Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 953
Filtrar
1.
Circ Res ; 133(11): 944-958, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37869877

RESUMEN

BACKGROUND: ß1AR (beta-1 adrenergic receptor) and ß2AR (beta-2 adrenergic receptor)-mediated cyclic adenosine monophosphate signaling has distinct effects on cardiac function and heart failure progression. However, the mechanism regulating spatial localization and functional compartmentation of cardiac ß-ARs remains elusive. Emerging evidence suggests that microtubule-dependent trafficking of mRNP (messenger ribonucleoprotein) and localized protein translation modulates protein compartmentation in cardiomyocytes. We hypothesized that ß-AR compartmentation in cardiomyocytes is accomplished by selective trafficking of its mRNAs and localized translation. METHODS: The localization pattern of ß-AR mRNA was investigated using single molecule fluorescence in situ hybridization and subcellular nanobiopsy in rat cardiomyocytes. The role of microtubule on ß-AR mRNA localization was studied using vinblastine, and its effect on receptor localization and function was evaluated with immunofluorescent and high-throughput Förster resonance energy transfer microscopy. An mRNA protein co-detection assay identified plausible ß-AR translation sites in cardiomyocytes. The mechanism by which ß-AR mRNA is redistributed post-heart failure was elucidated by single molecule fluorescence in situ hybridization, nanobiopsy, and high-throughput Förster resonance energy transfer microscopy on 16 weeks post-myocardial infarction and detubulated cardiomyocytes. RESULTS: ß1AR and ß2AR mRNAs show differential localization in cardiomyocytes, with ß1AR found in the perinuclear region and ß2AR showing diffuse distribution throughout the cell. Disruption of microtubules induces a shift of ß2AR transcripts toward the perinuclear region. The close proximity between ß2AR transcripts and translated proteins suggests that the translation process occurs in specialized, precisely defined cellular compartments. Redistribution of ß2AR transcripts is microtubule-dependent, as microtubule depolymerization markedly reduces the number of functional receptors on the membrane. In failing hearts, both ß1AR and ß2AR mRNAs are redistributed toward the cell periphery, similar to what is seen in cardiomyocytes undergoing drug-induced detubulation. This suggests that t-tubule remodeling contributes to ß-AR mRNA redistribution and impaired ß2AR function in failing hearts. CONCLUSIONS: Asymmetrical microtubule-dependent trafficking dictates differential ß1AR and ß2AR localization in healthy cardiomyocyte microtubules, underlying the distinctive compartmentation of the 2 ß-ARs on the plasma membrane. The localization pattern is altered post-myocardial infarction, resulting from transverse tubule remodeling, leading to distorted ß2AR-mediated cyclic adenosine monophosphate signaling.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Ratas , Animales , Hibridación Fluorescente in Situ , Insuficiencia Cardíaca/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , AMP Cíclico/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Microtúbulos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología
2.
Nature ; 622(7982): 383-392, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37731001

RESUMEN

CD8+ T cells are essential components of the immune response against viral infections and tumours, and are capable of eliminating infected and cancerous cells. However, when the antigen cannot be cleared, T cells enter a state known as exhaustion1. Although it is clear that chronic antigen contributes to CD8+ T cell exhaustion, less is known about how stress responses in tissues regulate T cell function. Here we show a new link between the stress-associated catecholamines and the progression of T cell exhaustion through the ß1-adrenergic receptor ADRB1. We identify that exhausted CD8+ T cells increase ADRB1 expression and that exposure of ADRB1+ T cells to catecholamines suppresses their cytokine production and proliferation. Exhausted CD8+ T cells cluster around sympathetic nerves in an ADRB1-dependent manner. Ablation of ß1-adrenergic signalling limits the progression of T cells towards the exhausted state in chronic infection and improves effector functions when combined with immune checkpoint blockade (ICB) in melanoma. In a pancreatic cancer model resistant to ICB, ß-blockers and ICB synergize to boost CD8+ T cell responses and induce the development of tissue-resident memory-like T cells. Malignant disease is associated with increased catecholamine levels in patients2,3, and our results establish a connection between the sympathetic stress response, tissue innervation and T cell exhaustion. Here, we uncover a new mechanism by which blocking ß-adrenergic signalling in CD8+ T cells rejuvenates anti-tumour functions.


Asunto(s)
Linfocitos T CD8-positivos , Catecolaminas , Receptores Adrenérgicos beta 1 , Sistema Nervioso Simpático , Agotamiento de Células T , Humanos , Antígenos/inmunología , Antígenos/metabolismo , Catecolaminas/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/terapia , Células T de Memoria/citología , Células T de Memoria/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Receptores Adrenérgicos beta 1/metabolismo , Sistema Nervioso Simpático/inmunología , Sistema Nervioso Simpático/fisiología , Estrés Fisiológico
3.
Life Sci ; 322: 121644, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37004731

RESUMEN

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.


Asunto(s)
Agonistas Adrenérgicos beta , Dobutamina , Humanos , Ratones , Ratas , Animales , Dobutamina/farmacología , Dobutamina/metabolismo , Agonistas Adrenérgicos beta/farmacología , Atrios Cardíacos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Isoproterenol/farmacología , Isoproterenol/metabolismo , Albuterol/farmacología , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/metabolismo
4.
J Neuroinflammation ; 20(1): 56, 2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36864439

RESUMEN

BACKGROUND: Noradrenergic neurons in the locus coeruleus (LC) are the primary source of norepinephrine (NE) in the brain and degeneration of these neurons is reported in the early stages of Parkinson's disease (PD), even prior to dopaminergic neuron degeneration in the substantia nigra (SN), which is a hallmark of PD pathology. NE depletion is generally associated with increased PD pathology in neurotoxin-based PD models. The effect of NE depletion in other models of PD-like α-synuclein-based models is largely unexplored. In PD models and in human patients, ß-adrenergic receptors' (AR) signaling is associated with a reduction of neuroinflammation and PD pathology. However, the effect of NE depletion in the brain and the extent of NE and ß-ARs signaling involvement in neuroinflammation, and dopaminergic neuron survival is poorly understood. METHODS: Two mouse models of PD, a 6OHDA neurotoxin-based model and a human α-synuclein (hα-SYN) virus-based model of PD, were used. DSP-4 was used to deplete NE levels in the brain and its effect was confirmed by HPLC with electrochemical detection. A pharmacological approach was used to mechanistically understand the impact of DSP-4 in the hα-SYN model of PD using a norepinephrine transporter (NET) and a ß-AR blocker. Epifluorescence and confocal imaging were used to study changes in microglia activation and T-cell infiltration after ß1-AR and ß2-AR agonist treatment in the hα-SYN virus-based model of PD. RESULTS: Consistent with previous studies, we found that DSP-4 pretreatment increased dopaminergic neuron loss after 6OHDA injection. In contrast, DSP-4 pretreatment protected dopaminergic neurons after hα-SYN overexpression. DSP-4-mediated protection of dopaminergic neurons after hα-SYN overexpression was dependent on ß-AR signaling since using a ß-AR blocker prevented DSP-4-mediated dopaminergic neuron protection in this model of PD. Finally, we found that the ß-2AR agonist, clenbuterol, reduced microglia activation, T-cell infiltration, and dopaminergic neuron degeneration, whereas xamoterol a ß-1AR agonist showed increased neuroinflammation, blood brain barrier permeability (BBB), and dopaminergic neuron degeneration in the context of hα-SYN-mediated neurotoxicity. CONCLUSIONS: Our data demonstrate that the effects of DSP-4 on dopaminergic neuron degeneration are model specific, and suggest that in the context of α-SYN-driven neuropathology, ß2-AR specific agonists may have therapeutic benefit in PD.


Asunto(s)
Síndromes de Neurotoxicidad , Enfermedad de Parkinson , Animales , Humanos , Ratones , alfa-Sinucleína , Neuronas Dopaminérgicas , Degeneración Nerviosa , Enfermedades Neuroinflamatorias , Neurotoxinas , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo
5.
Eur J Pharmacol ; 948: 175700, 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37001579

RESUMEN

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.


Asunto(s)
Monocitos , Propranolol , Humanos , Monocitos/metabolismo , Propranolol/farmacología , Benzodiazepinas , Diazepam/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo
6.
Cell Mol Immunol ; 20(2): 175-188, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36600052

RESUMEN

Upon viral infection, cytoplasmic pattern recognition receptors detect viral nucleic acids and activate the adaptor protein VISA/MAVS- or MITA/STING-mediated innate antiviral response. Whether and how the innate antiviral response is regulated by neuronal endocrine functions is unclear. Here, we show that viral infection reduced the serum levels of the ß-adrenergic hormones epinephrine and norepinephrine as well as the cellular levels of their receptors ADRB1 and ADRB2. We further show that an increase in epinephrine/norepinephrine level inhibited the innate antiviral response in an ADRB1-/2-dependent manner. Mechanistically, epinephrine/norepinephrine stimulation activated the downstream kinase PKA, which catalyzed the phosphorylation of MITA at S241, S243 and T263, inhibiting MITA activation and suppressing the innate immune response to DNA virus. In addition, phosphorylation of VISA at T54 by PKA antagonized the innate immune response to RNA virus. These findings reveal the regulatory mechanisms of innate antiviral responses by epinephrine/norepinephrine and provide a possible explanation for increased host susceptibility to viral infection in stressful and anxiety-promoting situations.


Asunto(s)
Proteínas de la Membrana , Virosis , Humanos , Antivirales , Epinefrina/farmacología , Inmunidad Innata/genética , Proteínas de la Membrana/genética , Norepinefrina/farmacología , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Activación Enzimática , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo
7.
J Pathol ; 259(1): 69-80, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36245401

RESUMEN

While multi-drug combinations and continuous treatment have become standard for multiple myeloma, the disease remains incurable. Repurposing drugs that are currently used for other indications could provide a novel approach to improve the therapeutic efficacy of standard multiple myeloma treatments. Here, we assessed the anti-tumor effects of cardiac drugs called ß-blockers as a single agent and in combination with commonly used anti-myeloma therapies. Expression of the ß2 -adrenergic receptor correlated with poor survival outcomes in patients with multiple myeloma. Targeting the ß2 -adrenergic receptor (ß2 AR) using either selective or non-selective ß-blockers reduced multiple myeloma cell viability, and induced apoptosis and autophagy. Blockade of the ß2 AR modulated cancer cell metabolism by reducing the mitochondrial respiration as well as the glycolytic activity. These effects were not observed by blockade of ß1 -adrenergic receptors. Combining ß2 AR blockade with the chemotherapy drug melphalan or the proteasome inhibitor bortezomib significantly increased apoptosis in multiple myeloma cells. These data identify the therapeutic potential of ß2 AR-blockers as a complementary or additive approach in multiple myeloma treatment and support the future clinical evaluation of non-selective ß-blockers in a randomized controlled trial. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Mieloma Múltiple , Humanos , Mieloma Múltiple/tratamiento farmacológico , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 1/uso terapéutico , Transducción de Señal , Bortezomib/farmacología , Bortezomib/uso terapéutico , Apoptosis
8.
Biomed Pharmacother ; 157: 114006, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36395608

RESUMEN

Depression has been associated with altered signal transduction of serotonergic, dopaminergic and adrenergic neurotransmitter systems in the brain. Signaling relies on receptor-ligand interactions and subsequent regulatory processes, but also on lateral receptor mobility. The aim of this study was to investigate the effect of the St. John's wort extract Ze 117 on the lateral mobility of SNAP-tagged ß1-adrenergic receptors (ß1AR) in the plasma membrane of C6 cells under both, non-stimulating and isoprenaline-stimulating conditions. Single particle tracking (SPT) was used, whereby the registered trajectories were evaluated by variational Bayesian treatment of a hidden Markov model (vbSPT) and packing coefficient (Pc) analysis with respect to diffusion coefficients, receptor state occupancies and confinement. Three different diffusion states were identified, differing in their diffusion coefficients. Treatment with Ze 117 [25 µg/ml] decreased the mobility of the ß1AR, which was manifested by a relative increase in the slow-diffusing state S1 (0.21-0.30) compared to control and by an increase in receptor confinement (79.4-68.1 nm). After isoprenaline stimulation of control cells, the slow-diffusing state was more pronounced, whereas confinement was not affected. In summary, SPT has been shown to be a powerful method to analyze lateral receptor mobility. Furthermore, the present study identified a correlation between Ze 117 treatment and ß1AR mobility.


Asunto(s)
Hypericum , Receptores Adrenérgicos beta 1/metabolismo , Teorema de Bayes , Extractos Vegetales/farmacología , Membrana Celular , Fitoterapia
9.
Iran J Med Sci ; 47(4): 367-378, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35919076

RESUMEN

Background: Cirrhotic cardiomyopathy is a well-recognized cardiac dysfunction in cirrhotic patients. Studies have confirmed the protective effects of silymarin in different types of cardiac injury. This study aimed to examine the effectiveness and molecular mechanism of silymarin against myocardial dysfunction and hypertrophy in a rat model of cirrhosis. Methods: The experiment was performed at Alborz University of Medical Sciences (Karaj, Iran) during 2020-2021. Thirty-two male Wistar rats were randomly divided into four groups of Sham-operated (control group for surgical procedures), Bile Duct Ligated (BDL), and two Silymarin extract (SE)-treated groups of 300 and 600 mg/Kg/day. After 28 days, serum levels of AST, ALT, GGT, and ALP, liver histopathological status, as well as cardiac mechanical function, were assessed. Cardiac ß1-adrenergic receptors (ß1-AR), L-type voltage-dependent calcium channels (L-VDCC), and GATA4 mRNA expression were also determined using real-time RT-PCR. Data analysis was performed using the one-way ANOVA followed by Duncan's multiple range test. Histological data has been analyzed with Kruskal-Wallis nonparametric test. The analysis was performed at P≤0.05. Results: BDL was associated with a significant elevation in serum AST, ALT, GGT, and ALP, development of necrosis and fibrosis of the liver texture, increased Heart Weight and Heart Weight to Body Weight ratio, enhanced cardiac mechanical function as well as a significant up-regulation of ventricular ß1-AR and L-VDCC. Administration of SE600, but not SE300, significantly reduced the serum levels of the enzymes and alleviated signs of liver necrosis and fibrosis. Cirrhotic-induced cardiac dysfunction was also restored by SE600, but not by the lower dose. In addition, cardiac expression of the ß1-AR and L-VDCC was down-regulated toward normal values by either higher or lower doses of the SE. Conclusion: Silymarin treatment in higher dose attenuated cirrhosis-associated cardiac remodeling and reduced cardiac mechanical dysfunctions.


Asunto(s)
Cardiomiopatías , Silimarina , Animales , Canales de Calcio Tipo L , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/metabolismo , Cirrosis Hepática/complicaciones , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Masculino , Necrosis/tratamiento farmacológico , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 1/metabolismo , Silimarina/farmacología , Silimarina/uso terapéutico
10.
J Chromatogr A ; 1677: 463298, 2022 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-35809523

RESUMEN

The discovery of beta1-adrenoceptor (ß1-AR) ligands is viewed as an enormous demand for fighting ailments mediated by the receptor including cardiovascular diseases. Such pursuit is gravely challenged due to the lack of lead screening methods with high efficiency. This work developed a chromatographic method for pursuing ß1-AR ligand from the herbal extract by fusing epidermal growth factor receptor (EGFR) as a tag at its C-terminus to stably express the fusion receptor in E. coli, immobilizing the expressed EGFR-tagged ß1-AR onto ibrutinib-derivatized amino microspheres, and applying the immobilized receptor in the analysis of ligand-receptor interaction and herbal extract. Comprehensive characterizations like X-ray photoelectron spectroscopy and retention behaviors of canonical drugs demonstrated high specificity and good stability of the immobilized ß1-AR prepared through the covalent reaction between the EGFR and ibrutinib decorated on the microsphere surface. Frontal analysis of atenolol, metoprolol, and esmolol confirmed their bindings to ß1-AR with association constants of 1.07 × 104, 6.54 × 103, and 1.45 × 104 M-1. The thermodynamic analysis provided proof of electrostatic interaction, hydrogen bonds, and van der Waals force driving those interactions. Pulegone was recognized as a bioactive compound that specifically binding to ß1-AR from the extract of Ziziphora clinopodioides Lam by analyzing the retention peak through reverse-phase high performance liquid chromatography coupled with tandem mass spectrometry. These results, taken together, indicated that the current method is possible to provide an alternative for discovering ß1-AR ligands with high efficiency from complex matrices like herbal extract.


Asunto(s)
Medicamentos Herbarios Chinos , Proteínas de Escherichia coli , Receptores Adrenérgicos beta 1/metabolismo , Ligasas de Carbono-Oxígeno , Cromatografía , Medicamentos Herbarios Chinos/química , Receptores ErbB , Escherichia coli/metabolismo , Ligandos , Receptores Adrenérgicos beta 2/química
11.
J Cardiovasc Pharmacol ; 80(3): 334-341, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35881897

RESUMEN

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.


Asunto(s)
Insuficiencia Cardíaca , Receptores Adrenérgicos beta , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Humanos , Miocitos Cardíacos , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal
12.
Basic Res Cardiol ; 117(1): 37, 2022 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-35842861

RESUMEN

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.


Asunto(s)
Corticosterona , Proteínas Quinasas Dependientes de AMP Cíclico , Adrenérgicos/metabolismo , Adrenérgicos/farmacología , Animales , Calcio/metabolismo , Catecolaminas/metabolismo , Catecolaminas/farmacología , Cationes/metabolismo , Cationes/farmacología , Corticosterona/metabolismo , Corticosterona/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Ratones , Monoaminooxidasa/metabolismo , Monoaminooxidasa/farmacología , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Fosforilación , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Retículo Sarcoplasmático
13.
Glia ; 70(9): 1777-1794, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35589612

RESUMEN

Norepinephrine exerts powerful influences on the metabolic, neuroprotective and immunoregulatory functions of astrocytes. Until recently, all effects of norepinephrine were believed to be mediated by receptors localized exclusively to the plasma membrane. However, recent studies in cardiomyocytes have identified adrenergic receptors localized to intracellular membranes, including Golgi and inner nuclear membranes, and have shown that norepinephrine can access these receptors via transporter-mediated uptake. We recently identified a high-capacity norepinephrine transporter, organic cation transporter 3 (OCT3), densely localized to outer nuclear membranes in astrocytes, suggesting that adrenergic signaling may also occur at the inner nuclear membrane in these cells. Here, we used immunofluorescence and western blot to show that ß1 -adrenergic receptors are localized to astrocyte inner nuclear membranes; that key adrenergic signaling partners are present in astrocyte nuclei; and that OCT3 and other catecholamine transporters are localized to astrocyte plasma and nuclear membranes. To test the functionality of nuclear membrane ß1 -adrenergic receptors, we monitored real-time protein kinase A (PKA) activity in astrocyte nuclei using a fluorescent biosensor. Treatment of astrocytes with norepinephrine induced rapid increases in PKA activity in the nuclear compartment. Pretreatment of astrocytes with inhibitors of catecholamine uptake blocked rapid norepinephrine-induced increases in nuclear PKA activity. These studies, the first to document functional adrenergic receptors at the nuclear membrane in any central nervous system cell, reveal a novel mechanism by which norepinephrine may directly influence nuclear processes. This mechanism may contribute to previously described neuroprotective, metabolic and immunoregulatory actions of norepinephrine.


Asunto(s)
Astrocitos , Norepinefrina , Adrenérgicos/farmacología , Astrocitos/metabolismo , Catecolaminas/metabolismo , Catecolaminas/farmacología , Norepinefrina/metabolismo , Norepinefrina/farmacología , Membrana Nuclear/metabolismo , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 1/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 322(3): H486-H491, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35148234

RESUMEN

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.


Asunto(s)
Miocitos Cardíacos , Receptores Adrenérgicos beta 1 , Catecolaminas/metabolismo , Miocitos Cardíacos/metabolismo , Proteolisis , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Transducción de Señal , Tripsina/metabolismo
15.
Cells ; 11(2)2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-35053304

RESUMEN

Intracellular Ca2+ overload secondary to chronic hemodynamic stimuli promotes the recruitment of Ca2+-dependent signaling implicated in cardiomyocyte hypertrophy. The present study tested the hypothesis that sympathetic-mediated hypertrophy of neonatal rat ventricular cardiomyocytes (NRVMs) translated to an increase in calcium influx secondary to the upregulation of CaV1.2 channel subunits. Confocal imaging of norepinephrine (NE)-treated NRVMs revealed a hypertrophic response compared to untreated NRVMs. L-type CaV1.2 peak current density was increased 4-fold following a 24-h stimulation with NE. NE-treated NRVMs exhibited a significant upregulation of CaVα2δ1 and CaVß3 protein levels without significant changes of CaVα1C and CaVß2 protein levels. Pre-treatment with the ß1-blocker metoprolol failed to inhibit hypertrophy or CaVß3 upregulation whereas CaVα2δ1 protein levels were significantly reduced. NE promoted the phosphorylation of ERK 1/2, and the response was attenuated by the ß1-blocker. U0126 pre-treatment suppressed NE-induced ERK1/2 phosphorylation but failed to attenuate hypertrophy. U0126 inhibition of ERK1/2 phosphorylation prevented NE-mediated upregulation of CaVα2δ1, whereas CaVß3 protein levels remained elevated. Thus, ß1-adrenergic receptor-mediated recruitment of the ERK1/2 plays a seminal role in the upregulation of CaVα2δ1 in NRVMs independent of the concomitant hypertrophic response. However, the upregulation of CaVß3 protein levels may be directly dependent on the hypertrophic response of NRVMs.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Ventrículos Cardíacos/citología , Sistema de Señalización de MAP Quinasas , Miocitos Cardíacos/metabolismo , Subunidades de Proteína/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Sistema Nervioso Simpático/metabolismo , Regulación hacia Arriba , Animales , Animales Recién Nacidos , Calcio/metabolismo , Hipertrofia , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Norepinefrina/farmacología , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Regulación hacia Arriba/efectos de los fármacos
16.
J Cardiovasc Pharmacol ; 80(3): 328-333, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35099166

RESUMEN

ABSTRACT: ß 1 -adrenergic receptors (ß 1 ARs) are the principle mediators of catecholamine actions in cardiomyocytes. ß 1 ARs rapidly adjust cardiac output and provide short-term hemodynamic support for the failing heart by activating a Gs-adenylyl cyclase pathway that increases 3'-5'-cyclic adenosine monophosphate and leads to the activation of protein kinase A and the phosphorylation of substrates involved in excitation-contraction coupling. However, chronic persistent ß 1 AR activation in the setting of heart failure leads to a spectrum of maladaptive changes that contribute to the evolution of heart failure. The molecular basis for ß 1 AR-driven maladaptive responses remains uncertain because chronic persistent ß 1 AR activation has been linked to the activation of both proapoptotic and antiapoptotic signaling pathways. Of note, studies to date have been predicated on the assumption that ß 1 ARs signal exclusively as full-length receptor proteins. Our recent studies show that ß 1 ARs are detected as both full-length and N-terminally truncated species in cardiomyocytes, that N-terminal cleavage is regulated by O-glycan modifications at specific sites on the ß 1 AR N-terminus, and that N-terminally truncated ß 1 ARs remain signaling competent, but their signaling properties differ from those of the full-length ß 1 AR. The N-terminally truncated form of the ß 1 AR constitutively activates the protein kinase B signaling pathway and confers protection against doxorubicin-dependent apoptosis in cardiomyocytes. These studies identify a novel signaling paradigm for the ß 1 AR, implicating the N-terminus as a heretofore-unrecognized structural determinant of ß 1 AR responsiveness that could be pharmacologically targeted for therapeutic advantage.


Asunto(s)
Insuficiencia Cardíaca , Miocitos Cardíacos , Adenilil Ciclasas/metabolismo , AMP Cíclico/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/fisiología
17.
Sci Rep ; 12(1): 41, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996972

RESUMEN

Heart failure has high morbidity and mortality in the developed countries. Autophagy is important for the quality control of proteins and organelles in the heart. Rubicon (Run domain Beclin-1-interacting and cysteine-rich domain-containing protein) has been identified as a potent negative regulator of autophagy and endolysosomal trafficking. The aim of this study was to investigate the in vivo role of Rubicon-mediated autophagy and endosomal trafficking in the heart. We generated cardiomyocyte-specific Rubicon-deficient mice and subjected the mice to pressure overload by means of transverse aortic constriction. Rubicon-deficient mice showed heart failure with left ventricular dilatation, systolic dysfunction and lung congestion one week after pressure overload. While autophagic activity was unchanged, the protein amount of beta-1 adrenergic receptor was decreased in the pressure-overloaded Rubicon-deficient hearts. The increases in heart rate and systolic function by beta-1 adrenergic stimulation were significantly attenuated in pressure-overloaded Rubicon-deficient hearts. In isolated rat neonatal cardiomyocytes, the downregulation of the receptor by beta-1 adrenergic agonist was accelerated by knockdown of Rubicon through the inhibition of recycling of the receptor. Taken together, Rubicon protects the heart from pressure overload. Rubicon maintains the intracellular recycling of beta-1 adrenergic receptor, which might contribute to its cardioprotective effect.


Asunto(s)
Proteínas Relacionadas con la Autofagia , Insuficiencia Cardíaca , Receptores Adrenérgicos beta 1 , Animales , Masculino , Ratones , Autofagia/fisiología , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Células Cultivadas , Endosomas/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocitos Cardíacos/metabolismo , Ratas Wistar , Receptores Adrenérgicos beta 1/metabolismo
18.
J Pharmacol Sci ; 148(1): 51-55, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34924129

RESUMEN

The present study aimed to characterize and compare ß-adrenoceptors in the rat bladder with those in the heart and lungs of SD rats (8-10 weeks old) using subtype-selective agonists and antagonists in a radioligand binding assay with (-)-[125I]cyanopindolol ([125I]CYP), and also to clarify alterations in ß-adrenoceptors in the bladder of spontaneously hypertensive rats (SHR) at 14 weeks old, from those of Wistar-Kyoto rats (WKY) and Wistar rats at the same age. A radioligand binding assay with [125I]CYP was used to measure ß-adrenoceptor binding activity in rat tissues. Metoprolol exhibited the highest affinity to specific binding sites of [125I]CYP in the rat heart, indicating the dominance of ß1-adrenoceptors. ß3-selective agonists (BRL37344 and CL316243) and antagonist (SR59230A) exhibited higher affinity to specific binding sites of [125I]CYP in the bladder than in the heart and lungs. Furthermore, the binding affinity of the ß2-selective antagonist, ICI118551 was the highest in the bladder. The Bmax of specific [125]CYP binding in the bladder was significantly lower in WKY and SHR than in Wistar rats. The present study provides further evidence for the coexistence of ß2-and ß3-adrenoceptors in the rat bladder, and indicates that ß-adrenoceptor density is lower in the bladders of WKY and SHR.


Asunto(s)
Pulmón/metabolismo , Miocardio/metabolismo , Ratas Endogámicas SHR/metabolismo , Receptores Adrenérgicos beta/metabolismo , Vejiga Urinaria/metabolismo , Animales , Ensayo de Unión Radioligante/métodos , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Ratas Wistar , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 3/metabolismo
19.
Nat Struct Mol Biol ; 28(11): 936-944, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34759376

RESUMEN

The ß1-adrenergic receptor (ß1-AR) can activate two families of G proteins. When coupled to Gs, ß1-AR increases cardiac output, and coupling to Gi leads to decreased responsiveness in myocardial infarction. By comparative structural analysis of turkey ß1-AR complexed with either Gi or Gs, we investigate how a single G-protein-coupled receptor simultaneously signals through two G proteins. We find that, although the critical receptor-interacting C-terminal α5-helices on Gαi and Gαs interact similarly with ß1-AR, the overall interacting modes between ß1-AR and G proteins vary substantially. Functional studies reveal the importance of the differing interactions and provide evidence that the activation efficacy of G proteins by ß1-AR is determined by the entire three-dimensional interaction surface, including intracellular loops 2 and 4 (ICL2 and ICL4).


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Estructura Terciaria de Proteína/fisiología , Receptores Adrenérgicos beta 1/metabolismo , Animales , Gasto Cardíaco/genética , Gasto Cardíaco/fisiología , Línea Celular , Microscopía por Crioelectrón , AMP Cíclico/metabolismo , Activación Enzimática/fisiología , Células HEK293 , Cardiopatías/patología , Humanos , Hipertensión/patología , Isoproterenol/química , Estructura Secundaria de Proteína/fisiología , Células Sf9 , Transducción de Señal/fisiología
20.
Mol Biol Rep ; 48(12): 7893-7900, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34716504

RESUMEN

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.


Asunto(s)
Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Cardiomiopatía de Takotsubo/genética , Adulto , Anciano , Estudios de Cohortes , ADN , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Polimorfismo de Nucleótido Simple , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Volumen Sistólico , Turquía , Función Ventricular Izquierda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA