MI but not in the group of patients with a history of MI (r = -0,137, p = 0,479). In the sample of patients with chronic CAD, in the myocardium of subjects with a history of MI, the relative expression of ADRB1 gene was higher compared to the group of patients without MI. In patients with different functional classes (FC) of HF and with different ejection fraction, both with MI and without it, ADRB1 gene expression was comparable. In the group of patients with a history of MI, an increase in ß-ARM was observed, i.e. decrease in the number or sensitivity of ß-AR. Among patients without MI, an inverse correlation was found between ß-ARM and LVEF.


Subject(s)
Cardiomyopathies , Myocardial Infarction , Adrenergic Agents , Adrenergic beta-Antagonists , Humans , Myocardial Infarction/genetics , Myocardium , Receptors, Adrenergic, beta-1/genetics , Stroke Volume
12.
J Cardiovasc Pharmacol ; 80(3): 334-341, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35881897

ABSTRACT

ABSTRACT: Adrenergic receptors are critical regulators of cardiac function with profound effects on cardiac output during sympathetic stimulation. Chronic stimulation of the adrenergic system of the heart under conditions of cardiac stress leads to cardiac dysfunction, hypertrophy, and ultimately failure. Emerging data have revealed that G protein-coupled receptors in intracellular compartments are functionally active and regulate distinct cellular processes from those at the cell surface. ß2 adrenergic receptors internalize onto endosomes in various cell types where they have recently been shown to continue to stimulate cAMP production to selectively regulate gene expression. Other studies have identified ß1 adrenergic receptors at the nuclear envelope and the Golgi apparatus. Here, we discuss data on signaling by ß1 and ß2 adrenergic receptors in the heart and the possible influence of their subcellular locations on their divergent physiological functions in cardiac myocytes and in cardiac pathology. Understanding the relative roles of these receptors at these locations could have a significant impact on pharmacological targeting of these receptors for the treatment of heart failure and cardiac diseases.


Subject(s)
Heart Failure , Receptors, Adrenergic, beta , Heart Failure/drug therapy , Heart Failure/metabolism , Humans , Myocytes, Cardiac , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
13.
Basic Res Cardiol ; 117(1): 37, 2022 07 17.
Article in English | MEDLINE | ID: mdl-35842861

ABSTRACT

We have recently identified a pool of intracellular ß1 adrenergic receptors (ß1ARs) at the sarcoplasmic reticulum (SR) crucial for cardiac function. Here, we aim to characterize the integrative control of intracellular catecholamine for subcellular ß1AR signaling and cardiac function. Using anchored Förster resonance energy transfer (FRET) biosensors and transgenic mice, we determined the regulation of compartmentalized ß1AR-PKA signaling at the SR and plasma membrane (PM) microdomains by organic cation transporter 3 (OCT3) and monoamine oxidase A (MAO-A), two critical modulators of catecholamine uptake and homeostasis. Additionally, we examined local PKA substrate phosphorylation and excitation-contraction coupling in cardiomyocyte. Cardiac-specific deletion of MAO-A (MAO-A-CKO) elevates catecholamines and cAMP levels in the myocardium, baseline cardiac function, and adrenergic responses. Both MAO-A deletion and inhibitor (MAOi) selectively enhance the local ß1AR-PKA activity at the SR but not PM, and augment phosphorylation of phospholamban, Ca2+ cycling, and myocyte contractile response. Overexpression of MAO-A suppresses the SR-ß1AR-PKA activity and PKA phosphorylation. However, deletion or inhibition of OCT3 by corticosterone prevents the effects induced by MAOi and MAO-A deletion in cardiomyocytes. Deletion or inhibition of OCT3 also negates the effects of MAOi and MAO-A deficiency in cardiac function and adrenergic responses in vivo. Our data show that MAO-A and OCT3 act in concert to fine-tune the intracellular SR-ß1AR-PKA signaling and cardiac fight-or-flight response. We reveal a drug contraindication between anti-inflammatory corticosterone and anti-depressant MAOi in modulating adrenergic regulation in the heart, providing novel perspectives of these drugs with cardiac implications.


Subject(s)
Corticosterone , Cyclic AMP-Dependent Protein Kinases , Adrenergic Agents/metabolism , Adrenergic Agents/pharmacology , Animals , Calcium/metabolism , Catecholamines/metabolism , Catecholamines/pharmacology , Cations/metabolism , Cations/pharmacology , Corticosterone/metabolism , Corticosterone/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/pharmacology , Mice , Monoamine Oxidase/metabolism , Monoamine Oxidase/pharmacology , Myocardial Contraction , Myocytes, Cardiac/metabolism , Phosphorylation , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Sarcoplasmic Reticulum
14.
J Mol Biol ; 434(16): 167687, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35717996

ABSTRACT

Cell-free expression enables direct cotranslational insertion of G protein coupled receptors (GPCRs) and other membrane proteins into the defined membrane environments of nanodiscs. This technique avoids GPCR contacts with detergents and allows rapid identification of lipid effects on GPCR function as well as fast screening of receptor derivatives. Critical steps of conventional GPCR preparation from cellular membranes followed by detergent-based reconstitution into nanodisc membranes are thus eliminated. We report the efficient cotranslational insertion of full-length human ß1-adrenergic receptor and of a truncated derivative into preformed nanodisc membranes. Their biochemical characterization revealed significant differences in lipid requirements, dimer formation and ligand binding activity. The truncated receptor showed a higher affinity to most tested ligands, in particular in presence of choline-containing lipids. However, introducing the naturally occurring G389R polymorphism in the full-length receptor resulted into an increased affinity to the antagonists alprenolol and carvedilol. Receptor quality was generally improved by coexpression with the agonist isoproterenol and the percentage of the ligand binding active fraction was twofold increased. Specific coupling of full-length and truncated human receptors in nanodisc membranes to Mini-Gαs protein as well as to purified Gs heterotrimer could be demonstrated and homogeneity of purified GPCR/Gs protein complexes in nanodiscs was demonstrated by negative stain single particle analysis.


Subject(s)
Nanostructures , Receptors, Adrenergic, beta-1 , Cell-Free System , Humans , Ligands , Lipids/chemistry , Nanostructures/chemistry , Polymorphism, Genetic , Protein Binding , Protein Biosynthesis , Protein Multimerization , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/genetics
15.
Endocr Relat Cancer ; 29(5): 267-272, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35258481

ABSTRACT

The release of excessive amounts of catecholamine by pheochromocytoma-paragangliomas (PPGL) can lead to life-threatening catecholamine-induced cardiomyopathy (CIC). Single-nucleotide polymorphisms of the beta1 and alpha-2c adrenergic receptors alter myocyte receptor function and enhanced norepinephrine release. We tested the hypothesis that such genetic variations may impact the risk of developing CIC in the context of PPGL. Thirty-one patients with PPGL, including nine with a history of CIC, were analyzed for alpha-2-adrenergic receptors: ADRA2C, beta-1 and beta-2 adrenergic receptors: ADRB1 and ADRB2 genotyping. CIC was defined either by a history of heart failure or cardiogenic shock associated with dilated or Takotsubo cardiomyopathy. Subjects were genotyped for ADRA2C (rs61767072 for del322_325), ADRB1 (rs1801252 for Ser49Gly and rs1801253 for Arg389Gly) and ADRB2 (rs1042713 for Arg16Gly and rs1042714 for Gln27Glu). Single-locus analysis revealed that variant in ADRA2C (alpha 2CDel322-325) was more common among patients with CIC than among controls (allele frequency, 0.44 vs 0.05; P< 0.001). The lack of alpha 2CDel322-325 polymorphism has a negative predictive value of 95% for the onset of CIC. In a replication cohort including 26 patients with PPGL whom eight have developed a CIC, the association between Alpha 2CDel322-325 and CIC was confirmed (allele frequency, 0.33 vs 0.; P= 0.0001). In conclusion, Alpha 2CDel322-325 through the identification of patients at low risk of developing CIC can help physicians to better determine the most appropriate therapeutic approach, notably in patients at high risk of surgical complications.


Subject(s)
Adrenal Gland Neoplasms , Cardiomyopathies , Paraganglioma , Pheochromocytoma , Receptors, Adrenergic, alpha-2 , Receptors, Adrenergic, beta-1 , Receptors, Adrenergic, beta-2 , Adrenal Gland Neoplasms/genetics , Biomarkers , Catecholamines , Genotype , Humans , Pheochromocytoma/genetics , Polymorphism, Single Nucleotide , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics
16.
Am J Physiol Heart Circ Physiol ; 322(3): H486-H491, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35148234

ABSTRACT

ß1-Adrenergic receptors (ß1ARs) are the principal mediators of catecholamine action in cardiomyocytes. We previously showed that ß1ARs accumulate as both full-length and NH2-terminally truncated species in cells, that maturational processing of full-length ß1ARs to an NH2-terminally truncated form is attributable to O-glycan-regulated proteolytic cleavage of the ß1AR NH2-terminus at R31 ↓ L32 by ADAM17, and that NH2-terminally truncated ß1ARs remain signaling competent but they acquire a distinct signaling phenotype. NH2-terminally truncated ß1ARs differ from full-length ß1ARs in their signaling bias to cAMP/PKA versus ERK pathways and only the NH2-terminally truncated form of the ß1AR constitutively activates AKT and confers protection against doxorubicin-dependent apoptosis in cardiomyocytes. Since the R31 ↓ L32 sequence conforms to a trypsin consensus cleavage site, we used immunoblotting methods to test the hypothesis that ß1ARs are also cleaved at R31 ↓ L32 by trypsin (an enzyme typically used to isolate cardiomyocytes from the intact ventricle). We show that full-length ß1ARs are cleaved by trypsin and that trypsin cleaves the full-length ß1AR NH2-terminus specifically at R31 ↓ L32 in CHO-Pro5 cells. Trypsin also cleaves ß1ARs in cardiomyocytes, but at a second site that results in the formation of ∼40-kDa NH2-terminal and ∼30-kDa COOH-terminal fragments. The observation that cardiomyocyte ß1ARs are cleaved by trypsin (a mechanism that constitutes a heretofore-unrecognized mechanism that would influence ß1AR-signaling responses) suggests that studies that use standard trypsin-based procedures to isolate adult cardiomyocytes from the intact ventricle should be interpreted with caution.NEW & NOTEWORTHY Current concepts regarding the molecular basis for ß1AR responses derive from literature predicated on the assumption that ß1ARs signal exclusively as full-length receptor proteins. However, we recently showed that ß1ARs accumulate as both full-length and NH2-terminally truncated forms. This manuscript provides novel evidence that ß1-adrenergic receptors can be cleaved by trypsin and that cell surface ß1AR cleavage constitutes a heretofore unrecognized mechanism to alter catecholamine-dependent signaling responses.


Subject(s)
Myocytes, Cardiac , Receptors, Adrenergic, beta-1 , Catecholamines/metabolism , Myocytes, Cardiac/metabolism , Proteolysis , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction , Trypsin/metabolism
17.
PLoS Med ; 19(2): e1003897, 2022 02.
Article in English | MEDLINE | ID: mdl-35113855

ABSTRACT

BACKGROUND: Epidemiological studies have reported conflicting findings on the potential adverse effects of long-term antihypertensive medication use on cancer risk. Naturally occurring variation in genes encoding antihypertensive drug targets can be used as proxies for these targets to examine the effect of their long-term therapeutic inhibition on disease outcomes. METHODS AND FINDINGS: We performed a mendelian randomization analysis to examine the association between genetically proxied inhibition of 3 antihypertensive drug targets and risk of 4 common cancers (breast, colorectal, lung, and prostate). Single-nucleotide polymorphisms (SNPs) in ACE, ADRB1, and SLC12A3 associated (P < 5.0 × 10-8) with systolic blood pressure (SBP) in genome-wide association studies (GWAS) were used to proxy inhibition of angiotensin-converting enzyme (ACE), ß-1 adrenergic receptor (ADRB1), and sodium-chloride symporter (NCC), respectively. Summary genetic association estimates for these SNPs were obtained from GWAS consortia for the following cancers: breast (122,977 cases, 105,974 controls), colorectal (58,221 cases, 67,694 controls), lung (29,266 cases, 56,450 controls), and prostate (79,148 cases, 61,106 controls). Replication analyses were performed in the FinnGen consortium (1,573 colorectal cancer cases, 120,006 controls). Cancer GWAS and FinnGen consortia data were restricted to individuals of European ancestry. Inverse-variance weighted random-effects models were used to examine associations between genetically proxied inhibition of these drug targets and risk of cancer. Multivariable mendelian randomization and colocalization analyses were employed to examine robustness of findings to violations of mendelian randomization assumptions. Genetically proxied ACE inhibition equivalent to a 1-mm Hg reduction in SBP was associated with increased odds of colorectal cancer (odds ratio (OR) 1.13, 95% CI 1.06 to 1.22; P = 3.6 × 10-4). This finding was replicated in the FinnGen consortium (OR 1.40, 95% CI 1.02 to 1.92; P = 0.035). There was little evidence of association of genetically proxied ACE inhibition with risk of breast cancer (OR 0.98, 95% CI 0.94 to 1.02, P = 0.35), lung cancer (OR 1.01, 95% CI 0.92 to 1.10; P = 0.93), or prostate cancer (OR 1.06, 95% CI 0.99 to 1.13; P = 0.08). Genetically proxied inhibition of ADRB1 and NCC were not associated with risk of these cancers. The primary limitations of this analysis include the modest statistical power for analyses of drug targets in relation to some less common histological subtypes of cancers examined and the restriction of the majority of analyses to participants of European ancestry. CONCLUSIONS: In this study, we observed that genetically proxied long-term ACE inhibition was associated with an increased risk of colorectal cancer, warranting comprehensive evaluation of the safety profiles of ACE inhibitors in clinical trials with adequate follow-up. There was little evidence to support associations across other drug target-cancer risk analyses, consistent with findings from short-term randomized controlled trials for these medications.


Subject(s)
Antihypertensive Agents/adverse effects , Mendelian Randomization Analysis/methods , Neoplasms/genetics , Peptidyl-Dipeptidase A/genetics , Receptors, Adrenergic, beta-1/genetics , Blood Pressure/drug effects , Blood Pressure/genetics , Female , Genome-Wide Association Study/methods , Humans , Male , Neoplasms/chemically induced , Neoplasms/epidemiology , Polymorphism, Single Nucleotide/drug effects , Polymorphism, Single Nucleotide/genetics , Risk Factors , Solute Carrier Family 12, Member 3/genetics
18.
Pharmacogenomics J ; 22(1): 62-68, 2022 02.
Article in English | MEDLINE | ID: mdl-34642472

ABSTRACT

Single nucleotide polymorphisms (SNPs) have been associated with differential beta-blocker (BB) effects on heart rate, blood pressure, and left ventricular ejection fraction in various patient populations. This study aimed to determine if SNPs previously associated with BB response are also associated with differential survival in heart failure (HF) patients receiving BBs. HF patient data were derived from electronic health records and the Social Security Death Index. Associations and interactions between BB dose, SNP genotype, and the outcome of death were assessed using a Cox proportional-hazard model adjusting for covariates known to be associated with differential survival in HF patients. Two SNPs, ADRB1 Arg389Gly and ADRB2 Glu27Gln, displayed significant interactions (Pint = 0.043 and Pint = 0.017, respectively) with BB dose and their association with mortality. Our study suggests that ADRB2 27Glu and ADRB1 389Arg may confer a larger survival benefit with higher BB doses in patients with HF.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Heart Failure/drug therapy , Heart Failure/mortality , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Adrenergic beta-Antagonists/administration & dosage , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Female , Genotype , Humans , Male , Middle Aged , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide , Proportional Hazards Models , Survival Analysis
19.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830474

ABSTRACT

ß-adrenergic receptor (ß-AR) stimulation represents a major mechanism of modulating cardiac output. In spite of its fundamental importance, its molecular basis on the level of cell signalling has not been characterised in detail yet. We employed mass spectrometry-based proteome and phosphoproteome analysis using SuperSILAC (spike-in stable isotope labelling by amino acids in cell culture) standardization to generate a comprehensive map of acute phosphoproteome changes in mice upon administration of isoprenaline (ISO), a synthetic ß-AR agonist that targets both ß1-AR and ß2-AR subtypes. Our data describe 8597 quantitated phosphopeptides corresponding to 10,164 known and novel phospho-events from 2975 proteins. In total, 197 of these phospho-events showed significantly altered phosphorylation, indicating an intricate signalling network activated in response to ß-AR stimulation. In addition, we unexpectedly detected significant cardiac expression and ISO-induced fragmentation of junctophilin-1, a junctophilin isoform hitherto only thought to be expressed in skeletal muscle. Data are available via ProteomeXchange with identifier PXD025569.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Phosphoproteins/genetics , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta/genetics , Amino Acids , Animals , Heart/drug effects , Humans , Isoproterenol/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Myocardium/metabolism , Myocardium/pathology , Phosphoproteins/antagonists & inhibitors , Phosphorylation/drug effects , Proteome/drug effects , Proteome/genetics , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2 , Signal Transduction/drug effects
20.
Mol Biol Rep ; 48(12): 7893-7900, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34716504

ABSTRACT

BACKGROUND: Takotsubo Cardiomyopathy (TC) is a rare disorder that is mostly caused by stress and is often misdiagnosed. We aimed to analyze Takotsubo Syndrome at the molecular level by using the Oxford Nanopore Minion Device and its protocol. METHODS AND RESULTS: Ten patients who were previously diagnosed with Takotsubo Syndrome (increased after decrease in ejection fraction and without critical stenosis in coronary arteries) and 10 healthy individuals in the control group were included in our project. The mean age was 53 ± 12.2 for the patient group and 52.4 ± 9.9 for the control group, and the left ventricular ejection fraction was 50.3 ± 11.5 for the patient group and 64.2 ± 2.8 for the control group (p < 0.05). Peripheral blood of patients and healthy individuals was taken and their DNA was obtained. By making long reads throughout the genome, the most studied regions responsible for ß-adrenergic signaling pathways; The gene expression level of cardiac ß-1 ADRB1 (rs1801253-ENST00000369295.4), G > C, (Gly389Arg) and cardiac ß-2 ADRB2 (rs1800888-ENSG00000169252), C > T, (Thr165Ile) adrenoceptors was investigated. As a result; no structural variation was detected leading to Takotsubo Cardiomyopathy. The results obtained from the bioinformatics analysis were also checked from the VarSome Tools and similar results were found. CONCLUSIONS: Many publications in TC susceptibility have that may lead to adrenergic pathway dysregulation, most studied adrenergic receptor genes in the similar literatures too. We searched for genetic variants in b1AR and b2AR genes in our study and however we could not find any variants in this study, we think larger numbers of cohort studies are needed.


Subject(s)
Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Takotsubo Cardiomyopathy/genetics , Adult , Aged , Cohort Studies , DNA , Gene Expression , Humans , Male , Middle Aged , Pilot Projects , Polymorphism, Single Nucleotide , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Stress, Physiological/genetics , Stress, Physiological/physiology , Stroke Volume , Turkey , Ventricular Function, Left
SELECTION OF CITATIONS
SEARCH DETAIL
...