Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 474
Filtrar
1.
Fish Shellfish Immunol ; 92: 188-195, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31176766

RESUMO

Octopamine (OA), a biogenic monoamine, is known to mediate several immune responses. This study analyzed the effects of OA on immunological regulation in the tiger shrimp Penaeus monodon. The immune parameters including total haemocyte count, differential haemocyte count, phenoloxidase activity, respiratory bursts, superoxide dismutase activity, and phagocytic activity and clearance efficiency in response to the pathogen, Photobacterium damselae, were determined when shrimp were individually injected with saline or OA at 100 or 1000 pmol shrimp-1. In addition, the intracellular second messengers in haemocyte such as Ca2+ and adenosine 3',5'-cyclic monophosphate (cAMP) were examined in shrimp receiving saline or OA at 1 or 10 nmol shrimp-1. Results showed that all of the immune parameters significantly increased at 2-4 h in OA-injected shrimp except hyaline cells in 100 pmol shrimp-1-injected shrimp at 4 h, but phenoloxidase activity per granulocyte significantly decreased at 2-4 h. However, these had returned to saline control levels after receiving OA for 8 h except differential haemocyte count and phenoloxidase activity per granulocyte for 16 h. An injection of OA also significantly increased the survival rate of shrimp challenged with Pho. damselae. Shrimp receiving OA at 1 and 10 nmol shrimp-1 significantly increased the intracellular Ca2+ concentration ([Ca2+]i) at 30-60 min and 30 min, and cAMP concentration [cAMP]i) at 5-15 min and 15 min, respectively. However, [Ca2+]i at 50-60 min, and [cAMP]i at 30-60 min returned to saline control when the shrimp received OA at 10 nmol shrimp-1, and at 1 and 10 nmol shrimp-1, respectively. These results suggest that OA administration by injection at ≤1000 pmol shrimp-1 mediates transient upregulation of immunity together with the increased resistance of P. monodon to Pho. damselae, which are modulated through intracellular Ca2+ and cAMP second messenger pathways.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Inata/efeitos dos fármacos , Octopamina/metabolismo , Penaeidae/genética , Penaeidae/imunologia , Transdução de Sinais/imunologia , Adjuvantes Imunológicos/farmacologia , Agonistas alfa-Adrenérgicos/administração & dosagem , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Cálcio/metabolismo , AMP Cíclico/metabolismo , Perfilação da Expressão Gênica , Octopamina/administração & dosagem , Photobacterium/fisiologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/imunologia
2.
EMBO J ; 32(11): 1613-25, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23591430

RESUMO

The sympathetic nervous system relies on distinct populations of neurons that use noradrenaline or acetylcholine as neurotransmitter. We show that fating of the sympathetic lineage at early stages results in hybrid precursors from which, genetic cell-lineage tracing reveals, all types progressively emerge by principal mechanisms of maintenance, repression and induction of phenotypes. The homeobox transcription factor HMX1 represses Tlx3 and Ret, induces TrkA and maintains tyrosine hydroxylase (Th) expression in precursors, thus driving segregation of the noradrenergic sympathetic fate. Cholinergic sympathetic neurons develop through cross-regulatory interactions between TRKC and RET in precursors, which lead to Hmx1 repression and sustained Tlx3 expression, thereby resulting in failure of TrkA induction and loss of maintenance of Th expression. Our results provide direct evidence for a model in which diversification of noradrenergic and cholinergic sympathetic neurons is based on a principle of cross-repressive functions in which the specific cell fates are directed by an active suppression of the expression of transcription factors and receptors that direct the alternative fate.


Assuntos
Diferenciação Celular , Neurônios Colinérgicos/citologia , Proteínas de Homeodomínio/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Fatores de Transcrição/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Acetilcolina/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Agonistas Colinérgicos/metabolismo , Neurônios Colinérgicos/fisiologia , Cromossomos Artificiais Bacterianos , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Norepinefrina/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptor trkC/genética , Receptor trkC/metabolismo , Receptores de Fatores de Crescimento/genética , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia , Fatores de Transcrição/genética , Tirosina 3-Mono-Oxigenase/metabolismo
3.
Exp Physiol ; 102(9): 1245-1258, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28681979

RESUMO

NEW FINDINGS: What is the central question of this study? We sought to determine whether human skeletal muscle feed arteries (SFMAs) express TRPV1 channels and what role they play in modulating vascular function. What is the main finding and its importance? Human SMFAs do express functional TRPV1 channels that modulate vascular function, specifically opposing α-adrenergic receptor-mediated vasocontraction and potentiating vasorelaxation, in an endothelium-dependent manner, as evidenced by the α1 -receptor-mediated responses. Thus, the vasodilatory role of TRPV1 channels, and their ligand capsaicin, could be a potential therapeutic target for improving vascular function. Additionally, given the 'sympatholytic' effect of TRPV1 activation and known endogenous activators (anandamide, reactive oxygen species, H+ , etc.), TRPV1 channels might contribute to functional sympatholysis during exercise. To examine the role of the transient receptor potential vanilloid type 1 (TRPV1 ) ion channel in the vascular function of human skeletal muscle feed arteries (SMFAs) and whether activation of this heat-sensitive receptor could be involved in modulating vascular function, SMFAs from 16 humans (63 ± 5 years old, range 41-89 years) were studied using wire myography with capsaicin (TRPV1 agonist) and without (control). Specifically, phenylephrine (α1 -adrenergic receptor agonist), dexmedetomidine (α2 -adrenergic receptor agonist), ACh and sodium nitroprusside concentration-response curves were established to assess the role of TRPV1 channels in α-receptor-mediated vasocontraction as well as endothelium-dependent and -independent vasorelaxation, respectively. Compared with control conditions, capsaicin significantly attenuated maximal vasocontraction in response to phenylephrine [control, 52 ± 8% length-tensionmax (LTmax ) and capsaicin, 21 ± 5%LTmax ] and dexmedetomidine (control, 29 ± 12%LTmax and capsaicin, 2 ± 3%LTmax ), while robustly enhancing maximal vasorelaxation with ACh (control, 78 ± 8% vasorelaxation and capsaicin, 108 ± 13% vasorelaxation) and less clearly enhancing the sodium nitroprusside response. Denudation of the endothelium greatly attenuated the maximal ACh-induced vasorelaxation equally in the control and capsaicin conditions (∼17% vasorelaxation) and abolished the attenuating effect of capsaicin on the maximal phenylephrine response (denuded + capsaicin, 61 ± 20%LTmax ). Immunohistochemistry identified a relatively high density of TRPV1 channels in the endothelium compared with the smooth muscle of the SMFAs, but because of the far greater volume of smooth muscle, total TRPV1 protein content was not significantly attenuated by denudation. Thus, SMFAs ubiquitously express functional TRPV1 channels, which alter vascular function, in terms of α1 -receptors, in a predominantly endothelium-dependent manner, conceivably contributing to the functional sympatholysis and unveiling a therapeutic target.


Assuntos
Artérias/metabolismo , Músculo Esquelético/metabolismo , Músculo Liso Vascular/metabolismo , Canais de Cátion TRPV/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Artérias/efeitos dos fármacos , Capsaicina/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Músculo Liso Vascular/irrigação sanguínea , Músculo Liso Vascular/efeitos dos fármacos , Nitroprussiato/farmacologia , Fenilefrina/farmacologia , Receptores Adrenérgicos alfa/metabolismo , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
4.
J Exp Biol ; 220(Pt 3): 336-340, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27875262

RESUMO

Most vertebrates increase cardiac output during activity by elevating heart rate with relatively stable stroke volume. However, several studies have demonstrated 'intrinsic autoregulation' of cardiac output where artificially increased heart rate is associated with decreased stroke volume, leaving cardiac output unchanged. We explored the capacity of noradrenaline to overcome autoregulation in the anaconda heart. Electrically pacing in situ perfused hearts from the intrinsic heart rate to the maximum attainable resulted in a proportional decrease in stroke volume. However, noradrenaline, which increased heart rate to the same frequency as pacing, maintained stroke volume and thus increased cardiac output. In atrial and ventricular preparations, noradrenaline significantly increased the force of contraction and contraction kinetics. Thus, the increased contractility associated with adrenergic stimulation ameliorates filling limitations at high heart rates. Although heart rate appears the primary regulated variable during activity, this may only be achieved with compensatory amendments in myocardial contractility provided by adrenergic stimulation.


Assuntos
Boidae/fisiologia , Coração/fisiologia , Norepinefrina/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Débito Cardíaco , Homeostase , Masculino , Contração Miocárdica , Volume Sistólico
5.
Proc Natl Acad Sci U S A ; 109(12): 4562-7, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393023

RESUMO

Female sperm storage is common among organisms with internal fertilization. It is important for extended fertility and, in cases of multiple mating, for sperm competition. The physiological mechanisms by which females store and manage stored sperm are poorly understood. Here, we report that the biogenic amines tyramine (TA) and octopamine (OA) in Drosophila melanogaster females play essential roles in sperm storage. D. melanogaster females store sperm in two types of organs, a single seminal receptacle and a pair of spermathecae. We examined sperm storage parameters in females mutant in enzymes required for the biochemical synthesis of tyrosine to TA and TA to OA, respectively. Postmating uterine conformational changes, which are associated with sperm entry and accumulation into storage, were unaffected by the absence of either TA or OA. However, sperm release from storage requires both TA and OA; sperm were retained in storage in both types of mutant females at significantly higher levels than in control flies. Absence of OA inhibited sperm depletion only from the seminal receptacle, whereas absence of both OA and TA perturbed sperm depletion from both storage organ types. We find innervation of the seminal receptacle and spermathecae by octopaminergic-tyraminergic neurons. These findings identify a distinct role for TA and OA in reproduction, regulating the release of sperm from storage, and suggest a mechanism by which Drosophila females actively regulate the release of stored sperm.


Assuntos
Neurotransmissores/metabolismo , Octopamina/metabolismo , Espermatozoides/metabolismo , Tiramina/metabolismo , Inibidores da Captação Adrenérgica/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Cruzamentos Genéticos , Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Feminino , Fertilização , Masculino , Modelos Biológicos , Mutação , Neurônios/metabolismo , Reprodução
6.
J Biol Chem ; 288(21): 15291-302, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23548902

RESUMO

Microglia, the resident immune cells of the central nervous system (CNS), monitor the brain for disturbances of tissue homeostasis by constantly moving their fine processes. Microglia respond to tissue damage through activation of ATP/ADP receptors followed by directional process extension to the damaged area. A common feature of several neurodegenerative diseases is the loss of norepinephrine, which might contribute to the associated neuroinflammation. We carried out a high resolution analysis of the effects of norepinephrine (NE) on microglial process dynamics in acute brain slices from mice that exhibit microglia-specific enhanced green fluorescent protein expression. Bath application of NE to the slices resulted in significant process retraction in microglia. Analysis of adrenergic receptor expression with quantitative PCR indicated that resting microglia primarily express ß2 receptors but switch expression to α2A receptors under proinflammatory conditions modeled by LPS treatment. Despite the differential receptor expression, NE caused process retraction in both resting and LPS-activated microglia cultured in the gelatinous substrate Matrigel in vitro. The use of subtype-selective receptor agonists and antagonists confirmed the involvement of ß2 receptors in mediating microglial process dynamics in resting cells and α2A receptors in activated cells. Co-application of NE with ATP to resting microglia blocked the ATP-induced process extension and migration in isolated microglia, and ß2 receptor antagonists prolonged ATP effects in brain slice tissues, suggesting the presence of cross-talk between adrenergic and purinergic signaling in microglia. These data show that the neurotransmitter NE can modulate microglial motility, which could affect microglial functions in pathogenic situations of either elevated or reduced NE levels.


Assuntos
Trifosfato de Adenosina/farmacologia , Agonistas alfa-Adrenérgicos/farmacologia , Movimento Celular/efeitos dos fármacos , Microglia/metabolismo , Norepinefrina/farmacologia , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Movimento Celular/fisiologia , Células Cultivadas , Camundongos , Camundongos Transgênicos , Microglia/citologia , Norepinefrina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
7.
Biochem J ; 450(2): 351-63, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23215897

RESUMO

ERK1/2 (extracellular-signal-regulated kinase 1/2) and their substrates RSKs (p90 ribosomal S6 kinases) phosphorylate different transcription factors, contributing differentially to transcriptomic profiles. In cardiomyocytes ERK1/2 are required for >70% of the transcriptomic response to endothelin-1. In the present study we investigated the role of RSKs in the transcriptomic responses to the G(q)-protein-coupled receptor agonists endothelin-1, phenylephrine (a generic α(1)-adrenergic receptor agonist) and A61603 (α(1A)-adrenergic receptor selective). Phospho-ERK1/2 and phospho-RSKs appeared in cardiomyocyte nuclei within 2-3 min of stimulation (endothelin-1>A61603≈phenylephrine). All agonists increased nuclear RSK2, but only endothelin-1 increased the nuclear RSK1 content. PD184352 (inhibits ERK1/2 activation) and BI-D1870 (inhibits RSKs) were used to dissect the contribution of RSKs to the endothelin-1-responsive transcriptome. Of the 213 RNAs up-regulated after 1 h, 51% required RSKs for their up-regulation, whereas 29% required ERK1/2 but not RSKs. The transcriptomic response to phenylephrine overlapped with, but was not identical with, endothelin-1. As with endothelin-1, PD184352 inhibited the up-regulation of most phenylephrine-responsive transcripts, but the greater variation in the effects of BI-D1870 suggests that differential RSK signalling influences global gene expression. A61603 induced similar changes in RNA expression in cardiomyocytes as phenylephrine, indicating that the signal was mediated largely through α(1A)-adrenergic receptors. A61603 also increased expression of immediate early genes in perfused adult rat hearts and, as in cardiomyocytes, up-regulation of the majority of genes was inhibited by PD184352. PD184352 or BI-D1870 prevented the increased surface area induced by endothelin-1 in cardiomyocytes. Thus RSKs play a significant role in regulating cardiomyocyte gene expression and hypertrophy in response to G(q)-protein-coupled receptor stimulation.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Regulação da Expressão Gênica , Miócitos Cardíacos/metabolismo , Receptor de Endotelina A/agonistas , Receptores Adrenérgicos alfa 1/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Benzamidas/farmacologia , Núcleo Celular/metabolismo , Masculino , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais
8.
J Immunol ; 185(10): 5762-8, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20935206

RESUMO

Although noradrenaline (NA), a stress-associated neurotransmitter, seems to affect the immune system, the precise mechanisms underlying NA-mediated immunoregulation are not fully understood. We examined the effect of NA on Ag uptake (endocytosis) by dendritic cells (DCs) using murine bone marrow-derived DCs and fluorescence-labeled endocytic tracers (dextran and OVA). Ag uptake by DCs notably increased following a very brief treatment (3 min) with NA. NA-induced endocytosis was completely blocked by treatment with α(2)-adrenoceptor antagonist yohimbine. Neither α(1)-adrenoceptor antagonist prazosin nor ß-adrenoceptor antagonist propranolol affected NA-induced endocytosis by DCs. A selective α(2)-adrenoceptor agonist, azepexole (B-HT 933), also significantly increased endocytosis by DCs. Thus, the α(2)-adrenoceptor seems to be responsible for NA-induced DC endocytosis. In parallel, NA markedly activated intracellular signaling pathways of PI3K and ERK1/2 in DCs. NA-mediated activation of these pathways was completely inhibited by yohimbine treatment. Blocking PI3K activation significantly reduced NA-induced endocytosis by DCs. Based on these results, NA rapidly enhances Ag capture by DCs via α(2) adrenoceptor-mediated PI3K activation, which may be associated with immune enhancement following acute stress.


Assuntos
Células Dendríticas/metabolismo , Ativação Enzimática/imunologia , Norepinefrina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Apresentação de Antígeno/imunologia , Western Blotting , Separação Celular , Células Dendríticas/imunologia , Endocitose/imunologia , Citometria de Fluxo , Camundongos , Fosfatidilinositol 3-Quinases/imunologia , Receptores Adrenérgicos alfa 2/imunologia
9.
Methods Mol Biol ; 2550: 95-100, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36180681

RESUMO

Pineal gland secretes the hormone melatonin at night with a circadian rhythm. The synthesis and secretion of melatonin are stimulated at night by norepinephrine released by sympathetic postganglionic neurons projecting from the superior cervical ganglia. Norepinephrine simultaneously activates α- and ß-adrenoceptors, triggering melatonin synthesis.To study the regulation of melatonin production and secretion, it is very convenient to use an ex vivo preparation. Thus, it is possible to keep intact pineal glands in culture and to study the actions of agonists, antagonists, modulators, toxic agents, etc., in melatonin synthesis. Artificial melatonin synthesis stimulation in vitro is usually achieved by using a ß-adrenergic agonist alone or in association with an α-adrenergic agonist. In this chapter, the methodology of cultured pineal glands will be described. Several papers were published by our group using this methodology, approaching the role played in melatonin synthesis control by angiotensin II and IV, insulin, glutamate, voltage-gated calcium channels, anhydroecgonine methyl ester (AEME, crack-cocaine product), monosodium glutamate (MSG), signaling pathways like NFkB, pathophysiological conditions like diabetes, etc.


Assuntos
Cocaína , Insulinas , Melatonina , Glândula Pineal , Agonistas alfa-Adrenérgicos/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Angiotensina II/metabolismo , Canais de Cálcio/metabolismo , Ritmo Circadiano/fisiologia , Melatonina/metabolismo , Norepinefrina , Glândula Pineal/metabolismo , Receptores Adrenérgicos beta/metabolismo , Glutamato de Sódio
10.
J Mol Cell Cardiol ; 51(1): 16-23, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21458461

RESUMO

Activation of coronary vascular α-adrenoceptors results in vasoconstriction which competes with metabolic vasodilation during sympathetic activation. Epicardial conduit vessel constriction is largely mediated by α(1)-adrenoceptors; the constriction of the resistive microcirculation largely by α(2)-adrenoceptors, but also by α(1)-adrenoceptors. There is no firm evidence that α-adrenergic coronary vasoconstriction exerts a beneficial effect on transmural blood flow distribution. In fact, α-blockade in anesthetized and conscious dogs improves blood flow to all transmural layers, during normoperfusion and hypoperfusion. Also, in patients with coronary artery disease, blockade of α(1)- and α(2)-adrenoceptors improves coronary blood flow, myocardial function and metabolism.


Assuntos
Circulação Coronária/fisiologia , Receptores Adrenérgicos alfa/metabolismo , Vasoconstrição/fisiologia , Agonistas alfa-Adrenérgicos/metabolismo , Antagonistas Adrenérgicos alfa/metabolismo , Animais , Doença da Artéria Coronariana , Doença das Coronárias/fisiopatologia , Vasos Coronários/metabolismo , Cães , Coração/fisiopatologia , Humanos , Isquemia , Vasodilatação/fisiologia
11.
Pharmacology ; 88(1-2): 55-64, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21791946

RESUMO

AIMS: We investigated how modification of levels of the sex hormones 17ß-estradiol and testosterone affects vascular contraction and nongenomic vascular effects of 17ß-estradiol. METHODS: Male and female rats were treated with vehicle, 17ß-estradiol (25 µg/kg/day) or testosterone (1 mg/kg/day) for 14 consecutive days after sham gonadectomy or gonadectomy was performed. Isometric tensions were then measured from mesenteric arteries of each group of rats. RESULTS: Contraction to phenylephrine was increased in mesenteric arteries from rats with or without gonadectomy treated with testosterone for 14 days compared to their intact controls. Contraction to phenylephrine was reduced in mesenteric arteries of rats with or without gonadectomy treated with 17ß-estradiol for 14 days compared to their intact controls. Incubation of mesenteric arteries with 17ß-estradiol (1 nmol/l) for 30 min reduced contraction to phenylephrine in mesenteric arteries of rats that were treated with testosterone for 14 days. This acute incubation of 17ß-estradiol had no effect on arteries from rats that were treated with 17ß-estradiol for 14 days. The acute effect of 17ß-estradiol (1 nmol/l) is preserved in arteries without endothelium. CONCLUSION: Our results suggest that 14 days' testosterone treatment enhances while 14 days' 17ß-estradiol treatment suppresses contraction as well as the nongenomic effects of 17ß-estradiol in the vascular smooth muscles.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Hormônios Esteroides Gonadais/sangue , Artérias Mesentéricas/fisiologia , Agonistas alfa-Adrenérgicos/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Peso Corporal , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Endotélio Vascular/fisiologia , Estradiol/sangue , Estrogênios/sangue , Feminino , Hormônios Esteroides Gonadais/farmacologia , Humanos , Masculino , Músculo Liso Vascular/fisiologia , Fenilefrina/metabolismo , Fenilefrina/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Testosterona/sangue , Testosterona/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
12.
Biochim Biophys Acta ; 1793(5): 871-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19250949

RESUMO

Muscle fructose 1,6-bisphosphatase (FBPase), a well-known regulatory enzyme of glyconeogenic pathway has recently been found inside nuclei of several cell types (cardiomyocytes, smooth muscle cells, myogenic progenitor cells). This surprising finding raised a question concerning the role of FBPase in this compartment of the cell, and of the extracellular signals regulating nuclear transport of the enzyme. In the present paper we show that, in HL-1 cardiomyocyte cell line, the activity of adenylyl cyclase and cAMP-dependent protein kinase A is essential to nuclear import of FBPase. The import is also stimulated by isoproterenol (a nonselective beta-adrenergic receptors agonist) and inhibited by metoprolol (a selective beta1 antagonist), strongly suggesting that nucleo-cytoplasmic shuttling of FBPase is under the control of beta1-adrenergic receptor-dependent Gs protein signaling cascade.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Núcleo Celular/enzimologia , Frutose-Bifosfatase/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Miócitos Cardíacos , Receptores Adrenérgicos beta 1/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Adenilil Ciclases/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Agonistas Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/metabolismo , Animais , Linhagem Celular , Colforsina/metabolismo , Meios de Cultura/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Isoproterenol/metabolismo , Metoprolol/metabolismo , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Norepinefrina
13.
Int J Neuropsychopharmacol ; 13(9): 1219-31, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20128952

RESUMO

Stimulation of the subiculum/CA1 of the hippocampal formation evokes monosynaptic field potentials in the prefrontal cortex (PFC). High-frequency stimulation of the hippocampus (HPC) can induce long-term potentiation (LTP) in this hippocampo-prefrontal cortical (hippo-PFC) pathway. Previous studies have shown that dopamine and serotonin modulate hippo-PFC LTP. Here, we investigated whether the locus coeruleus (LC) and noradrenaline (NA) can modulate LTP in the rat hippo-PFC pathway. Stimulation of the LC in combination with stimulation of the HPC increased hippo-PFC LTP. Infusion of lidocaine into the LC reduced hippo-PFC LTP. Administration of the noradrenaline reuptake inhibitor, nisoxetine or the alpha2 adrenoceptor antagonist, idazoxan prior to high-frequency stimulation of the HPC enhanced hippo-LTP. In contrast, administration of clonidine, an alpha2 adrenoceptor agonist, impaired hippo-PFC LTP. Partial noradrenergic (NAergic) lesioning with DSP-4 also impaired hippo-PFC LTP. In conclusion, the LC and NAergic mechanisms modulate hippo-PFC LTP.


Assuntos
Hipocampo/efeitos dos fármacos , Locus Cerúleo/efeitos dos fármacos , Potenciação de Longa Duração , Norepinefrina/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas alfa-Adrenérgicos/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Benzilaminas/metabolismo , Benzilaminas/farmacologia , Clonidina/metabolismo , Clonidina/farmacologia , Dopamina/metabolismo , Dopamina/fisiologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Idazoxano/metabolismo , Idazoxano/farmacologia , Locus Cerúleo/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Córtex Pré-Frontal/fisiologia , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Serotonina/fisiologia
14.
J Mol Cell Cardiol ; 47(1): 149-55, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19362092

RESUMO

Spontaneously hypertensive rats (SHR) are known to have cardiac noradrenergic hyperactivity due to an impaired nitric oxide (NO)-cGMP pathway. We hypothesized that dietary l-arginine supplementation may correct this autonomic phenotype. Male SHR and Wistar Kyoto rats (WKY) aged 16-18 weeks were given l-arginine (10 g/L in drinking water) for 1 week. Separate control groups received no supplementation. The SHR control had a significantly lower plasma l-arginine than WKY control, but this was increased to a comparable level following l-arginine. Atrial cGMP was lower in the SHR control compared with the WKY control (2.4+/-0.4 pmol/mg vs 3.9+/-0.5 pmol/mg, p<0.05), but increased to 4.1+/-0.5 pmol/mg protein (n=8, p<0.05) with l-arginine. Evoked [(3)H]norepinephrine release in isolated spontaneously beating right atria from the SHR control (328+/-19%, n=19) was 28% higher than the WKY control (256+/-20%, n=14, p<0.05), but was reduced to 258+/-11% with l-arginine feeding (n=24, p<0.01). Soluble guanylyl cyclase (sGC) inhibition caused a greater increase of evoked norepinephrine release in the l-arginine fed SHR compared with the non-fed SHR. l-arginine feeding did not reduce evoked norepinephrine release in the WKY. In-vitro heart rate response to exogenous norepinephrine (0.1-5 mumol/L) was similar between l-arginine fed (n=13) and non-fed SHR (n=10), suggesting that l-arginine supplementation worked pre-synaptically. Myocardial tyrosine hydroxylase protein was decreased in SHR following l-arginine supplementation, providing a link to reduced synthesis of norepinephrine. In conclusion, l-arginine supplementation corrects local cardiac noradrenergic hyperactivity in the SHR, probably via increased pre-synaptic substrate availability of NOS-sGC-cGMP pathway and reduced tyrosine hydroxylase levels.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Arginina/farmacologia , Norepinefrina/farmacologia , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Arginina/sangue , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Guanilato Ciclase/antagonistas & inibidores , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Masculino , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Norepinefrina/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Guanilil Ciclase Solúvel
15.
Science ; 192(4241): 791-3, 1976 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-4894

RESUMO

A radioactively labeled alpha-adrenergic antagonist, [3H]dihydroergocryptine, binds specifically to a site on rabbit uterine membranes. Binding is rapid, reaching equilibrium in less than 17 minutes at 25 degrees C. Adrenergic agonists compete for this binding site with an order of affinities identical to the pharmacological potency order of these agents as alpha-adrenergic agonists (epinephrine greater than norepinephrine greater than isoprotereonl). The (-) stereoisomers of epinephrine and norepinephrine are 30 times more potent in competing for the site than the corresponding (+) stereoisomers. alpha-Adrenergic antagonists, such as phentolamine and phenoxybenzamine, potently compete for the binding sites while the beta-adrenergic antagonist propranolol does not. Structural analogs of catecholamines that are devoid of alpha-adrenergic physiological activity do not compete for [3H]dihydroergocryptine binding sites. These data suggest that alpha-adrenergic receptors can be directly identified and studied by [3H]dihydroergocryptine binding.


Assuntos
Agonistas alfa-Adrenérgicos/metabolismo , Antagonistas Adrenérgicos alfa/metabolismo , Mesilatos Ergoloides/metabolismo , Receptores Adrenérgicos , Animais , Ligação Competitiva , Dopamina/metabolismo , Feminino , Cinética , Propranolol/metabolismo , Coelhos , Estereoisomerismo , Útero/metabolismo
16.
Mol Cell Biochem ; 323(1-2): 61-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19039652

RESUMO

This study was designed to test whether increased sympathetic stimulation during polymicrobial sepsis modulates the profile of the syntaxin1A and norepinephrine transporter (NET) in the heart. Sepsis of mild and severe intensity was induced in male Sprague-Dawley rats (275-350 g) using the cecal inoculum (CI) and cecal ligation and puncture (CLP) methods, respectively. The heart samples were isolated from sham, 1, 3, and 7 day post-sepsis in the CI model and at 2 and 20 h post-sepsis in the CLP model. In the CI model, the plasma concentration of norepinephrine (NE) significantly increased at 1, 3, and 7 days post-CI compared to the sham group. The myocardial syntaxin1A mRNA and protein expression also significantly increased at 1 day post-CI compared to the sham group. However, the sepsis-induced increase in syntaxin1A returned to the baseline values at 3 and 7 days post-CI. The expressions of myocardial NET mRNA and protein were not altered at 1 day post-CI but significantly decreased at 3 days post-CI compared to the sham and 1 day post-CI groups. The immunohistochemical analyses revealed an increased localization of NET and syntaxin1A in the heart tissue sections of the 1 day post-CI group. In the CLP model of severe sepsis, the myocardial syntaxin1A mRNA protein expressions significantly increased at 2 h post-CLP, but remained unchanged at 20 h post-CLP compared to the sham group. In contrast, the myocardial expressions of NET mRNA and protein significantly decreased at both 2 and 20 h post-CLP compared to the sham group. It appears that during severe sepsis (CLP model), both the upregulation of syntaxin1A and the downregulation of NET contribute to increased concentrations of NE during the early and late stages of sepsis.


Assuntos
Miocárdio/metabolismo , Sepse , Sintaxina 1/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Masculino , Norepinefrina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Ratos , Ratos Sprague-Dawley , Sepse/metabolismo , Sepse/microbiologia , Sintaxina 1/genética , Regulação para Cima
17.
Brain Res ; 1707: 54-61, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30468724

RESUMO

Type I Diabetes (T1D) is associated with reduced leptin levels and increased stress axis activity marked by elevations in norepinephrine (NE) levels in the paraventricular nucleus (PVN) of the hypothalamus. We hypothesized that leptin suppresses stress axis activity in T1D through central and peripheral mechanisms. In the first experiment, adult male Sprague Dawley rats were implanted with a cannula in the PVN and randomly divided into a non-diabetic group treated with vehicle (n = 6) and a diabetic group treated with streptozotocin (n = 13). Food intake and water intake was measured for 14 days. On the last day, a subset of diabetic rats were treated with 500 µg of leptin i.p. Rats were subjected to push-pull perfusion of the PVN and hourly blood sampling for 5 h. In the next experiment, diabetic rats were treated either with an alpha-2 adrenergic agonist, clonidine (CLON), or a beta adrenergic agonist isoproterenol (ISO), to reverse the effects of leptin. Rats were subjected to push pull perfusion and hourly blood sampling. In experiment 1, T1D increased food intake, water intake, NE release in the PVN and circulating CS levels. Leptin treatment decreased NE release modestly but produced a robust reduction in corticosterone (CS) levels. In experiment 2, CLON but not ISO was able to reverse the effect of leptin on NE levels in the PVN, however, both agonists were capable of blocking leptin's effects on circulating CS. These results suggest that in diabetic rats, the sensitivity of the hypothalamus to beta adrenergic agonists is altered, while the adrenals remain sensitive to both alpha and beta adrenergic agonists.


Assuntos
Leptina/metabolismo , Norepinefrina/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Agonistas Adrenérgicos/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animais , Clonidina/farmacologia , Corticosterona/análise , Corticosterona/sangue , Diabetes Mellitus Experimental/fisiopatologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Isoproterenol/farmacologia , Leptina/fisiologia , Masculino , Norepinefrina/fisiologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
18.
J Cell Physiol ; 216(2): 486-97, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18314882

RESUMO

Hepatocytes express adrenergic receptors (ARs) that modulate several functions, including liver regeneration, hepatocyte proliferation, glycogenolysis, gluconeogenesis, synthesis of urea and fatty acid metabolism. Adrenergic hepatic function in adults is mainly under the control of alpha(1)-ARs; however, the mechanism through which they influence diverse processes remains incompletely understood. This study describes a novel alpha(1)-AR-mediated transactivation of signal transducer and activator of transcription-3 (Stat3) in primary and transformed hepatocytes. Treatment of primary rat hepatocytes with the alpha(1)-AR agonist, phenylephrine (PE), induced a rapid phosphorylation of Stat3. PE also increased Stat3 phosphorylation, DNA binding and transcription activity in transformed human hepatocellular carcinoma cells (Hep3B). The PE-induced Stat3 phosphorylation, DNA binding and reporter activity were completely blocked by the selective alpha(1)-AR antagonist, prazosin. In addition, transfection of Hep3B cells with human alpha(1B)-AR expression vector also enhanced Stat3 phosphorylation and reporter activity. Moreover, overexpression of RGS2, a protein inhibitor of G(q/11) signaling, blocked PE-induced Stat3 phosphorylation and reporter activity. The observations that PE induced the formation of c-Src-Stat3 binding complex and phosphorylation of epidermal growth factor receptor (EGFR) and that inhibiting Src and EGFR prevented PE-induced Stat3 activation indicate the involvement of Src and EGFR. Taken together, these observations demonstrate a novel alpha(1)-AR-mediated Stat3 activation that involves G(q/11), Src, and EGFR in hepatic cells.


Assuntos
Receptores ErbB/metabolismo , Hepatócitos/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Fator de Transcrição STAT3/metabolismo , Quinases da Família src/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Antagonistas Adrenérgicos alfa/metabolismo , Adulto , Animais , Linhagem Celular Tumoral , Células Cultivadas , Ativação Enzimática , Receptores ErbB/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hepatócitos/citologia , Humanos , Masculino , Fenilefrina/metabolismo , Prazosina , Proteínas RGS/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores Adrenérgicos alfa 1/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/fisiologia , Transcrição Gênica , Quinases da Família src/genética
19.
J Clin Invest ; 101(4): 812-8, 1998 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9466976

RESUMO

This study tested the hypothesis that nitric oxide (NO) and atrial natriuretic peptide (ANP) can attenuate the effects of adrenergic agonists on the growth of cardiac myocytes and fibroblasts. In ventricular cells cultured from neonatal rat heart, ANP and the NO donor S-nitroso-N-acetyl-D,L-penicillamine (SNAP) caused concentration-dependent decreases in the norepinephrine (NE)-stimulated incorporation of [3H]leucine in myocytes and [3H]thymidine in fibroblasts. In myocytes, the NO synthase inhibitor NG-monomethyl-L-arginine potentiated NE-stimulated [3H]leucine incorporation. In both cell types, ANP and SNAP increased intracellular cGMP levels, and their growth-suppressing effects were mimicked by the cGMP analogue 8-bromo-cGMP. Furthermore, in myocytes, 8-bromo-cGMP attenuated the alpha1-adrenergic receptor-stimulated increases in c-fos. Likewise, ANP and 8-bromo-cGMP attenuated the alpha1-adrenergic receptor- stimulated increase in prepro-ANP mRNA and the alpha1-adrenergic receptor-stimulated decrease in sarcoplasmic reticulum calcium ATPase mRNA. The L-type Ca2+ channel blockers verapamil and nifedipine inhibited NE-stimulated incorporation of [3H]leucine in myocytes and [3H]thymidine in fibroblasts, and these effects were not additive with those of ANP, SNAP, or 8-bromo-cGMP. In myocytes, the Ca2+ channel agonist BAY K8644 caused an increase in [3H]leucine incorporation which was inhibited by ANP. These findings indicate that NO and ANP can attenuate the effects of NE on the growth of cardiac myocytes and fibroblasts, most likely by a cGMP-mediated inhibition of NE-stimulated Ca2+ influx.


Assuntos
Agonistas alfa-Adrenérgicos/metabolismo , Fator Natriurético Atrial/metabolismo , GMP Cíclico/metabolismo , Fibroblastos/metabolismo , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Norepinefrina/metabolismo , Inibidores da Síntese de Proteínas/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Fator Natriurético Atrial/biossíntese , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/farmacologia , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/biossíntese , ATPases Transportadoras de Cálcio/genética , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Coração/efeitos dos fármacos , Miocárdio/citologia , Óxido Nítrico Sintase/antagonistas & inibidores , Norepinefrina/farmacologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/biossíntese , Ratos , S-Nitroso-N-Acetilpenicilamina , Retículo Sarcoplasmático/enzimologia , Timidina/farmacocinética , Trítio/farmacocinética , ômega-N-Metilarginina/farmacologia
20.
Surgery ; 142(2): 170-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17689682

RESUMO

BACKGROUND: The systemic response to injury is characterized by massive release of norepinephrine (NE) into the circulation as a result of global sympathetic activation. We have recently demonstrated that NE modulates the recruitment of macrophages to the cutaneous wound. We hypothesized that NE suppresses wound macrophage phagocytic function through canonical adrenergic signaling pathways. METHODS: Murine wound macrophages were harvested at 5 days after injury and treated with physiologic and pharmacologic dose norepinephrine. Phagocytosis of green fluorescent protein-labeled Escherichia coli was assayed by flow cytometry. The signaling pathways mediating NE modulation of wound macrophage phagocytosis were interrogated by pharmacologic manipulation of alpha- and beta-adrenoreceptors (ARs), intracellular cyclic adenosine monophosphate (cAMP), and protein kinase A (PKA). Tissue specificity was determined by comparison of wound macrophages to splenic macrophages. RESULTS: Both physiologic and pharmacologic dose NE suppressed wound macrophage phagocytic efficiency. This effect was mediated by alpha- and beta-ARs in a dose-dependent fashion. Direct stimulation of cAMP-suppressed phagocytic efficiency and blockade of PKA signaling prevented NE-mediated suppression of phagocytic efficiency. Splenic macrophage phagocytic efficiency was less than that of wound macrophages and was not altered by NE. CONCLUSIONS: NE has a profound immunosuppressive effect on wound macrophage function that is tissue specific and appears to be mediated through adrenergic receptors and their canonical downstream signaling pathway. Attenuation of post-injury immunosuppression represents another potential mechanism by which beta-AR blockade may reduce morbidity and mortality after severe injury.


Assuntos
Agonistas alfa-Adrenérgicos/imunologia , Macrófagos/metabolismo , Norepinefrina/imunologia , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Ferimentos e Lesões/imunologia , Agonistas alfa-Adrenérgicos/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Escherichia coli/imunologia , Isoquinolinas/farmacologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Inibidores de Proteínas Quinases/farmacologia , Baço/citologia , Baço/imunologia , Sulfonamidas/farmacologia , Ferimentos e Lesões/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa