Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.659
Filtrar
1.
J Mol Cell Cardiol ; 161: 86-97, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34375616

RESUMEN

Delayed rectifier K+ current (IKs) is a key contributor to repolarization of action potentials. This study investigated the mechanisms underlying the adrenoceptor-induced potentiation of IKs in pulmonary vein cardiomyocytes (PVC). PVC were isolated from guinea pig pulmonary vein. The action potentials and IKs current were recorded using perforated and conventional whole-cell patch-clamp techniques. The expression of IKs was examined using immunocytochemistry and Western blotting. KCNQ1, a IKs pore-forming protein was detected as a signal band approximately 100 kDa in size, and its immunofluorescence signal was found to be mainly localized on the cell membrane. The IKs current in PVC was markedly enhanced by both ß1- and ß2-adrenoceptor stimulation with a negative voltage shift in the current activation, although the potentiation was more effectively induced by ß2-adrenoceptor stimulation than ß1-adrenoceptor stimulation. Both ß-adrenoceptor-mediated increases in IKs were attenuated by treatment with the adenylyl cyclase (AC) inhibitor or protein kinase A (PKA) inhibitor. Furthermore, the IKs current was increased by α1-adrenoceptor agonist but attenuated by the protein kinase C (PKC) inhibitor. PVC exhibited action potentials in normal Tyrode solution which was slightly reduced by HMR-1556 a selective IKs blocker. However, HMR-1556 markedly reduced the ß-adrenoceptor-potentiated firing rate. The stimulatory effects of ß- and α1-adrenoceptor on IKs in PVC are mediated via the PKA and PKC signal pathways. HMR-1556 effectively reduced the firing rate under ß-adrenoceptor activation, suggesting that the functional role of IKs might increase during sympathetic excitation under in vivo conditions.


Asunto(s)
Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Miocitos Cardíacos/metabolismo , Venas Pulmonares/metabolismo , Receptores Adrenérgicos/metabolismo , Potenciales de Acción/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Cobayas , Atrios Cardíacos/metabolismo , Isoproterenol/farmacología , Canal de Potasio KCNQ1/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Proteína Quinasa C/metabolismo , Venas Pulmonares/citología , Transducción de Señal
2.
Cytokine ; 142: 155497, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33770644

RESUMEN

In brain astrocytes, noradrenaline (NA) has been shown to up-regulate IL-6 production via ß-adrenoceptors (ARs). However, the underlying intracellular mechanisms for this regulation are not clear, and it remains unknown whether α-ARs are involved. In this study, we investigated the AR-mediated regulation of IL-6 mRNA levels in the cultured astrocytes from rat spinal cord. NA, the α1-agonist phenylephrine, and the ß-agonist isoproterenol increased IL-6 mRNA levels. The phenylephrine-induced IL-6 increase was accompanied by an increase in ERK phosphorylation, and these effects were blocked by inhibitors of PKC and ERK. The isoproterenol-induced IL-6 increase was accompanied by an increase in CREB phosphorylation, and these effects were blocked by a PKA inhibitor. Our results indicate that IL-6 increases by α1- and ß-ARs are mediated via the PKC/ERK and cAMP/PKA/CREB pathways, respectively. Moreover, conditioned medium collected from astrocytes treated with the α2-AR agonist dexmedetomidine, increased IL-6 mRNA in other astrocytes. In this study, we elucidate that α1- and α2-ARs, in addition to ß-ARs, promote IL-6 transcription through different pathways in spinal cord astrocytes.


Asunto(s)
Astrocitos/metabolismo , Interleucina-6/genética , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Médula Espinal/citología , Transcripción Genética , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Interleucina-6/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
3.
J Pharmacol Sci ; 145(3): 228-240, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33602503

RESUMEN

Astrocytes are glial cells with numerous fine processes which are important for the functions of the central nervous system. The activation of ß-adrenoceptors induces process formation of astrocytes via cyclic AMP (cAMP) signaling. However, the role of α-adrenoceptors in the astrocyte morphology has not been elucidated. Here, we examined it by using cultured astrocytes from neonatal rat spinal cords and cortices. Exposure of these cells to noradrenaline and the ß-adrenoceptor agonist isoproterenol increased intracellular cAMP levels and induced the formation of processes. Noradrenaline-induced process formation was enhanced with the α1-adrenoceptor antagonist prazosin and α2-adrenoceptor antagonist atipamezole. Atipamezole also enhanced noradrenaline-induced cAMP elevation. Isoproterenol-induced process formation was not inhibited by the α1-adrenoceptor agonist phenylephrine but was inhibited by the α2-adrenoceptor agonist dexmedetomidine. Dexmedetomidine also inhibited process formation induced by the adenylate cyclase activator forskolin and the membrane-permeable cAMP analog dibutyryl-cAMP. Moreover, dexmedetomidine inhibited cAMP-independent process formation induced by adenosine or the Rho-associated kinase inhibitor Y27632. In the presence of propranolol, noradrenaline inhibited Y27632-induced process formation, which was abolished by prazosin or atipamezole. These results demonstrate that α-adrenoceptors inhibit both cAMP-dependent and -independent astrocytic process formation.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Receptores Adrenérgicos alfa/fisiología , Receptores Adrenérgicos beta/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Dexmedetomidina/farmacología , Imidazoles/farmacología , Isoproterenol/farmacología , Norepinefrina/farmacología , Prazosina/farmacología , Ratas Wistar , Transducción de Señal
4.
Am J Physiol Regul Integr Comp Physiol ; 320(2): R105-R116, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33175586

RESUMEN

Hypoxia tolerance in the vertebrate brain often involves chemical modulators that arrest neuronal activity to conserve energy. However, in intact networks, it can be difficult to determine whether hypoxia triggers modulators to stop activity in a protective manner or whether activity stops because rates of ATP synthesis are insufficient to support network function. Here, we assessed the extent to which neuromodulation or metabolic limitations arrest activity in the respiratory network of bullfrogs-a circuit that survives moderate periods of oxygen deprivation, presumably, by activating an inhibitory noradrenergic pathway. We confirmed that hypoxia and norepinephrine (NE) reduce network output, consistent with the view that hypoxia may cause the release of NE to inhibit activity. However, these responses differed qualitatively; hypoxia, but not NE, elicited a large motor burst and silenced the network. The stereotyped response to hypoxia persisted in the presence of both NE and an adrenergic receptor blocker that eliminates sensitivity to NE, indicating that noradrenergic signaling does not cause the arrest. Pharmacological inhibition of glycolysis and mitochondrial respiration recapitulated all features of hypoxia on network activity, implying that reduced ATP synthesis underlies the effects of hypoxia. Finally, activating modulatory mechanisms that dampen neuronal excitability when ATP levels fall, KATP channels and AMP-dependent protein kinase, did not resemble the hypoxic response. These results suggest that energy failure-rather than inhibitory modulation-silences the respiratory network during hypoxia and emphasize the need to account for metabolic limitations before concluding that modulators arrest activity as an adaptation for energy conservation in the nervous system.


Asunto(s)
Tronco Encefálico/fisiología , Metabolismo Energético/fisiología , Consumo de Oxígeno/fisiología , Rana catesbeiana/fisiología , Adenosina Trifosfato/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Desoxiglucosa/farmacología , Femenino , Humanos , Ácido Yodoacético/farmacología , Norepinefrina/farmacología , Prazosina/farmacología
5.
Biomed Res Int ; 2020: 6632359, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33381567

RESUMEN

INTRODUCTION: Bladder outlet obstruction (BOO) occurs in more than 20 percent of the adult population and may lead to changes in the structure and function of the bladder. The main objective of the study was to evaluate the expression of Toll-like receptor 4 (TLR 4) and Toll-like receptor 9 (TLR 9) in the animal model of BOO as potential triggers of the inflammation phase in the bladder. In addition, the modulating effect of alpha-1 adrenergic antagonist (tamsulosin) on TLR 4 and TLR 9 expression and inflammatory markers was assessed. Material and Methods. Thirty-two male, 9-week-old Sprague Dawley rats were randomly divided into 4 groups: SOP-sham-operated rats with a placebo (water); SOB-sham-operated rats with an alpha-1 adrenergic antagonist; BOOP-rats with BOO and a placebo; and BOOB-rats with BOO and an alpha-1 adrenergic antagonist. The rats were given a placebo or alpha-1 adrenergic antagonist for 15 days. Next, urine and the bladder were collected from the rats for histopathological and biochemical study. RESULTS: Histopathological analysis showed chronic inflammation without acute inflammation in the bladder. TLR 4 showed positive cytoplasmic reactivity in the urothelium and the smooth muscles of the bladder. TLR 9 showed positive cytoplasmic reactivity only in the urothelium. BOO caused an increase in TLR 4 and TLR 9 expression. Furthermore, treatment with an alpha-1 adrenergic antagonist had no significant effect on TLR 4 and TLR 9 expression in rats with BOO. BOO caused a significant increase in urine concentration of interleukin 6 (IL-6), while alpha-1 antagonist reduced the urine concentration of IL-6 and the concentration of interleukin 18 (IL-18). CONCLUSIONS: The results suggest the participation of TLR 4 and TLR 9 receptors in the induction of inflammation in the bladder, which is the first phase in the development of pathophysiological changes in BOO.


Asunto(s)
Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Animales , Peso Corporal , Modelos Animales de Enfermedad , Inflamación , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 1/metabolismo , Tamsulosina/farmacología , Vejiga Urinaria/metabolismo
6.
Nat Commun ; 11(1): 6157, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33268792

RESUMEN

Norepinephrine adjusts sensory processing in cortical networks and gates plasticity enabling adaptive behavior. The actions of norepinephrine are profoundly altered by recreational drugs like ethanol, but the consequences of these changes on distinct targets such as astrocytes, which exhibit norepinephrine-dependent Ca2+ elevations during vigilance, are not well understood. Using in vivo two-photon imaging, we show that locomotion-induced Ca2+ elevations in mouse astroglia are profoundly inhibited by ethanol, an effect that can be reversed by enhancing norepinephrine release. Vigilance-dependent astroglial activation is abolished by deletion of α1A-adrenergic receptor from astroglia, indicating that norepinephrine acts directly on these ubiquitous glial cells. Ethanol reduces vigilance-dependent Ca2+ transients in noradrenergic terminals, but has little effect on astroglial responsiveness to norepinephrine, suggesting that ethanol suppresses their activation by inhibiting norepinephrine release. Since abolition of astroglia Ca2+ activation does not affect motor coordination, global suppression of astroglial networks may contribute to the cognitive effects of alcohol intoxication.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Calcio/metabolismo , Etanol/farmacología , Norepinefrina/farmacología , Vigilia/efectos de los fármacos , Intoxicación Alcohólica/genética , Intoxicación Alcohólica/metabolismo , Intoxicación Alcohólica/fisiopatología , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Transportador 1 de Aminoácidos Excitadores/deficiencia , Transportador 1 de Aminoácidos Excitadores/genética , Femenino , Regulación de la Expresión Génica , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Noqueados , Microscopía de Fluorescencia por Excitación Multifotónica , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Norepinefrina/antagonistas & inhibidores , Receptores Adrenérgicos alfa 1/deficiencia , Receptores Adrenérgicos alfa 1/genética , Vigilia/fisiología , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
7.
FASEB J ; 34(11): 15448-15461, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32985027

RESUMEN

The LPBN (lateral parabrachial nucleus) plays an important role in feeding control. CGRP (calcitonin gene-related peptide) LPBN neurons activation mediates the anorectic effects of different gut-derived peptides, including amylin. Amylin and its long acting analog sCT (salmon calcitonin) exert their anorectic actions primarily by directly activating neurons located in the area postrema (AP). A large proportion of projections from the AP and the adjacent nucleus of the solitary tractNTS to the LPBN, are noradrenergic (NA), and amylin-activated NAAP neurons are critical in mediating amylin's hypophagic effects. Here, we determine the functional role of NAAP amylin activated neurons to activate CGRP and non-CGRP LPBN neurons. To this end, NA was specifically depleted in the rat LPBN through a stereotaxic microinfusion of 6-OHDA, a neurotoxic agent that destroys NA terminals. While amylin (50 µg/kg) and sCT (5 µg/kg) reduced eating in sham-lesioned rats, no reduction in feeding occurred in NA-depleted animals. Further, the amylin-induced c-Fos response in the LPBN and c-Fos/CGRP colocalization were reduced in NA-depleted animals compared to controls. We conclude that AP â†’ LPBN NA signaling, through the activation of LPBN CGRP neurons, mediates part of amylin's hypophagic effect.


Asunto(s)
Anorexia/tratamiento farmacológico , Calcitonina/metabolismo , Ingestión de Alimentos/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Neuronas/efectos de los fármacos , Norepinefrina/farmacología , Núcleos Parabraquiales/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Agonistas de los Receptores de Amilina/farmacología , Animales , Anorexia/metabolismo , Anorexia/patología , Calcitonina/genética , Ingestión de Alimentos/efectos de los fármacos , Masculino , Núcleos Parabraquiales/metabolismo , Ratas , Ratas Sprague-Dawley
8.
FASEB J ; 34(11): 14892-14904, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32939891

RESUMEN

Renal fibrosis is a common pathological hallmark of chronic kidney disease (CKD). Renal sympathetic nerve activity is elevated in patients and experimental animals with CKD and contributes to renal interstitial fibrosis in obstructive nephropathy. However, the mechanisms underlying sympathetic overactivation in renal fibrosis remain unknown. Norepinephrine (NE), the main sympathetic neurotransmitter, was found to promote TGF-ß1-induced epithelial-mesenchymal transition (EMT) and fibrotic gene expression in the human renal proximal epithelial cell line HK-2. Using both genetic and pharmacological approaches, we identified that NE binds Gαq-coupled α1-adrenoceptor (α1-AR) to enhance EMT of HK-2 cells by activating p38/Smad3 signaling. Inhibition of p38 diminished the NE-exaggerated EMT process and increased the fibrotic gene expression in TGF-ß1-treated HK-2 cells. Moreover, the pharmacological blockade of α1-AR reduced the kidney injury and renal fibrosis in a unilateral ureteral obstruction mouse model by suppressing EMT in the kidneys. Thus, sympathetic overactivation facilitates EMT of renal epithelial cells and fibrosis via the α1-AR/p38/Smad3 signaling pathway, and α1-AR inhibition may be a promising approach toward treating renal fibrosis.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Insuficiencia Renal Crónica/metabolismo , Tamsulosina/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapéutico , Agonistas alfa-Adrenérgicos/farmacología , Animales , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/etiología , Proteína smad3/metabolismo , Tamsulosina/uso terapéutico , Factor de Crecimiento Transformador beta/farmacología , Obstrucción Uretral/complicaciones , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Nat Prod Res ; 34(23): 3369-3372, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30698035

RESUMEN

Oxalis pes-caprae L. is a plant of the Oxalidaceae family, from which several compounds have been previously identified. Recently, we showed that an Oxalis pes-caprae L. extract inhibits the vasopressor effect of noradrenaline. In this work we aimed to explore the mechanisms involved in this effect. The results confirmed that the flavonoid fraction present in the extract inhibits noradrenaline-induced contractions and that this effect is concentration-dependent. Also, a parallel shift to the right in the noradrenaline concentration-response curve was observed, suggesting a decrease in efficacy and also in potency. Together these results support the assumption that the extract could exert a non-competitive antagonism on the α-adrenergic receptors. However, experiments in the presence of competitive antagonists for α-adrenergic receptor sub-types (i.e. prazosin, yohimbine and phentolamine) showed that the effect may not be directly mediated by α-adrenergic receptors. Thus, the interaction of this extract with the adrenergic system remains to be confirmed.


Asunto(s)
Oxalidaceae/química , Extractos Vegetales/farmacología , Polifenoles/análisis , Arterias Torácicas/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Humanos , Persona de Mediana Edad , Norepinefrina/farmacología , Extractos Vegetales/química , Hojas de la Planta/química , Prazosina/farmacología , Receptores Adrenérgicos alfa/efectos de los fármacos , Receptores Adrenérgicos alfa/metabolismo , Yohimbina/farmacología
10.
Bosn J Basic Med Sci ; 20(1): 88-98, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-29984678

RESUMEN

Catecholamines have both anti-inflammatory and vasoactive properties. A decreased cardiac response to catecholamines has been associated with a high risk of death in sepsis and septic shock. The aim of this study was to investigate the effects of epinephrine (EPI) on heart rate variability (HRV) and autonomic balance, as well as cytokine levels, in a rat sepsis model. Thirty-six male Sprague-Dawley rats were assigned to 4 experimental groups and 2 control groups of 6 rats each. The rats in the experimental groups were inoculated with a lipopolysaccharide (LPS, endotoxin) to establish a sepsis model. Group A received only LPS; group B received LPS, antecedent EPI and the nonselective ß-blocker propranolol; group C received LPS and antecedent EPI; and group D received LPS, antecedent EPI and the selective ß1-blocker esmolol. One control group received EPI and the other received saline placebo. Heart rate variability (HRV) was analyzed and tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1ß (IL-1ß) levels were measured. Measurements were carried out at baseline, at 0 hour after EPI infusion, and at 0.5, 2, and 4 hours after LPS inoculation. There were significant differences in HRV and cytokine levels between the groups, indicating that LPS infusion caused autonomic imbalance. Antecedent EPI significantly decreased the level of TNF-α in group C compared with group A in which TNF-α level peaked at 2 hours and then declined. Propranolol (group B) but not esmolol (group D) administration resulted in elevated TNF-α levels, comparable to those observed in group A. In conclusion, antecedent administration of EPI in a rat sepsis model inhibits the production of TNF-α possibly via the ß2-adrenoceptor.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Citocinas/efectos de los fármacos , Epinefrina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Sepsis/metabolismo , Sepsis/fisiopatología , Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Frecuencia Cardíaca/fisiología , Masculino , Propanolaminas/farmacología , Propranolol/farmacología , Ratas , Ratas Sprague-Dawley , Sepsis/tratamiento farmacológico
11.
Anaesthesia ; 74(11): 1389-1396, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31389614

RESUMEN

We evaluated the effect of adrenaline on human skin microcirculation (nutritive and sub-papillary) and systemic cardiovascular variables after it was added to lidocaine in infraclavicular brachial plexus blocks. Twelve healthy, non-smoking male volunteers were included, each attending two study sessions 2 weeks apart, and they were studied using a crossover design. In both sessions, they received an ultrasound-guided infraclavicular brachial plexus block in the non-dominant arm with 0.4 ml.kg-1 lidocaine, 15 mg.ml-1 with or without adrenaline 5 µg.ml-1 . Microcirculation was assessed by laser Doppler fluxmetry (sub-papillary blood flow), capillary video microscopy (nutritive blood flow) and continuous temperature measurements. Heart rate and arterial pressure were recorded continuously and non-invasively. Median (IQR [range]) sub-papillary blood flow increased substantially 30 min after the brachial plexus block, from 8.5 (4.4-13.5 [2.9-28.2]) to 162.7 (111.0-197.8 [9.5-206.7]) arbitrary units with adrenaline (p = 0.017), and from 6.9 (5.3-28.5 [1.8-42.1] to 133.7 (16.5-216.7 [1.0-445.0] arbitrary units without adrenaline (p = 0.036). Nutritive blood flow (functional capillary density, capillaries.mm-2 , measured at the dorsal side of the hand) decreased in the blocked extremity when adrenaline was used as adjuvant, from median (IQR [range]) 45 (36-52 [26-59]) to 38 (29-41 [26-42]), p = 0.028, whereas no significant change occurred without adrenaline. Median finger skin temperature (°C) increased by 44% (data pooled) with no significant differences between the groups. No significant changes were found in the systemic cardiovascular variables with or without adrenaline. We conclude that lidocaine infraclavicular brachial plexus blocks caused an increase in skin sub-papillary blood flow. The addition of adrenaline produced stronger and longer lasting blocks, but decreased the nutritive blood flow.


Asunto(s)
Anestésicos Locales/farmacología , Bloqueo del Plexo Braquial/métodos , Epinefrina/farmacología , Hemodinámica/efectos de los fármacos , Lidocaína/farmacología , Microcirculación/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Adulto , Estudios Cruzados , Método Doble Ciego , Quimioterapia Combinada , Hemodinámica/fisiología , Humanos , Masculino , Microcirculación/fisiología , Persona de Mediana Edad , Estudios Prospectivos , Valores de Referencia , Ultrasonografía Intervencional/métodos , Adulto Joven
12.
Psychoneuroendocrinology ; 105: 44-50, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30318393

RESUMEN

BACKGROUND: Stress-induced prothrombotic changes are mediated by the sympathetic nervous system and critically involved in mental triggering of acute coronary syndromes, but the underlying psychobiology is not fully understood. We tested the hypothesis that a norepinephrine (NE) infusion to mimic effects of stress-induced NE release on blood coagulation elicits prothrombotic changes and examined to what extent these would be mediated by an alpha-adrenergic mechanism. METHODS AND RESULTS: In a single-blind placebo-controlled within-subjects design, 24 middle-aged, non-smoking, non-obese and normotensive men participated in three experimental trials with an interval between one and two weeks. Each trial applied two sequential infusions of 1 and 15 min duration with varying substances [i.e., saline as placebo, the non-specific α-blocker phentolamine (2.5 mg/min), and NE (5 µg/min)]: trial 1=saline + saline; trial 2=saline + NE, and trial 3=phentolamine + NE. Plasma levels of clotting factor VIII activity (FVIII:C), fibrinogen, and D-dimer were assessed from blood samples collected immediately before and 1 min and 20 min after infusion procedures. Compared to saline + saline, saline + NE induced increases over time in FVIII:C, fibrinogen, and D-dimer levels. With phentolamine + NE, fibrinogen levels remained increased compared to saline + saline, but changes in FVIII:C and D-dimer levels were no more different. Coagulation changes did not differ between saline + NE and phentolamine + NE. CONCLUSIONS: NE infusion activates blood coagulation. The resulting prothrombotic state could be one psychobiological mechanism underlying mental triggering of acute coronary syndromes. Blockade of α-adrenergic receptors partly attenuated NE effects on coagulation and could be implied to have preventive potential in susceptible individuals.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Factores de Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Norepinefrina/sangre , Norepinefrina/farmacología , Fentolamina/farmacología , Estrés Psicológico/sangre , Agonistas alfa-Adrenérgicos/administración & dosificación , Antagonistas Adrenérgicos alfa/administración & dosificación , Adulto , Epinefrina/sangre , Factor VIII/efectos de los fármacos , Productos de Degradación de Fibrina-Fibrinógeno/efectos de los fármacos , Fibrinógeno/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Norepinefrina/administración & dosificación , Fentolamina/administración & dosificación , Método Simple Ciego
13.
Int J Mol Sci ; 19(12)2018 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-30469522

RESUMEN

Primary adipose tissue-derived multipotent stem/stromal cells (adMSCs) demonstrate unusual signaling regulatory mechanisms, i.e., increased of sensitivity to catecholamines in response to noradrenaline. This phenomenon is called "heterologous sensitization", and was previously found only in embryonic cells. Since further elucidation of the molecular mechanisms that are responsible for such sensitization in primary adMSCs was difficult due to the high heterogeneity in adrenergic receptor expression, we employed immortalized adipose-derived mesenchymal stem cell lines (hTERT-MSCs). Using flow cytometry and immunofluorescence microscopy, we demonstrated that the proportion of cells expressing adrenergic receptor isoforms does not differ significantly in hTERT-MSCs cells compared to the primary adMSCs culture. However, using analysis of Ca2+-mobilization in single cells, we found that these cells did not demonstrate the sensitization seen in primary adMSCs. Consistently, these cells did not activate cAMP synthesis in response to noradrenaline. These data indicate that immortalized adipose-derived mesenchymal stem cell lines demonstrated impaired ability to respond to noradrenaline compared to primary adMSCs. These data draw attention to the usage of immortalized cells for MSCs-based regenerative medicine, especially in the field of pharmacology.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Norepinefrina/farmacología , Tejido Adiposo/citología , Señalización del Calcio , Línea Celular , Células Cultivadas , AMP Cíclico/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo
14.
Endocrinology ; 159(10): 3565-3578, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30124804

RESUMEN

Intrauterine growth restriction (IUGR) increases the risk of developing diabetes in later life, which indicates developmental programming of islets. IUGR fetuses with placental insufficiency develop hypoxemia, elevating epinephrine and norepinephrine (NE) concentrations throughout late gestation. To isolate the programming effects of chronically elevated catecholamines, NE was continuously infused into normally grown sheep fetuses for 7 days. High plasma NE concentrations suppress insulin, but after the NE infusion was terminated, persistent hypersecretion of insulin occurred. Our objective was to identify differential gene expression with RNA sequencing (RNAseq) in fetal islets after chronic adrenergic stimulation. After determining the NE-regulated genes, we identified the subset of differentially expressed genes that were common to both islets from NE fetuses and fetuses with IUGR to delineate the adrenergic-induced transcriptional responses. A portion of these genes were investigated in mouse insulinoma (Min6) cells chronically treated with epinephrine to better approximate the ß-cell response. In islets from NE fetuses, RNAseq identified 321 differentially expressed genes that were overenriched for metabolic and hormone processes, and the subset of 96 differentially expressed genes common to IUGR islets were overenriched for protein digestion, vitamin metabolism, and cell replication pathways. Thirty-eight of the 96 NE-regulated IUGR genes changed similarly between models with functional enrichment for proliferation. In Min6 cells, chronic epinephrine stimulation slowed proliferation and augmented insulin secretion after treatment. These data establish molecular mechanisms underlying persistent adrenergic stimulation in hyperfunctional fetal islets and identify a subset of genes dysregulated by catecholamines in IUGR islets that may represent programming of ß-cell proliferation capacity.


Asunto(s)
Feto/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Norepinefrina/farmacología , Agonistas alfa-Adrenérgicos/administración & dosificación , Agonistas alfa-Adrenérgicos/farmacología , Animales , Línea Celular Tumoral , Femenino , Retardo del Crecimiento Fetal/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/embriología , Ratones , Norepinefrina/administración & dosificación , Embarazo , Ovinos
15.
Psychopharmacology (Berl) ; 235(10): 3055-3063, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30112578

RESUMEN

Hypoglycemia can alter arousal and negatively impact mood. This study tests the hypothesis that acute drops in glucose metabolism cause an aversive state mediated by monoamine activity. In experiment 1, male Sprague-Dawley rats were either food deprived (FD) or pre-fed (PF) and tested on conditioned place avoidance (CPA; biased place conditioning design; 3 pairings drug/vehicle, each 30 min-long) induced by the glucose antimetabolite 2-deoxy-D-glucose (2-DG; 0, 300 or 500 mg/kg, SC). Locomotion and blood glucose were also assessed. Experiment 2 examined whether clonidine (noradrenergic α2 agonist, 0, 10 or 40 µg/kg, SC) or bupropion (monoamine reuptake blocker, 0, 10 or 30 mg/kg, SC) could alter CPA induced by 500 mg/kg 2-DG. In experiment 3, blood corticosterone (CORT) was measured in response to 500 mg/kg 2-DG, alone or in combination with 40 µg/kg clonidine or 30 mg/kg bupropion. Finally, experiment 4 controlled for possible place conditioning induced by 10 or 40 µg/kg clonidine, or 10 or 30 mg/kg bupropion injected without 2-DG. It was found that 2-DG increased blood glucose and produced a robust CPA. The feeding status of the animals modulated these effects, including CORT levels. Both clonidine and bupropion attenuated the effects of 2-DG on CPA and CORT, but only bupropion reversed suppression of locomotion. Taken together, these results in rats suggest that impaired glucose metabolism can negatively impact arousal and mood via effects on HPA and monoamine systems.


Asunto(s)
Conducta Animal/fisiología , Glucemia/metabolismo , Desoxiglucosa/fisiología , Hipoglucemia/psicología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Análisis de Varianza , Animales , Bupropión/farmacología , Clonidina/farmacología , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Inhibidores de Captación de Dopamina/farmacología , Hipoglucemia/metabolismo , Locomoción/fisiología , Masculino , Norepinefrina/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Nat Commun ; 9(1): 2067, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802242

RESUMEN

Testosterone deficiency in men is associated with increased risk for autoimmunity and increased B cell numbers through unknown mechanisms. Here we show that testosterone regulates the cytokine BAFF, an essential survival factor for B cells. Male mice lacking the androgen receptor have increased splenic B cell numbers, serum BAFF levels and splenic Baff mRNA. Testosterone deficiency by castration causes expansion of BAFF-producing fibroblastic reticular cells (FRCs) in spleen, which may be coupled to lower splenic noradrenaline levels in castrated males, as an α-adrenergic agonist decreases splenic FRC number in vitro. Antibody-mediated blockade of the BAFF receptor or treatment with the neurotoxin 6-hydroxydopamine revert the increased splenic B cell numbers induced by castration. Among healthy men, serum BAFF levels are higher in men with low testosterone. Our study uncovers a previously unrecognized regulation of BAFF by testosterone and raises important questions about BAFF in testosterone-mediated protection against autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Testosterona/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Animales , Enfermedades Autoinmunes/inmunología , Factor Activador de Células B/sangre , Receptor del Factor Activador de Células B/antagonistas & inhibidores , Receptor del Factor Activador de Células B/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Castración , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Norepinefrina/metabolismo , Oxidopamina/farmacología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Testosterona/sangre , Testosterona/deficiencia , Testosterona/inmunología
17.
Exp Dermatol ; 27(7): 763-768, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29637626

RESUMEN

When applied topically, oxymetazoline and brimonidine reduce the persistent facial erythema of rosacea; this effect is mediated by cutaneous vasoconstriction induced by postsynaptic activation of α-adrenoceptors. We investigated the α-adrenergic pharmacology of oxymetazoline and brimonidine. Functional activity on α-adrenoceptors was evaluated in vitro in HEK293 cells stably expressing single receptor subtypes using a fluorometric imaging plate reader Ca2+ influx assay. Oxymetazoline was an α1 -adrenoceptor agonist with partial α2 -adrenoceptor activity, whereas brimonidine was a highly selective full α2 -adrenoceptor agonist. In vivo pharmacology was investigated in a mouse model of ultraviolet B light (UVB)-induced skin erythema. To selectively inhibit α-adrenoceptor subtypes, mice were injected with prazosin (an α1 -selective antagonist) or rauwolscine (an α2 -selective antagonist) following UVB exposure. Oxymetazoline cream 1.0%, brimonidine gel 0.33% or vehicle control was applied topically, and erythema was measured using a chromameter. Oxymetazoline and brimonidine reduced UVB-induced erythema compared with vehicle control (P < .01). The effect of oxymetazoline was impaired in prazosin-pretreated but not rauwolscine-pretreated mice. Conversely, the effect of brimonidine was impaired in rauwolscine-pretreated but not prazosin-pretreated mice. These data suggest that while oxymetazoline and brimonidine produce cutaneous vasoconstriction, they do so through different α-adrenergic mechanisms, with oxymetazoline primarily acting via α1 -adrenoceptors and brimonidine acting via α2 -adrenoceptors.


Asunto(s)
Tartrato de Brimonidina/farmacología , Eritema/tratamiento farmacológico , Eritema/etiología , Oximetazolina/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Eritema/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Pelados , Prazosina/farmacología , Rosácea/tratamiento farmacológico , Rosácea/etiología , Rosácea/metabolismo , Rayos Ultravioleta/efectos adversos , Vasoconstricción/efectos de los fármacos , Yohimbina/farmacología
18.
Neurourol Urodyn ; 37(7): 2128-2134, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29675856

RESUMEN

AIM: To examine putative interaction between adrenergic and muscarinic contractile activation in the human urinary outflow tract. METHODS: Tissue from the trigone and prostatic urethra was obtained from 12 cystectomy and 16 prostatectomy specimen. Contractions were elicited by exposure to exogenous agonists before and after inhibition of Rho kinase and protein kinase c (PKC). Immunofluorescence and Western-blot studies were performed using antibodies to muscarinic M3-receptors (M3-R) and alpha1A-adrenoreceptors (alpha1A-AR). The study is registered with ClinicalTrials.gov, number NCT01227447. RESULTS: There was strong co-localization of M3-R and alpha1A-AR on trigonal and urethral myocytes. Western blot analysis revealed a significantly higher expression of alpha1A-AR in the superficial than in the deep trigone. Phenylephrine (PE, 1 µm) augmented contractions induced by carbachol (CA, 3 µm) to more than threefold control in the male superficial trigone, and to about sevenfold control in the proximal urethra. No such potentiation could be detected in female bladder outlet. Both PKC inhibitor GF 109203X and Rho kinase inhibitor Y-27632 reduced responses to 1 µM PE as well as to 3 µM CA significantly. However, the synergistic effect of the combined intervention remained proportionally unaffected. CONCLUSIONS: Muscarinic and adrenergic receptor activation exerts a strong synergistic effect in the male human bladder trigone and proximal urethra.


Asunto(s)
Receptor Muscarínico M3/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Sistema Urinario/inervación , Agonistas alfa-Adrenérgicos/farmacología , Anciano , Anciano de 80 o más Años , Carbacol/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Agonistas Muscarínicos/farmacología , Contracción Muscular/efectos de los fármacos , Fenilefrina/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Receptor Muscarínico M3/efectos de los fármacos , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Caracteres Sexuales , Uretra/efectos de los fármacos , Uretra/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores
19.
J Smooth Muscle Res ; 54(0): 1-12, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29540622

RESUMEN

Object We identified the ß-adrenoceptor (ß-AR) subtypes responsible for the relaxant responses to adrenaline (AD) and noradrenaline (NA) in the rat thoracic aorta and examined the role of cAMP which is involved in these relaxant responses. Methods The effects of ß-AR antagonists or the adenylyl cyclase inhibitor SQ 22,536 on AD- and NA-induced relaxant responses in phenylephrine-induced contraction and increases in cAMP levels were examined in isolated, endothelium-denuded rat thoracic aorta segments. Results AD-induced relaxation was completely suppressed by propranolol (10-7 M) or by ICI-118,551 (10-8 M) plus atenolol (10-6 M), and was also very strongly inhibited by ICI-118,551 (10-8 M) alone. AD (10-5 M) increased tissue cAMP levels by approximately 1.9-fold compared with that in non-stimulated aortic tissue, but did not significantly increase cAMP levels in the presence of ICI-118,551 (10-8 M) or SQ 22,536 (10-4 M). AD-induced relaxation was strongly suppressed by SQ 22,536 (10-4 M). NA-induced relaxation was almost completely suppressed by atenolol (10-6 M) plus ICI-118,551 (10-8 M) although it was hardly affected by ICI-118,551 (10-8 M) alone. NA (10-5 M) increased tissue cAMP levels by approximately 2.2-fold compared with that in non-stimulated aortic tissue, but did not significantly increase cAMP levels in the presence of atenolol (10-6 M) or SQ 22,536 (10-4 M). NA-induced relaxation was strongly suppressed by SQ 22,536 (10-4 M). Conclusion In rat thoracic aorta, AD- and NA-induced relaxations, which are both strongly dependent on increased tissue cAMP levels, are mainly mediated through ß2- and ß1-adrenoceptors respectively.


Asunto(s)
Aorta Torácica/fisiología , AMP Cíclico/metabolismo , Epinefrina/farmacología , Relajación Muscular/fisiología , Norepinefrina/farmacología , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Masculino , Relajación Muscular/efectos de los fármacos , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química , Transducción de Señal
20.
Neuropsychopharmacology ; 43(10): 2118-2125, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29472646

RESUMEN

Medications that target the noradrenergic system are important therapeutics for depression and anxiety disorders. More recently, clinical studies have shown that the α2-noradrenergic receptor (α2AR) agonist guanfacine can decrease stress-induced smoking relapse during acute abstinence, suggesting that targeting the noradrenergic system may aid in smoking cessation through effects on stress pathways in the brain. Acetylcholine (ACh), like the nicotine in tobacco, acts at nicotinic acetylcholine receptors (nAChRs) to regulate behaviors related to anxiety and depression. We therefore investigated interactions between guanfacine and ACh signaling in tests of anxiolytic and antidepressant efficacy in female and male C57BL/6J mice, focusing on the amygdala as a potential site of noradrenergic/cholinergic interaction. The antidepressant-like effects of guanfacine were blocked by shRNA-mediated knockdown of α2AR in amygdala. Knockdown of the high-affinity ß2 nAChR subunit in amygdala also prevented antidepressant-like effects of guanfacine, suggesting that these behavioral effects require ACh signaling through ß2-containing nAChRs in this brain area. Ablation of NE terminals prevented the anxiolytic- and antidepressant-like effects of the nicotinic partial agonist cytisine, whereas administration of the cholinesterase antagonist physostigmine induced a depression-like phenotype that was not altered by knocking down α2AR in the amygdala. These studies suggest that ACh and NE have opposing actions on behaviors related to anxiety and depression and that cholinergic signaling through ß2-containing nAChRs and noradrenergic signaling through α2a receptors in neurons of the amygdala are critical for regulation of these behaviors.


Asunto(s)
Amígdala del Cerebelo/fisiología , Ansiedad/psicología , Depresión/psicología , Sistema Nervioso Parasimpático/fisiología , Transducción de Señal/fisiología , Sistema Nervioso Simpático/fisiología , Acetilcolina/agonistas , Acetilcolina/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Alcaloides/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Azocinas/farmacología , Inhibidores de la Colinesterasa/farmacología , Femenino , Técnicas de Silenciamiento del Gen , Guanfacina/antagonistas & inhibidores , Guanfacina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Agonistas Nicotínicos/farmacología , Norepinefrina/fisiología , Sistema Nervioso Parasimpático/efectos de los fármacos , Quinolizinas/farmacología , Receptores Adrenérgicos alfa 2/genética , Transducción de Señal/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA