Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Naunyn Schmiedebergs Arch Pharmacol ; 397(9): 6551-6562, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38457039

RESUMEN

Sepsis/septic shock activates the sympathetic nervous system (SNS) to deal with the infection stress. However, an imbalanced or maladaptive response due to excessive or uncontrolled activation characterizes autonomic dysfunction. Our hypothesis was that reducing this excessive activation of the autonomic nervous system would impact positively in sepsis. Using ganglionic blockers as a pharmacological approach, the main aim of the present report was to assess the role of ganglionic transmission in the vascular dysfunction associated with sepsis.Sepsis was induced in rats by cecal ligation and puncture (CLP). One hour after CLP surgery, rats were treated subcutaneously with hexamethonium (15 mg/kg; ganglionic blocker), pentolinium (5 mg/kg; a blocker with a higher selectivity for sympathetic ganglia compared to hexamethonium), or vehicle (PBS). Basal blood pressure and the response to adrenergic agonists were evaluated at 6 and 24 h after CLP surgery. Reactivity to vasoconstrictors, nitric oxide (NO) synthase 2 (NOS-2) expression, IL-1 and TNF plasma levels, and density of α1 adrenergic receptors were evaluated in the aorta 24 h after CLP.Septic shock resulted in hypotension and hyporesponsiveness to norepinephrine and phenylephrine, increased plasma cytokine levels and NOS-2 expression in the aorta, and decreased α1 receptor density in the same vessel. Pentolinium but not hexamethonium recovered responsiveness and α1 adrenergic receptor density in the aorta. Both blockers normalized the in vivo response to vasoconstrictors, and reduced plasma IL-1 and NOx levels and NOS-2 expression in the aorta.Blockade of ganglionic sympathetic transmission reduced the vascular dysfunction in experimental sepsis. This beneficial effect seems to be, at least in part, due to the preservation of α1 adrenergic receptor density and to reduced NOS-2 expression and may lead to adjuvant ways to treat human sepsis.


Asunto(s)
Ganglios Simpáticos , Choque Séptico , Animales , Choque Séptico/fisiopatología , Choque Séptico/tratamiento farmacológico , Choque Séptico/metabolismo , Masculino , Ganglios Simpáticos/efectos de los fármacos , Ganglios Simpáticos/fisiopatología , Ganglios Simpáticos/metabolismo , Bloqueadores Ganglionares/farmacología , Ratas Wistar , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Vasoconstrictores/farmacología
2.
Eur J Pharmacol ; 945: 175603, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-36804548

RESUMEN

The motor activity of the epididymis duct is an essential process for male fertility and it is regulated by hormonal, neuronal and epithelial mechanisms. However, although there is evidence for the presence of histamine in the epididymis, its effects on epididymal motor activity are unknown. This study sought to evaluate the contractile effects of histamine on the rat distal cauda epididymis duct. Segments of the distal cauda epididymis duct from male Wistar rats were isolated and used in isolated organ bath experiments to evaluate the contractile effects of histamine in the absence or presence of antagonists of histamine receptors, α1-adrenoceptors and muscarinic acetylcholine receptors. The effects of histamine on noradrenaline induced contractions were also investigated. Histamine was able to induce phasic contractions on rat distal cauda epididymis duct which were prevented by promethazine 10-1000 nM (H1 receptor antagonist), ranitidine 1-100 µM (H2 receptor antagonist), atropine 100 nM (muscarinic antagonist), and prazosin 100 nM (α1-adrenoceptor antagonist). In addition, histamine was also able to modify noradrenaline-induced contractions possibly via activation of H1 and H2 receptors. In conclusion, this study demonstrates that histamine can induce phasic contractions of rat distal cauda epididymis via H2 receptors and autonomic neurotransmitters. Histamine may also exert modulatory actions on contractions of rat distal cauda epididymis duct induced by adrenergic receptor agonists. Further studies are necessary to unveil the localization of histamine receptors within the epididymal duct and the consequences of manipulation of the histaminergic system on epididymal function and male fertility.


Asunto(s)
Epidídimo , Histamina , Ratas , Masculino , Animales , Ratas Wistar , Histamina/farmacología , Prazosina/farmacología , Norepinefrina/farmacología , Receptores Adrenérgicos alfa 1
3.
Mini Rev Med Chem ; 22(18): 2383-2405, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35507746

RESUMEN

Adrenoceptors are the receptors for catecholamines, adrenaline, and noradrenaline. They are divided in α (α1 and α2) and ß (ß1, ß2 and ß3). α1-adrenoceptors are subdivided in α1A, α1B and α1D. Most tissues express mixtures of α1-adrenoceptors subtypes, which appear to coexist in different densities and ratios, and in most cases, their responses are probably due to the activation of more than one type. The three subtypes of α1-adrenoceptors are G-protein-coupled receptors (GPCR), specifically coupled to Gq/11. Additionally, the activation of these receptors may activate other signaling pathways or different components of these pathways, which leads to a great variety of possible cellular effects. The first clinically used α1 antagonist was Prazosin for Systemic Arterial Hypertension (SAH). It was followed by its congeners, Terazosin and Doxazosin. Nowadays, there are many classes of α-adrenergic antagonists with different selectivity profiles. In addition to SAH, the α1-adrenoceptors are used to treat Benign Prostatic Hyperplasia (BPH) and urolithiasis. This antagonism may be part of the mechanism of action of tricyclic antidepressants. Moreover, the activation of these receptors may lead to adverse effects such as orthostatic hypotension, similar to what happens with antidepressants and with some antipsychotics. Structure-activity relationships can explain, in part, how antagonists work and how selective they can be for each one of the subtypes. However, it is necessary to develop new molecules which antagonize the α1- adrenoceptors or make chemical modifications in these molecules to improve the selectivity and pharmacokinetic profile and/or reduce the adverse effects of known drugs.


Asunto(s)
Antipsicóticos , Doxazosina , Antagonistas Adrenérgicos alfa/farmacología , Antidepresivos Tricíclicos , Epinefrina , Norepinefrina , Prazosina/metabolismo , Receptores Adrenérgicos alfa 1/análisis , Receptores Adrenérgicos alfa 1/metabolismo
4.
Ann Hepatol ; 27 Suppl 1: 100570, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34706275

RESUMEN

INTRODUCTION AND OBJECTIVES: This study aimed to investigate miR-3682 as a biomarker in hepatocellular carcinoma (HCC). MATERIALS AND METHODS: MiRNA and RNA profiles of 375 HCC tissues and 50 normal liver samples were downloaded from The Cancer Genome Atlas (TCGA) database. Multivariate Cox regression and Kaplan-Meier analyses were applied to examine the prognostic value of factors. Target genes of miR-3682 were analyzed by TargetScan and dual-luciferase reporter assay. Online Database for Annotation, Visualization, and Integrated Discovery (DAVID) to perform KEGG pathway enrichment. Cell counting kit-8, colony formation and migration and invasion assays were performed to analyze biological behaviors of HCC cells. RESULTS: MiR-3682 was identified to be highly expressed in HCC tissues and cell lines. And miR-3682 was negatively and independently associated with the outcome of HCC patients. Inhibition of miR-3682 suppressed HCC cell viability and mobility. ADRA1A, predicted and confirmed as the novel target of miR-3682, was an independent and positive prognostic predictor for HCC. In addition, the knockdown of ADRA1A partially offset the inhibitory effect of miR-3682 inhibitor on the growth and mobility of HCC cells. DAVID enrichment and western blot of key signaling-related proteins analyses revealed that miR-3682 inactivated 5'-AMP-activated protein kinase (AMPK) signaling by negatively regulating ADRA1A. Mechanically, it was partially through suppressing AMPK signaling via targeting ADRA1A that miR-3682 supported the HCC cell malignant phenotype. CONCLUSIONS: This study implicates that miR-3682 plays an oncogenetic role in HCC and can be considered a novel therapeutic target and prognostic indicator of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/patología , MicroARNs/genética , MicroARNs/metabolismo , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo
5.
Clin Sci (Lond) ; 135(20): 2341-2356, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34622918

RESUMEN

Renal vascular reactivity to vasoconstrictors is preserved in sepsis in opposition to what happens in the systemic circulation. We studied whether this distinct behavior was related to α1 adrenergic receptor density, G protein-coupled receptor kinase 2 (GRK2) and the putative role of nitric oxide (NO). Sepsis was induced in female mice by cecal ligation and puncture (CLP). Wildtype mice were treated with prazosin 12 h after CLP or nitric oxide synthase 2 (NOS-2) inhibitor, 30 min before and 6 and 12 h after CLP. In vivo experiments and biochemistry assays were performed 24 h after CLP. Sepsis decreased the systemic mean arterial pressure (MAP) and the vascular reactivity to phenylephrine. Sepsis also reduced basal renal blood flow which was normalized by treatment with prazosin. Sepsis led to a substantial decrease in GRK2 level associated with an increase in α1 adrenergic receptor density in the kidney. The disappearance of renal GRK2 was prevented in NOS-2-KO mice or mice treated with 1400 W. Treatment of non-septic mice with an NO donor reduced GRK2 content in the kidney. Therefore, our results show that an NO-dependent reduction in GRK2 level in the kidney leads to the maintenance of a normal α1 adrenergic receptor density. The preservation of the density and/or functionality of this receptor in the kidney together with a higher vasoconstrictor tonus in sepsis lead to vasoconstriction. Thus, the increased concentration of vasoconstrictor mediators together with the preservation (and even increase) of the response to them may help to explain sepsis-induced acute kidney injury.


Asunto(s)
Lesión Renal Aguda/etiología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Riñón/metabolismo , Sepsis/complicaciones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Presión Arterial , Modelos Animales de Enfermedad , Femenino , Riñón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Circulación Renal , Sepsis/metabolismo , Sepsis/fisiopatología , Factores de Tiempo
6.
Clin Transl Oncol ; 23(8): 1705-1710, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33644847

RESUMEN

BACKGROUND: Neurotransmitters released from the sympathetic nervous system attach to the adrenergic receptors on the surface of tumoral cells in response to stress, and alter the expression of genes programming cellular activity. This study aimed to assess the expression of α1 adrenergic receptors in the serum and saliva of patients with oral squamous cell carcinoma (OSCC) compared with healthy controls. MATERIALS AND METHODS: In this case-control study, serum and stimulated and unstimulated saliva samples were collected from 26 OSCC patients and 26 healthy controls. ELISA kits were used for measurement of the serum and salivary levels of α1 adrenergic receptors. RESULTS: The level of α1 adrenergic receptors was significantly higher in the stimulated and unstimulated saliva of OSCC patients than healthy controls (P = 0.000). However, their serum level was not significantly different between the two groups (P = 0.389). The serum level of α1 adrenergic receptors significantly increased by an increase in OSCC grade. No significant correlation was noted between the serum and salivary levels of α1 adrenergic receptors in OSCC patients. The salivary level of α1 adrenergic receptors was significantly higher in patients with tumors located in the gingiva, compared with other sites. CONCLUSION: Significantly higher salivary level of α1 adrenergic receptors in OSCC patients compared with healthy controls, and no significant difference in their serum level between the two groups may indirectly indicate the over-expression of these receptors in OSCC cells, compared with normal oral mucosa. Further studies and particularly histological analyses are required to confirm this finding.


Asunto(s)
Neoplasias de Cabeza y Cuello/metabolismo , Receptores Adrenérgicos alfa 1/análisis , Saliva/química , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Neoplasias de Cabeza y Cuello/sangre , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Persona de Mediana Edad , Receptores Adrenérgicos alfa 1/sangre , Carcinoma de Células Escamosas de Cabeza y Cuello/sangre , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
7.
Pflugers Arch ; 473(2): 253-271, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33140200

RESUMEN

The bed nucleus of the stria terminalis (BNST) is a forebrain structure, involved in the modulation of neuroendocrine, cardiovascular and autonomic responses. One of the responses is baroreflex activity, which consists in a neural mechanism responsible for keeping the blood pressure within a narrow range of variation. It has been reported that blockade of BNST α1-adrenoceptors increased the bradycardic component of baroreflex. In addition, such receptors are able to modulate glutamate release in this structure. Interestingly, BNST NMDA receptor antagonism and neuronal nitric oxide synthase (nNOS) inhibition led to the same effect of the α1-adrenoceptors blockade on baroreflex bradycardic response. Therefore, the hypothesis of the present study is that BNST noradrenergic transmission interacts with NMDA/NO pathway through α1 adrenoceptors to modulate the baroreflex activity. Male Wistar rats had stainless steel guide cannulas bilaterally implanted in the BNST. Subsequently, a catheter was inserted into the femoral artery for cardiovascular recordings, and into the femoral vein for assessing baroreflex activation. Injection of the noradrenaline reuptake inhibitor reboxetine in the BNST did not modify the tachycardic, but significantly decreased the bradycardic component of baroreflex. Administration of an α1, but not an α2 antagonist into the BNST prior to reboxetine prevented this effect. Likewise, previous injection of NMDA/NO pathway blockers inhibited the effect of reboxetine on bradycardic response. In conclusion, it was demonstrated for the first time the existence of an interaction between BNST noradrenergic, glutamatergic and nitrergic neurotransmissions in the modulation of bradycardic baroreflex response.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Barorreflejo , Corazón/inervación , Óxido Nítrico/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleos Septales/metabolismo , Animales , Presión Arterial , Sistema Nervioso Autónomo/efectos de los fármacos , Barorreflejo/efectos de los fármacos , Frecuencia Cardíaca , Masculino , Neurotransmisores/farmacología , Ratas Wistar , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Núcleos Septales/efectos de los fármacos , Factores de Tiempo
8.
Artículo en Inglés | MEDLINE | ID: mdl-33166680

RESUMEN

Centruroides margaritatus scorpion stings are common in Colombia. However, the cardiovascular toxicity of the venom has not been clarified. AIM: To study the effect and mechanisms of action of the complete venom of C. margaritatus (CmV) on the murine cardiovascular system. METHODS: We evaluated the in vivo effect of CmV LD50 on the mean arterial pressure (MABP), heart rate, and surface electrocardiogram in male adult normotensive Wistar rats. Ex vivo, we evaluated the vascular reactivity of rat aortic rings to increasing concentrations (1 to 60 µg/mL) of CmV using the blockers L-NAME, indomethacin, seratrodast, and prazosin. RESULTS: In the first hour of poisoning, CmV increased the MABP. In the second hour after poisoning, the heart rate decreased as the normalized PR interval and QT corrected increased. After that, cardiovascular shock was demonstrated by a drastic fall in the MABP and signs of cardiac conduction system block. In aortic rings, CmV caused a direct vasoconstrictor effect mediated by alpha-1 adrenergic receptors and counteracted by nitric oxide. CONCLUSION: The direct vascular and probably the cardiac alpha-1 effects likely explain the transient hypertension and the maintenance of cardiac function, while interval lengthening may be due to K+ channel blockage. Afterwards, the effects of both the alpha-1 pathway and the K+ channel pathway converged, resulting in fatal cardiovascular shock. This knowledge could aid in understanding the dynamics of the effects of the venom and in designing treatments to address its cardiovascular effects.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Venenos de Escorpión/toxicidad , Escorpiones/química , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Presión Sanguínea/efectos de los fármacos , Electrocardiografía/métodos , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratas Wistar , Transducción de Señal/efectos de los fármacos
9.
J Cardiovasc Pharmacol ; 76(6): 698-707, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33105324

RESUMEN

Cellular death and survival signaling plays a key role in the progress of adverse cardiac remodeling after acute myocardial infarction (AMI). Therapeutic strategies, such as co-treatment with beta-blocker carvedilol and thyroid hormones (THs), give rise to new approaches that can sustain the cellular homeostasis after AMI. Therefore, we sought to investigate the effects of carvedilol and TH co-administration on apoptosis and survival proteins and on cardiac remodeling after AMI. Male Wistar rats were distributed in 5 groups as follows: sham-operated group (SHAM), infarcted group (MI), infarcted plus carvedilol group (MI+C), infarcted plus TH group (MI+TH), and infarcted plus carvedilol and TH co-treatment group (MI+C+TH). Echocardiographic analysis was performed, and hearts were collected for western blot evaluation. The MI group presented systolic posterior wall thickness loss, an increase in the wall tension index, and an increase in atrial natriuretic peptide tissue levels than the SHAM group. However, in the MI+C+TH group, these parameters were equally to the SHAM group. Moreover, whereas the MI group showed Bax protein expression elevated in relation to the SHAM group, the MI+C+TH group presented Bax reduction and also Akt activation compared with the MI group. In addition, the MI+TH group revealed beta-1 adrenergic receptor (ß1AR) upregulation compared with the MI and MI+C groups, whereas the MI+C+TH group presented lower levels of ß1AR in relation to the SHAM and MI+TH groups. In conclusion, we suggest that carvedilol and TH co-administration may mediate its cardioprotective effects against adverse cardiac remodeling post-AMI through the Bax reduction, Akt activation, and ß1AR decrease.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Carvedilol/farmacología , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Tiroxina/farmacología , Triyodotironina/farmacología , Animales , Modelos Animales de Enfermedad , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Ratas Wistar , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal
10.
Eur J Pharmacol ; 885: 173423, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32750368

RESUMEN

In a cell line, stably expressing α1A-adrenoceptors fused to the mCherry red fluorescent protein, noradrenaline, methoxamine, and oxymetazoline induced concentration-dependent increases in intracellular calcium. All of these agents increase α1A-adrenoceptor phosphorylation and internalization. Transient co-expression of these receptors with Rab proteins tagged with the enhanced Green Fluorescent Protein was employed to estimate α1A-adrenoceptor-Rab interaction using Förster Resonance Energy Transfer. Noradrenaline and methoxamine increased α1A-adrenoceptor interaction with Rab5 and Rab7 but did not modify it with Rab9. Oxymetazoline induced adrenoceptor interaction with Rab5 and Rab9 and only an insignificant increase in Rab7 signal. Phorbol myristate acetate increased α1A-adrenoceptor interaction with Rab5 and Rab9 but did not modify it with Rab7. The agonists and the active phorbol ester, all of which induce receptor phosphorylation and internalization, favor receptor interaction with Rab5, i.e., association with early endosomes. Cell stimulation with phorbol myristate acetate induced the α1A-adrenoceptors to interact with the late endosomal marker, Rab9, suggesting that the receptors are directed to slow recycling endosomes once they have transited to the Trans-Golgi network to be retrieved to the plasma membrane. The agonists noradrenaline and methoxamine likely induce a faster recycling and might direct some of the adrenoceptors toward degradation and/or very slow recycling to the plasma membrane. Oxymetazoline produced a mixed pattern of interaction with the Rab proteins. These data indicate that α1A-adrenoceptor agonists can trigger different vesicular traffic and receptor fates within the cells.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Ésteres del Forbol/farmacología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Proteínas de Unión al GTP rab/efectos de los fármacos , Calcio/metabolismo , Línea Celular , Endosomas/efectos de los fármacos , Humanos , Proteínas Luminiscentes , Metoxamina/farmacología , Norepinefrina/farmacología , Oximetazolina/farmacología , Fosforilación , Acetato de Tetradecanoilforbol/farmacología , Proteínas de Unión al GTP rab5/efectos de los fármacos , Red trans-Golgi/efectos de los fármacos , Proteína Fluorescente Roja
11.
J Trace Elem Med Biol ; 62: 126569, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32563862

RESUMEN

Boron is an essential trace element in cellular metabolism; however, the molecular mechanism of boron in the heart is unclear. In this study, we examined the effect of sodium tetraborate (as boron source) as a possible protective agent or competitive inhibitor of cardiac hypertrophy in an in vitro murine model. We evaluated different previously reported sodium tetraborate concentrations and it was found that 13 µM improves viability without affecting the cellular structure. We demonstrated that cardiomyocytes pretreated with sodium tetraborate prevents cellular damage induced by isoproterenol (cardioprotective effect) by increasing proliferation rate and inhibiting apoptosis. In addition, the reduction of the expression of the α1AR and ß1AR adrenergic receptors as well as Erk1/2 was notable. Consequently, the expression of the early response genes c-myc, c-fos and c-jun was delayed. Also, the expression of GATA-4, NFAT, NKx2.5 and myogenin transcription factors involved in sarcomere synthesis declined. In contrast, cardiomyocytes, when treated simultaneously with sodium tetraborate and isoproterenol, did not increase their size (cytoplasmic gain), but an increase in apoptosis levels was observed; therefore, the proliferation rate was reduced. Although the mRNA levels of α1AR and ß1AR as well as Erk1/2 and Akt1 were low at 24 h, their expression increased to 48 h. Notably, the mRNA of expression levels of c-myc, c-fos and c-jun were lower than those determined in the control, while the transcription factors GATA-4, MEF2c, Nkx2.5, NFAT and CDk9 were determined in most cells. These results suggest that pretreatment with sodium tetraborate in cardiomyocytes inhibits the hypertrophic effect. However, sodium tetraborate attenuates isoproterenol induced hypertrophy damage in cardiomyocytes when these two compounds are added simultaneously.


Asunto(s)
Boratos/farmacología , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Cardiotónicos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Boratos/administración & dosificación , Boratos/efectos adversos , Cardiotónicos/efectos adversos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Isoproterenol/administración & dosificación , Isoproterenol/efectos adversos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos BALB C , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos beta 2/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
12.
Exp Brain Res ; 238(5): 1293-1303, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32322928

RESUMEN

Somatosensory afferent transmission strength is controlled by several presynaptic mechanisms that reduce transmitter release at the spinal cord level. We focused this investigation on the role of α-adrenoceptors in modulating sensory transmission in low-threshold myelinated afferents and in pathways mediating primary afferent depolarization (PAD) of neonatal mouse spinal cord. We hypothesized that the activation of α-adrenoceptors depresses low threshold-evoked synaptic transmission and inhibits pathways mediating PAD. Extracellular field potentials (EFPs) recorded in the deep dorsal horn assessed adrenergic modulation of population monosynaptic transmission, while dorsal root potentials (DRPs) recorded at root entry zone assessed adrenergic modulation of PAD. We found that noradrenaline (NA) and the α1-adrenoceptor agonists phenylephrine and cirazoline depressed synaptic transmission (by 15, 14 and 22%, respectively). DRPs were also depressed by NA, phenylephrine and cirazoline (by 62, 30, and 64%, respectively), and by the α2-adrenoceptor agonist clonidine, although to a lower extent (20%). We conclude that NA depresses monosynaptic transmission of myelinated afferents onto deep dorsal horn neurons via α1-adrenoceptors and inhibits interneuronal pathways mediating PAD through the activation of α1- and α2-adrenoceptors. The functional significance of these modulatory actions in shaping cutaneous and muscle sensory information during motor behaviors requires further study.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Fenómenos Electrofisiológicos/fisiología , Fibras Nerviosas Mielínicas/fisiología , Neuronas Aferentes/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Receptores Adrenérgicos alfa 2/fisiología , Asta Dorsal de la Médula Espinal/fisiología , Transmisión Sináptica/fisiología , Animales , Animales Recién Nacidos , Fenómenos Electrofisiológicos/efectos de los fármacos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Vías Nerviosas/fisiología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
13.
J Biol Chem ; 295(21): 7404-7417, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32303636

RESUMEN

G protein-coupled receptors (GPCRs) use a series of conserved microswitches to transmit signals across the cell membrane via an allosteric network encompassing the ligand-binding site and the G protein-binding site. Crystal structures of GPCRs provide snapshots of their inactive and active states, but poorly describe the conformational dynamics of the allosteric network that underlies GPCR activation. Here, we analyzed the correlation between ligand binding and receptor conformation of the α1A-adrenoreceptor, a GPCR that stimulates smooth muscle contraction in response to binding noradrenaline. NMR of [13CϵH3]methionine-labeled α1A-adrenoreceptor variants, each exhibiting differing signaling capacities, revealed how different classes of ligands modulate the conformational equilibria of this receptor. [13CϵH3]Methionine residues near the microswitches exhibited distinct states that correlated with ligand efficacies, supporting a conformational selection mechanism. We propose that allosteric coupling among the microswitches controls the conformation of the α1A-adrenoreceptor and underlies the mechanism of ligand modulation of GPCR signaling in cells.


Asunto(s)
Receptores Adrenérgicos alfa 1/química , Regulación Alostérica , Cristalografía por Rayos X , Humanos , Ligandos , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Receptores Adrenérgicos alfa 1/metabolismo
14.
Eur J Cell Biol ; 99(2-3): 151072, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32113707

RESUMEN

The possibility that glycogen synthase kinase 3 (GSK3) could modulate α1A-adrenergic receptor (α1A-AR) function and regulation was tested employing LNCaP and HEK293 cells transfected to express the enhanced green fluorescent protein-tagged human α1A-AR. Receptor phosphorylation and internalization, intracellular free calcium, α1A-AR-GSK3 colocalization, and coimmunoprecipitation were studied. The effects of the pharmacological GSK3 inhibitor, SB-216763, and the coexpression of a dominant-negative mutant of this kinase, as well as the signaling, desensitization, and internalization of receptors with S229, S258, S352, and S381 substitutions for alanine or aspartate, were also determined. SB-216763 inhibited agonist- and phorbol myristate acetate (PMA)-mediated α1A-AR phosphorylation, reduced oxymetazoline-induced desensitization, and magnified that induced by PMA. Agonists and PMA increased receptor-GSK3 colocalization and coimmunoprecipitation. Expression of a dominant-negative GSK3 mutant reduced agonist- but not PMA-induced receptor internalization. α1A-AR with the GSK3 putative target sites mutated to alanine exhibited reduced phosphorylation and internalization in response to agonists and increased PMA-induced desensitization. Agonist-induced, but not PMA-induced, receptor-ß arrestin intracellular colocalization was diminished in cells expressing the GSK3 putative target sites mutated to alanine. Our data indicated that GSK3 exerts a dual action on α1A-AR participating in agonist-mediated desensitization and internalization and avoiding PMA-induced desensitization.


Asunto(s)
Glucógeno Sintasa Quinasa 3/uso terapéutico , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/farmacología , Humanos
15.
Int J Mol Sci ; 21(3)2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31991781

RESUMEN

The ventral tegmental area (VTA) plays an important role in the reward and motivational processes that facilitate the development of drug addiction. Presynaptic α1-AR activation modulates glutamate and Gamma-aminobutyric acid (GABA) release. This work elucidates the role of VTA presynaptic α1-ARs and their modulation on glutamatergic and GABAergic neurotransmission during cocaine sensitization. Excitatory and inhibitory currents (EPSCs and IPSCs) measured by a whole cell voltage clamp show that α1-ARs activation increases EPSCs amplitude after 1 day of cocaine treatment but not after 5 days of cocaine injections. The absence of a pharmacological response to an α1-ARs agonist highlights the desensitization of the receptor after repeated cocaine administration. The desensitization of α1-ARs persists after a 7-day withdrawal period. In contrast, the modulation of α1-ARs on GABA neurotransmission, shown by decreases in IPSCs' amplitude, is not affected by acute or chronic cocaine injections. Taken together, these data suggest that α1-ARs may enhance DA neuronal excitability after repeated cocaine administration through the reduction of GABA inhibition onto VTA dopamine (DA) neurons even in the absence of α1-ARs' function on glutamate release and protein kinase C (PKC) activation. α1-AR modulatory changes in cocaine sensitization increase our knowledge of the role of the noradrenergic system in cocaine addiction and may provide possible avenues for therapeutics.


Asunto(s)
Cocaína/metabolismo , Neuronas Dopaminérgicas/metabolismo , Ácido Glutámico/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Cocaína/administración & dosificación , Trastornos Relacionados con Cocaína/etiología , Trastornos Relacionados con Cocaína/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Masculino , Modelos Biológicos , Técnicas de Placa-Clamp , Terminales Presinápticos/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
16.
Eur J Pharmacol ; 867: 172846, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31811856

RESUMEN

Cells expressing eGFP-tagged Rab5 (wild-type or the GDP-Rab5 mutant) and the DsRed-tagged α1B-adrenergic receptors were employed and the roles of GRK2 were studied utilizing paroxetine and the dominant-negative mutant of GRK2 (DN-GRK2). The following parameters were studied: a) FRET (as an index of α1B-adrenergic receptor-Rab5 interaction): b) intracellular accumulation of DsRed fluorescence (receptor internalization); c) α1B-adrenergic receptor phosphorylation, and d) noradrenaline-induced increase in intracellular calcium concentration. Noradrenaline increased α1B-adrenergic receptor-Rab5 interaction, which was blocked by paroxetine and by expression of the dominant-negative GRK2 mutant. Similarly, paroxetine and expression of the DN-GRK2 or the GDP-Rab5 mutants markedly decreased receptor internalization, α1B-adrenergic receptor phosphorylation, and attenuated the ability of the adrenergic agonist to induce homologous desensitization (calcium signaling). The S406, 410,412A α1B-adrenergic receptor mutant did not reproduce the actions of GRK2 inhibition. The data indicate that GRK2 and Rab5 play key roles in α1B-adrenergic receptor phosphorylation, internalization, and desensitization. The possibility that Rab5 might form part of a signaling complex is suggested, as well as that GDP-Rab5 might interfere with the ability of GRK2 to catalyze α1B-adrenergic receptor phosphorylation.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Transferencia Resonante de Energía de Fluorescencia , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Células HEK293 , Humanos , Mutación , Norepinefrina/farmacología , Paroxetina/farmacología , Fosforilación/efectos de los fármacos , Proteínas de Unión al GTP rab5/genética
17.
Life Sci ; 239: 117048, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31730867

RESUMEN

Benign prostatic hyperplasia (BPH) is an aging-related and progressive disease linked to an up-regulation of α1-adrenoceptors. The participation of EGF receptors (EGFR) in the GPCRs' signalosome has been described but so far data about the contribution of these receptors to prostatic stromal hyperplasia are scanty. We isolated and cultured vimentin-positive prostate stromal cells obtained from BPH patients. According to intracellular Ca2+ measurements, cell proliferation and Western blotting assays, these cultured hyperplastic stromal cells express functional α1-adrenoceptors and EGFR, and proliferate in response to the α1-adrenoceptor agonist phenylephrine. Interestingly, in these cells the inhibition of EGFR signaling with GM6001, CRM197, AG1478 or PD98059 was associated with full blockage of α1-adrenoceptor-mediated cell proliferation, while cell treatment with each inhibitor alone did not alter basal cell growth. Moreover, the co-incubation of AG1478 (EGFR inhibitor) with α1A/α1D-adrenoceptor antagonists showed no additive inhibitory effect. These findings highlight a putative role of EGFR signaling to α1-adrenoceptor-mediated human prostate hyperplasia, suggesting that the inhibition of this transactivation cascade could be useful to reduce BPH progression.


Asunto(s)
Receptores ErbB/metabolismo , Hiperplasia Prostática/metabolismo , Receptores Adrenérgicos alfa 1/fisiología , Antagonistas de Receptores Adrenérgicos alfa 1/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Humanos , Hiperplasia/metabolismo , Masculino , Piperazinas/farmacología , Cultivo Primario de Células , Próstata/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma
18.
Biochim Biophys Acta Mol Cell Res ; 1866(10): 1509-1519, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325464

RESUMEN

Phosphorylation of the human α1B-adrenergic receptor (fused with the green fluorescent protein) was studied employing the inducible Flp-ln HEK293 T-Rex system for expression. Serine/alanine substitutions were performed in five sites corresponding to those previously identified as phosphorylation targets in the hamster ortholog. Desensitization was decreased in these mutants but receptor phosphorylation was still clearly detected. The protein phosphorylation of the wild-type receptor (fused to the green fluorescent protein) was studied, using mass spectrometry, under baseline and stimulated conditions (noradrenaline or phorbol myristate acetate). Basal phosphorylation was detected at sites located at the intracellular loop 3 and carboxyl terminus, and the number of sites detected increased under agonist activation and stimulation of protein kinase C. The phosphorylation patterns differed under the distinct conditions. Three of the phosphorylation sites detected in this work corresponded to those observed in the hamster receptor. The phosphorylation sites detected included the following: a) at the intracellular loop 3: serines 246, 248, 257, 267, and 277; and threonines 252, 264, and 268, and b) at the carboxyl terminus: serines 396, 400, 402, 406, 423, 425, 427, 455, and 470, and threonines 387, 392, 420, and 475. Our data indicate that complex phosphorylation patterns exist and suggest the possibility that such differences could be relevant in receptor function and subcellular localization.


Asunto(s)
Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Acetato de Tetradecanoilforbol/metabolismo , Sustitución de Aminoácidos , Animales , Cricetinae , Quinasas MAP Reguladas por Señal Extracelular , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Fosforilación , Proteína Quinasa C/metabolismo
19.
Eur J Pharmacol ; 858: 172498, 2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31238063

RESUMEN

Metformin is a widely used drug for the treatment of type 2 Diabetes Mellitus. Several studies have also suggested that metformin decreases blood pressure; although an interaction with α-adrenoceptors has been proposed, this mechanism needs to be further investigated. Since α1-adrenoceptors play a significant role to regulate vascular tone, this study has analysed the potential ability of metformin to block α1-adrenoceptors in rat aorta and tail artery. For this purpose, the contractile responses induced by noradrenaline, methoxamine, and phenylephrine were determined in the absence or presence of metformin in rat aorta and tail artery rings. In both arteries, noradrenaline, methoxamine, and phenylephrine produced concentration-dependent contractile responses. Interestingly, the contractile responses to noradrenaline, methoxamine, and phenylephrine were significantly and differentially blocked by metformin (1, 3.1 and/or 10 mM) but not by vehicle. These results suggest that metformin is capable to block α1-adrenoceptors and may explain, at least in part, the anti-hypertensive effect observed in several clinical trials.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Aorta/efectos de los fármacos , Aorta/fisiología , Metformina/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Cola (estructura animal)/irrigación sanguínea , Animales , Masculino , Metoxamina/farmacología , Fenilefrina/farmacología , Ratas , Ratas Wistar , Vasoconstricción/efectos de los fármacos
20.
Br J Pharmacol ; 176(14): 2343-2357, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30740663

RESUMEN

α1 -Adrenoceptors are seven transmembrane domain GPCRs involved in numerous physiological functions controlled by the endogenous catecholamines, noradrenaline and adrenaline, and targeted by drugs useful in therapeutics. Three separate genes, whose products are named α1A -, α1B -, and α1D - adrenoceptors, encode these receptors. Although the existence of multiple α1 -adrenoceptors has been acknowledged for almost 25 years, the specific functions regulated by each subtype are still largely unknown. Despite the limited comprehension, the identification of a single class of subtype-selective ligands for the α1A - adrenoceptors, the so-called α-blockers for prostate dysfunction, has led to major improvement in therapeutics, demonstrating the need for continued efforts in the field. This review article surveys the tissue distribution of the three α1 -adrenoceptor subtypes in the cardiovascular system, genitourinary system, and CNS, highlighting the functions already identified as mediated by the predominant activation of specific subtypes. In addition, this review covers the recent advances in the understanding of the molecular mechanisms involved in the regulation of each of the α1 -adrenoceptor subtypes by phosphorylation and interaction with proteins involved in their desensitization and internalization. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.


Asunto(s)
Receptores Adrenérgicos alfa 1/metabolismo , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA