Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Endocrinol Metab ; 320(2): E359-E378, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33284094

RESUMO

Cold- and diet-induced recruitment of brown adipose tissue (BAT) and the browning of white adipose tissue (WAT) are dynamic processes, and the recruited state attained is a state of dynamic equilibrium, demanding continuous stimulation to be maintained. An involvement of macrophages, classical proinflammatory (M1) or alternatively activated anti-inflammatory (M2), is presently discussed as being an integral part of these processes. If these macrophages play a mediatory role in the recruitment process, such an involvement would have to be maintained in the recruited state. We have, therefore, investigated whether the recruited state of these tissues is associated with macrophage accretion or attrition. We found no correlation (positive or negative) between total UCP1 mRNA levels (as a measure of recruitment) and proinflammatory macrophages in any adipose depot. We found that in young chow-fed mice, cold-induced recruitment correlated with accretion of anti-inflammatory macrophages; however, such a correlation was not seen when cold-induced recruitment was studied in diet-induced obese mice. Furthermore, the anti-inflammatory macrophage accretion was mediated via ß1/ß2-adrenergic receptors; yet, in their absence, and thus in the absence of macrophage accretion, recruitment proceeded normally. We thus conclude that the classical recruited state in BAT and inguinal (brite/beige) WAT is not paralleled by macrophage accretion or attrition. Our results make mediatory roles for macrophages in the recruitment process less likely.NEW & NOTEWORTHY A regulatory or mediatory role-positive or negative-for macrophages in the recruitment of brown adipose tissue is presently discussed. As the recruited state in the tissue is a dynamic process, maintenance of the recruited state would need persistent alterations in macrophage complement. Contrary to this expectation, we demonstrate here an absence of alterations in macrophage complement in thermogenically recruited brown-or brite/beige-adipose tissues. Macrophage regulation of thermogenic capacity is thus less likely.


Assuntos
Tecido Adiposo Bege/fisiologia , Tecido Adiposo Marrom/fisiologia , Macrófagos/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Termogênese , Tecido Adiposo Bege/citologia , Tecido Adiposo Marrom/citologia , Animais , Dieta/efeitos adversos , Regulação da Expressão Gênica , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
2.
Mol Cell Neurosci ; 109: 103563, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33039519

RESUMO

Recent work demonstrated that sympathetic neurons innervate the skeletal muscle near the neuromuscular junction (NMJ), and muscle sympathectomy and sympathomimetic agents strongly influence motoneuron synaptic vesicle release ex vivo. Moreover, reports attest that the pontine nucleus locus coeruleus (LC) projects to preganglionic sympathetic neurons and regulates human mobility and skeletal muscle physiology. Thus, we hypothesized that peripheral and central sympathetic neurons projecting directly or indirectly to the skeletal muscle regulate NMJ transmission. The aim of this study was to define the specific neuronal groups in the peripheral and central nervous systems that account for such regulation in adult mice in vivo by using optogenetics and NMJ transmission recordings in 3-5-month-old, male and female ChR2(H134R/EYFP)/TH-Cre mice. After detecting ChR2(H134R)/EYFP fluorescence in the paravertebral ganglia and LC neurons, we tested whether optostimulating the plantar nerve near the lumbricalis muscle or LC neurons effectively modulates motor nerve terminal synaptic vesicle release in living mice. Nerve optostimulation increased motor synaptic vesicle release in vitro and in vivo, while the presynaptic adrenoceptor blockers propranolol (ß1/ß2) and atenolol (ß1) prevented this outcome. The effect is primarily presynaptic since miniature end-plate potential (MEPP) kinetics remained statistically unmodified after stimulation. In contrast, optostimulation of LC neurons did not regulate NMJ transmission. In summary, we conclude that postganglionic sympathetic neurons, but not LC neurons, increased NMJ transmission by acting on presynaptic ß1-adrenergic receptors in vivo.


Assuntos
Locus Cerúleo/fisiologia , Neurônios Motores/fisiologia , Junção Neuromuscular/fisiologia , Optogenética/métodos , Transmissão Sináptica/fisiologia , Nervo Tibial/fisiologia , Animais , Channelrhodopsins/análise , Channelrhodopsins/genética , Dependovirus/fisiologia , Feminino , Gânglios Simpáticos/fisiologia , Genes Reporter , Proteínas de Fluorescência Verde/análise , Lasers , Luz , Masculino , Camundongos , Camundongos Transgênicos , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Neurônios Motores/efeitos da radiação , Mutação de Sentido Incorreto , Receptores Adrenérgicos beta 1/fisiologia , Proteínas Recombinantes de Fusão/análise , Fibras Simpáticas Pós-Ganglionares/fisiologia , Transmissão Sináptica/efeitos da radiação , Nervo Tibial/efeitos da radiação
3.
Am J Physiol Heart Circ Physiol ; 318(2): H264-H282, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31834834

RESUMO

The ß1-adrenergic regulation of cardiac myocyte contraction plays an important role in regulating heart function. Activation of this system leads to an increased heart rate and stronger myocyte contraction. However, chronic stimulation of the ß1-adrenergic signaling system can lead to cardiac hypertrophy and heart failure. To understand the mechanisms of action of ß1-adrenoceptors, a mathematical model of cardiac myocyte contraction that includes the ß1-adrenergic system was developed and studied. The model was able to simulate major experimental protocols for measurements of steady-state force-calcium relationships, cross-bridge release rate and force development rate, force-velocity relationship, and force redevelopment rate. It also reproduced quite well frequency and isoproterenol dependencies for intracellular Ca2+ concentration ([Ca2+]i) transients, total contraction force, and sarcomere shortening. The mathematical model suggested the mechanisms of increased contraction force and myocyte shortening on stimulation of ß1-adrenergic receptors is due to phosphorylation of troponin I and myosin-binding protein C and increased [Ca2+]i transient resulting from activation of the ß1-adrenergic signaling system. The model was used to simulate work-loop contractions and estimate the power during the cardiac cycle as well as the effects of 4-aminopyridine and tedisamil on the myocyte contraction. The developed mathematical model can be used further for simulations of contraction of ventricular myocytes from genetically modified mice and myocytes from mice with chronic cardiac diseases.NEW & NOTEWORTHY A new mathematical model of mouse ventricular myocyte contraction that includes the ß1-adrenergic system was developed. The model simulated major experimental protocols for myocyte contraction and predicted the effects of 4-aminopyridine and tedisamil on the myocyte contraction. The model also allowed for simulations of work-loop contractions and estimation of the power during the cardiac cycle.


Assuntos
Ventrículos do Coração , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Algoritmos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Cardiotônicos/farmacologia , Proteínas de Transporte/metabolismo , Simulação por Computador , Frequência Cardíaca/fisiologia , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Camundongos , Modelos Teóricos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação , Bloqueadores dos Canais de Potássio/farmacologia , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Sarcômeros/fisiologia , Troponina I/metabolismo , Troponina I/fisiologia
4.
J Physiol ; 597(7): 1819-1831, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30693527

RESUMO

KEY POINTS: The prevailing dogma about neurogenic regulation of vascular tone consists of major vasodilatation caused by CGRP (and possibly substance P) released from sensory-motor nerves and vasoconstriction caused by noradrenaline, ATP and neuropeptode Y release from sympathetic nerves. Most studies on perivascular nerve-mediated vasodilatation are made in vitro. In the present study, we provide evidence indicating that in vivo electrical perivascular nerve stimulation in rat mesenteric small arteries causes a large ß1-adrenoceptor-mediated vasodilatation, which contrasts with a smaller vasodilatation caused by endogenous CGRP that is only visible after inhibition of Y1 NPY receptors. ABSTRACT: Mesenteric arteries are densely innervated and the nerves are important regulators of vascular tone and hence blood pressure and blood flow. Perivascular sensory-motor nerves have been shown to cause vasodilatation in vitro. However, less is known about their function in vivo. Male Wistar rats (10-12 weeks old; n = 72) were anaesthetized with ketamine (3 mg kg-1 ) and xylazine (0.75 mg kg-1 ) or pentobarbital (60 mg kg-1 ). After a laparotomy, a section of second-order mesenteric artery was visualized in an organ bath after minimal removal of perivascular adipose tissue. The effects of electrical field stimulation (EFS) and drugs on artery diameter and blood flow were recorded with intravital microscopy and laser speckle imaging. EFS caused vasodilatation in arteries constricted with 1 µm U46619 in the presence of 140 µm suramin and 1 µm prazosin. The vasodilatation was inhibited by 1 µm tetrodotoxin and 5 µm guanethidine, although not by the 1 µm of the CGRP receptor antagonist BIBN4096bs. In the presence of 0.3 µm Y1 receptor antagonist BIBP3226, BIBN4096bs partly inhibited the vasodilatation. Atenolol at a concentration 1 µm inhibited the vasodilatation, whereas 0.1 µm of the ß2 -adrenoceptor selective antagonist ICI-118,551 had no effect. Increasing the extracellular [K+ ] to 20 mm caused vasodilatation but was converted to vasoconstriction in the presence of 1 µm BIBN4096bs, and constriction to 30 mm potassium was potentiated by BIBN4096bs. Atenolol but not BIBN4096bs increased contraction to EFS in the absence of suramin and prazosin. In mesenteric small arteries of anaesthetized rats, EFS failed to stimulate major dilatation via sensory-motor nerves but induced sympathetic ß1 -adrenoceptor-mediated dilatation.


Assuntos
Artérias Mesentéricas/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Vasodilatação/fisiologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Animais , Antinematódeos/farmacologia , Atenolol/farmacologia , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Piperazinas/farmacologia , Prazosina/farmacologia , Quinazolinas/farmacologia , Ratos , Ratos Wistar , Suramina/farmacologia , Técnicas de Cultura de Tecidos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos
5.
Eur J Neurosci ; 50(7): 3141-3163, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31162753

RESUMO

Despite vast literature on catecholaminergic neuromodulation of auditory cortex functioning in general, knowledge about its role for long-term memory formation is scarce. Our previous pharmacological studies on cortex-dependent frequency-modulated tone-sweep discrimination learning of Mongolian gerbils showed that auditory-cortical D1/5 -dopamine receptor activity facilitates memory consolidation and anterograde memory formation. Considering overlapping functions of D1/5 -dopamine receptors and ß-adrenoceptors, we hypothesised a role of ß-adrenergic signalling in the auditory cortex for sweep discrimination learning and memory. Supporting this hypothesis, the ß1/2 -adrenoceptor antagonist propranolol bilaterally applied to the gerbil auditory cortex after task acquisition prevented the discrimination increment that was normally monitored 1 day later. The increment in the total number of hurdle crossings performed in response to the sweeps per se was normal. Propranolol infusion after the seventh training session suppressed the previously established sweep discrimination. The suppressive effect required antagonist injection in a narrow post-session time window. When applied to the auditory cortex 1 day before initial conditioning, ß1 -adrenoceptor-antagonising and ß1 -adrenoceptor-stimulating agents retarded and facilitated, respectively, sweep discrimination learning, whereas ß2 -selective drugs were ineffective. In contrast, single-sweep detection learning was normal after propranolol infusion. By immunohistochemistry, ß1 - and ß2 -adrenoceptors were identified on the neuropil and somata of pyramidal and non-pyramidal neurons of the gerbil auditory cortex. The present findings suggest that ß-adrenergic signalling in the auditory cortex has task-related importance for discrimination learning of complex sounds: as previously shown for D1/5 -dopamine receptor signalling, ß-adrenoceptor activity supports long-term memory consolidation and reconsolidation; additionally, tonic input through ß1 -adrenoceptors may control mechanisms permissive for memory acquisition.


Assuntos
Córtex Auditivo/fisiologia , Aprendizagem por Discriminação/fisiologia , Memória/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Estimulação Acústica , Agonistas de Receptores Adrenérgicos beta 1/administração & dosagem , Antagonistas de Receptores Adrenérgicos beta 2/administração & dosagem , Animais , Gerbillinae , Masculino , Propranolol/administração & dosagem
6.
Proc Natl Acad Sci U S A ; 111(46): 16604-9, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25368166

RESUMO

Cardiac disorders are the main cause of mortality in autosomal-dominant polycystic kidney disease (ADPKD). However, how mutated polycystins predispose patients with ADPKD to cardiac pathologies before development of renal dysfunction is unknown. We investigate the effect of decreased levels of polycystin 2 (PC2), a calcium channel that interacts with the ryanodine receptor, on myocardial function. We hypothesize that heterozygous PC2 mice (Pkd2(+/-)) undergo cardiac remodeling as a result of changes in calcium handling, separate from renal complications. We found that Pkd2(+/-) cardiomyocytes have altered calcium handling, independent of desensitized calcium-contraction coupling. Paradoxically, in Pkd2(+/-) mice, protein kinase A (PKA) phosphorylation of phospholamban (PLB) was decreased, whereas PKA phosphorylation of troponin I was increased, explaining the decoupling between calcium signaling and contractility. In silico modeling supported this relationship. Echocardiography measurements showed that Pkd2(+/-) mice have increased left ventricular ejection fraction after stimulation with isoproterenol (ISO), a ß-adrenergic receptor (ßAR) agonist. Blockers of ßAR-1 and ßAR-2 inhibited the ISO response in Pkd2(+/-) mice, suggesting that the dephosphorylated state of PLB is primarily by ßAR-2 signaling. Importantly, the Pkd2(+/-) mice were normotensive and had no evidence of renal cysts. Our results showed that decreased PC2 levels shifted the ßAR pathway balance and changed expression of calcium handling proteins, which resulted in altered cardiac contractility. We propose that PC2 levels in the heart may directly contribute to cardiac remodeling in patients with ADPKD in the absence of renal dysfunction.


Assuntos
Sinalização do Cálcio/fisiologia , Acoplamento Excitação-Contração/fisiologia , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Canais de Cátion TRPP/deficiência , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Pressão Sanguínea/genética , Cafeína/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/metabolismo , Estimulação Cardíaca Artificial , Acoplamento Excitação-Contração/efeitos dos fármacos , Acoplamento Excitação-Contração/genética , Heterozigoto , Isoproterenol/farmacologia , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Mensageiro/biossíntese , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/fisiologia , Troponina I/metabolismo , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/genética , Remodelação Ventricular/fisiologia
7.
Circ Res ; 115(2): 273-83, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24807785

RESUMO

RATIONALE: The sympathetic nervous system plays a fundamental role in the regulation of myocardial function. During chronic pressure overload, overactivation of the sympathetic nervous system induces the release of catecholamines, which activate ß-adrenergic receptors in cardiomyocytes and lead to increased heart rate and cardiac contractility. However, chronic stimulation of ß-adrenergic receptors leads to impaired cardiac function, and ß-blockers are widely used as therapeutic agents for the treatment of cardiac disease. MicroRNA-133 (miR-133) is highly expressed in the myocardium and is involved in controlling cardiac function through regulation of messenger RNA translation/stability. OBJECTIVE: To determine whether miR-133 affects ß-adrenergic receptor signaling during progression to heart failure. METHODS AND RESULTS: Based on bioinformatic analysis, ß1-adrenergic receptor (ß1AR) and other components of the ß1AR signal transduction cascade, including adenylate cyclase VI and the catalytic subunit of the cAMP-dependent protein kinase A, were predicted as direct targets of miR-133 and subsequently validated by experimental studies. Consistently, cAMP accumulation and activation of downstream targets were repressed by miR-133 overexpression in both neonatal and adult cardiomyocytes following selective ß1AR stimulation. Furthermore, gain-of-function and loss-of-function studies of miR-133 revealed its role in counteracting the deleterious apoptotic effects caused by chronic ß1AR stimulation. This was confirmed in vivo using a novel cardiac-specific TetON-miR-133 inducible transgenic mouse model. When subjected to transaortic constriction, TetON-miR-133 inducible transgenic mice maintained cardiac performance and showed attenuated apoptosis and reduced fibrosis compared with control mice. CONCLUSIONS: miR-133 controls multiple components of the ß1AR transduction cascade and is cardioprotective during heart failure.


Assuntos
AMP Cíclico/fisiologia , MicroRNAs/fisiologia , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Regiões 3' não Traduzidas/fisiologia , Adenilil Ciclases/fisiologia , Animais , Apoptose , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Progressão da Doença , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Masculino , Metoprolol/farmacologia , Metoprolol/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/genética , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética
8.
BJU Int ; 117(6): 993-1001, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26332379

RESUMO

OBJECTIVES: To clarify the involvement of bladder α1-adrenoceptors (α1-ARs) in afferent pathways by investigating the effects of silodosin and BMY7378, selective α1A- or α1D-AR antagonists, respectively, on single-unit afferent nerve fibre activity (SAA) of the primary bladder afferent nerves and their relationship with bladder microcontractions in rats. MATERIALS AND METHODS: A total of 63 female Sprague-Dawley rats were anaesthetized with urethane. The SAA of Aδ and C fibres generated from the left L6 dorsal roots was determined using electrical stimulation of the left pelvic nerve and bladder distension. After measuring baseline SAA during constant filling cystometry, the procedure was repeated with i.v. (0.3-30 µg/kg) or intravesical (10 µm) administration of each antagonist. In separate rats, the bladder was filled with saline until the intravesical pressure reached 30 cmH2 O, and was kept under isovolumetric conditions, then the recording was performed with i.v.-administered vehicle and silodosin (0.3 µg/kg). RESULTS: A total of Aδ fibres and 33 C fibres were isolated from 63 rats. The SAA of Aδ fibres, but not C fibres, were dose-dependently decreased after both i.v. and intravesical administrations of each of the antagonists. In the experiments under bladder isovolumetric conditions, silodosin administration significantly decreased the SAA of Aδ fibres, but not C fibres, compared with vehicle administration. There were no significant effects on either the mean basal bladder pressure or microcontractions. CONCLUSION: The present study suggests that both α1A- or α1D-ARs in the rat bladder are involved in the activation of the bladder mechanosensory Aδ fibres during bladder filling, and that this activation may not be related to bladder microcontractions.


Assuntos
Receptores Adrenérgicos beta 1/fisiologia , Bexiga Urinária/fisiologia , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Animais , Feminino , Contração Muscular/efeitos dos fármacos , Fibras Nervosas Amielínicas/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Ratos , Ratos Sprague-Dawley , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/inervação
9.
Circ Res ; 114(6): 993-1003, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24508724

RESUMO

RATIONALE: ß-Adrenoceptor activation contributes to sudden death risk in heart failure. Chronic ß-adrenergic stimulation, as occurs in patients with heart failure, causes potentially arrhythmogenic reductions in slow delayed-rectifier K(+) current (IKs). OBJECTIVE: To assess the molecular mechanisms of IKs downregulation caused by chronic ß-adrenergic activation, particularly the role of exchange protein directly activated by cAMP (Epac). METHODS AND RESULTS: Isolated guinea pig left ventricular cardiomyocytes were incubated in primary culture and exposed to isoproterenol (1 µmol/L) or vehicle for 30 hours. Sustained isoproterenol exposure decreased IKs density (whole cell patch clamp) by 58% (P<0.0001), with corresponding decreases in potassium voltage-gated channel subfamily E member 1 (KCNE1) mRNA and membrane protein expression (by 45% and 51%, respectively). Potassium voltage-gated channel, KQT-like subfamily, member 1 (KCNQ1) mRNA expression was unchanged. The ß1-adrenoceptor antagonist 1-[2-((3-Carbamoyl-4-hydroxy)phenoxy)ethylamino]-3-[4-(1-methyl-4-trifluoromethyl-2-imidazolyl)phenoxy]-2-propanol dihydrochloride (CGP-20712A) prevented isoproterenol-induced IKs downregulation, whereas the ß2-antagonist ICI-118551 had no effect. The selective Epac activator 8-pCPT-2'-O-Me-cAMP decreased IKs density to an extent similar to isoproterenol exposure, and adenoviral-mediated knockdown of Epac1 prevented isoproterenol-induced IKs/KCNE1 downregulation. In contrast, protein kinase A inhibition with a cell-permeable highly selective peptide blocker did not affect IKs downregulation. 1,2-Bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetate-AM acetoxymethyl ester (BAPTA-AM), cyclosporine, and inhibitor of nuclear factor of activated T cell (NFAT)-calcineurin association-6 (INCA6) prevented IKs reduction by isoproterenol and INCA6 suppressed isoproterenol-induced KCNE1 downregulation, consistent with signal-transduction via the Ca(2+)/calcineurin/NFAT pathway. Isoproterenol induced nuclear NFATc3/c4 translocation (immunofluorescence), which was suppressed by Epac1 knockdown. Chronic in vivo administration of isoproterenol to guinea pigs reduced IKs density and KCNE1 mRNA and protein expression while inducing cardiac dysfunction and action potential prolongation. Selective in vivo activation of Epac via sp-8-pCPT-2'-O-Me-cAMP infusion decreased IKs density and KCNE1 mRNA/protein expression. CONCLUSIONS: Prolonged ß1-adrenoceptor stimulation suppresses IKs by downregulating KCNE1 mRNA and protein via Epac-mediated Ca(2+)/calcineurin/NFAT signaling. These results provide new insights into the molecular basis of K(+) channel remodeling under sustained adrenergic stimulation.


Assuntos
Agonistas Adrenérgicos beta/toxicidade , Canais de Potássio de Retificação Tardia/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Ativação do Canal Iônico/efeitos dos fármacos , Isoproterenol/toxicidade , Receptores Adrenérgicos beta 1/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Calcineurina/fisiologia , Cálcio/farmacologia , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Cobaias , Hipertrofia Ventricular Esquerda/etiologia , Imidazóis/farmacologia , Ativação do Canal Iônico/fisiologia , Isoproterenol/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC/metabolismo , Técnicas de Patch-Clamp , Potássio/metabolismo , Propanolaminas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Sistemas do Segundo Mensageiro/fisiologia
10.
Pharmacogenet Genomics ; 25(11): 555-63, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26313487

RESUMO

INTRODUCTION: The ADRB1 389 polymorphism affects responses to the ß-1 adrenergic receptor (ß1AR) agonist in vitro. Previous studies on its effect on the response to dobutamine stress echocardiography were conflicting. In addition, sex differences in the response to dobutamine have been suggested. The aim of this study was to determine whether the ADRB1 389 polymorphism affects the hemodynamic response to dobutamine in healthy individuals including men and women. PARTICIPANTS AND METHODS: Healthy individuals were recruited according to their ADRB1 49 and 389 genotypes [15 Arg389Arg, 10 Gly389Arg, and 10 Gly389Gly individuals, (all Ser49Ser), 21 men and 14 women]. Dobutamine was infused at 2, 4, and 6 mcg/kg/min. Standardized exercise was performed during the last minute of each infusion. RESULTS: Resting heart rate (HR) response to 6 mcg/kg/min dobutamine (ΔHR) was 4.7-fold larger in Arg389Arg than in Gly389Gly [(mean ± SD) 12.95 ± 6.99, 2.75 ± 1.65 bpm, respectively, PANOVA=0.012]. Renin response to dobutamine (ΔRenin) was 3.9-fold greater in Arg389Arg than in Gly389Gly (PANOVA=0.032). Among Arg389Gly heterozygotes, ΔHR and ΔRenin were not significantly different from either homozygote group. In multivariate analysis for ΔHR variance, significant contributions were observed for genotype (P=0.011), baseline HR (P=0.011), and borderline effect for sex (P=0.049). CONCLUSION: In healthy individuals, HR and renin responses to dobutamine were more than three-fold greater among ADRB1 Arg389 compared with Gly389 homozygotes. Future studies on the effect of the ADRB1 389 polymorphism on dobutamine stress echocardiography should compare Arg389 and Gly389 homozygotes.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Dobutamina/farmacologia , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/genética , Polimorfismo de Nucleotídeo Único , Receptores Adrenérgicos beta 1/genética , Adulto , Exercício Físico/fisiologia , Feminino , Genótipo , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/genética , Homozigoto , Humanos , Masculino , Análise Multivariada , Receptores Adrenérgicos beta 1/fisiologia , Renina/sangue , Caracteres Sexuais , Adulto Jovem
11.
Circulation ; 127(8): 913-22, 2013 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23363625

RESUMO

BACKGROUND: ß-Adrenergic receptor (ß-AR) activation can provoke cardiac arrhythmias mediated by cAMP-dependent alterations of Ca(2+) signaling. However, cAMP can activate both protein kinase A and an exchange protein directly activated by cAMP (Epac), but their functional interaction is unclear. In heart, selective Epac activation can induce potentially arrhythmogenic sarcoplasmic reticulum (SR) Ca(2+) release that involves Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) effects on the ryanodine receptor (RyR). METHODS AND RESULTS: We tested whether physiological ß-AR activation causes Epac-mediated SR Ca(2+) leak and arrhythmias and whether it requires Epac1 versus Epac2, ß(1)-AR versus ß(2)-AR, and CaMKIIδ-dependent phosphorylation of RyR2-S2814. We used knockout (KO) mice for Epac1, Epac2, or both. All KOs exhibited unaltered basal cardiac function, Ca(2+) handling, and hypertrophy in response to pressure overload. However, SR Ca(2+) leak induced by the specific Epac activator 8-CPT in wild-type mice was abolished in Epac2-KO and double-KO mice but was unaltered in Epac1-KO mice. ß-AR-induced arrhythmias were also less inducible in Epac2-KO versus wild-type mice. ß-AR activation with protein kinase A inhibition mimicked 8-CPT effects on SR Ca(2+) leak and was prevented by blockade of ß(1)-AR but not ß(2)-AR. CaMKII inhibition (KN93) and genetic ablation of either CaMKIIδ or CaMKII phosphorylation on RyR2-S2814 prevented 8-CPT-induced SR Ca(2+) leak. CONCLUSIONS: ß(1)-AR activates Epac2 to induce SR Ca(2+) leak via CaMKIIδ-dependent phosphorylation of RyR2-S2814. This pathway contributes to ß-AR-induced arrhythmias and reduced cardiac function.


Assuntos
Arritmias Cardíacas/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/deficiência , Cálcio/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Retículo Sarcoplasmático/metabolismo , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/fisiopatologia , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Retículo Sarcoplasmático/patologia
12.
Pharmacology ; 93(5-6): 272-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25096537

RESUMO

The prelimbic region of the medial prefrontal cortex (mPFC) in the brain is crucial for memory. Norepinephrine elicits an important influence on mPFC functions. The stimulation of ß-adrenoceptors (ß-ARs) may play a critical role in the consolidation of long-term memory. The present study examines the possible role of ß1-ARs located in the mPFC on morphine-induced amnesia in rats. The animals were bilaterally implanted with chronic cannulas in the mPFC, trained in a step-through-type passive avoidance task and tested 24 h after training to measure step-through latency. Our present results indicated that posttraining intraperitoneal administration of morphine (2.5, 5 and 7.5 mg/kg) dose-dependently reduced the step-through latency. Different doses of xamoterol (0.01, 0.1 and 1 µg/rat) have shown no significant change in the step-through latency, but posttraining intra-mPFC microinjection of atenolol (0.2 and 0.4 µg/rat) had an amnesic effect. Moreover, atenolol-caused amnesia was reversed by an ineffective dose of xamoterol (0.1 µg/rat). On the other hand, coadministration of an ineffective dose of atenolol (0.1 µg/rat) with an ineffective dose of morphine (2.5 mg/kg) induced an amnesic effect. Meanwhile, xamoterol had no effect on morphine-induced amnesia. These results suggest that ß1-ARs of the prelimbic region in the mPFC may play an important role in morphine-induced amnesia.


Assuntos
Amnésia/fisiopatologia , Córtex Pré-Frontal/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Amnésia/induzido quimicamente , Animais , Atenolol/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Morfina , Ratos Wistar , Xamoterol/farmacologia
13.
J Neurophysiol ; 110(2): 279-85, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23615548

RESUMO

Long-term effects of stress during pregnancy on brain and behavior have been analyzed extensively in recent years. These effects include changes in emotional behavior, a reduction in learning capacity, and ability to generate long-term potentiation (LTP) in the offspring. In earlier studies, we and others have described a difference in ability to express LTP in dorsal and ventral sectors of the hippocampus (DH and VH, respectively) and its modification by prior stress. We now found that norepinephrine (NE) facilitated conversion of short-term potentiation to LTP in the normal DH but not in VH. Prenatal stress (PS) switched the locus of the facilitating action of NE from the DH to the VH. The effects of NE are likely to be mediated by activation of calcium stores. PS also facilitated (S)-3,5-dihydroxyphenylglycine hydrate (DHPG)-induced LTD in the VH, assumed to be mediated by release of calcium from stores. These observations have important implications for the role of the hippocampus in cognitive and emotional memories.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Potenciação de Longa Duração/fisiologia , Exposição Materna , Receptores Adrenérgicos beta 1/fisiologia , Estresse Fisiológico/fisiologia , Animais , Região CA1 Hipocampal/embriologia , Feminino , Técnicas In Vitro , Gravidez , Ratos , Ratos Wistar
14.
Exp Physiol ; 98(5): 973-87, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23243148

RESUMO

Secretin has been implicated in cardiovascular regulation through its specific receptors, as well as through ß-adrenoceptors and nitric oxide, although data on its direct effect on coronary blood flow and cardiac function have remained scarce. The present study aimed to determine the primary in vivo effect of secretin on cardiac function and perfusion and the mechanisms related to the autonomic nervous system, secretin receptors and NO. In addition, in coronary endothelial cells the intracellular pathways involved in the effects of secretin on NO release were also examined. In 30 pigs, intracoronary secretin infusion at 2.97 pg for each millilitre per minute of coronary blood flow at constant heart rate and aortic blood pressure increased coronary blood flow, maximal rate of change of left ventricular pressure, segmental shortening, cardiac output and coronary NO release (P<0.05). These responses were graded in a further five pigs. Moreover, while blockade of muscarinic cholinoreceptors (n=5) and of α-adrenoceptors (n=5) did not abolish the observed responses to secretin, blockade of ß1-adrenoceptors (n=5) prevented the effects of secretin on cardiac function. In addition, blockade of ß2-adrenoceptors (n=5) and NO synthase inhibition (n=5) prevented the coronary response and the effect of secretin on NO release. All these effects were abolished by a secretin receptor inhibitor (n=5). In coronary endothelial cells, the increased NO production caused by secretin was found to be related to cAMP/protein kinase A signalling activated as downstream effectors of stimulation of secretin receptors and ß2-adrenoceptors. In conclusion, in anaesthetized pigs secretin primarily increased cardiac function and perfusion through the involvement of specific receptors, ß-adrenoceptors and NO release.


Assuntos
Coração/efeitos dos fármacos , Óxido Nítrico/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Secretina/fisiologia , Animais , Células Cultivadas , Circulação Coronária/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Coração/fisiologia , Hemodinâmica/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Perfusão , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Secretina/administração & dosagem , Secretina/farmacologia , Suínos , Vasodilatação/efeitos dos fármacos
15.
Proc Natl Acad Sci U S A ; 107(36): 15868-73, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20713709

RESUMO

Ghrelin, an octanoylated peptide hormone produced in the stomach, rises dramatically in mouse plasma during chronic severe calorie deprivation, an event that is essential to maintain life. The mechanism for this increase is not understood. Here, we study the control of ghrelin secretion in tissue culture cells derived from mice bearing ghrelinomas induced by a tissue-specific SV40 T-antigen transgene. We found that the ghrelin-secreting cells express high levels of mRNA encoding beta(1)-adrenergic receptors. Addition of norepinephrine or epinephrine to the culture medium stimulated ghrelin secretion, and this effect was blocked by atenolol, a selective beta(1)-adrenergic antagonist. When WT mice were treated with reserpine to deplete adrenergic neurotransmitters from sympathetic neurons, the fasting-induced increase in plasma ghrelin was blocked. Inhibition was also seen following atenolol administration. We conclude that ghrelin secretion during fasting is induced by adrenergic agents released by sympathetic neurons and acting directly on beta(1) receptors on the ghrelin-secreting cells of the stomach.


Assuntos
Grelina/metabolismo , Neoplasias Experimentais/metabolismo , Receptores Adrenérgicos beta 1/fisiologia , Animais , Perfilação da Expressão Gênica , Camundongos , Neoplasias Experimentais/patologia , RNA Mensageiro/genética , Ratos , Receptores Adrenérgicos beta 1/genética
16.
EMBO J ; 27(2): 384-93, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18188154

RESUMO

Beta1- and beta2-adrenergic receptors (betaARs) are highly homologous, yet they play clearly distinct roles in cardiac physiology and pathology. Myocyte contraction, for instance, is readily stimulated by beta1AR but not beta2AR signaling, and chronic stimulation of the two receptors has opposing effects on myocyte apoptosis and cell survival. Differences in the assembly of macromolecular signaling complexes may explain the distinct biological outcomes. Here, we demonstrate that beta1AR forms a signaling complex with a cAMP-specific phosphodiesterase (PDE) in a manner inherently different from a beta2AR/beta-arrestin/PDE complex reported previously. The beta1AR binds a PDE variant, PDE4D8, in a direct manner, and occupancy of the receptor by an agonist causes dissociation of this complex. Conversely, agonist binding to the beta2AR is a prerequisite for the recruitment of a complex consisting of beta-arrestin and the PDE4D variant, PDE4D5, to the receptor. We propose that the distinct modes of interaction with PDEs result in divergent cAMP signals in the vicinity of the two receptors, thus, providing an additional layer of complexity to enforce the specificity of beta1- and beta2-adrenoceptor signaling.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular , Células Cultivadas , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Humanos , Imunoprecipitação , Camundongos , Modelos Biológicos , Células Musculares/citologia , Células Musculares/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/fisiologia , Transdução de Sinais
17.
J Pharmacol Exp Ther ; 343(2): 451-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22904357

RESUMO

Ethanol (EtOH) potentiation of GABAergic neurotransmission in the basolateral amygdala (BLA) may contribute to the acute anxiolytic effects of this drug. Previous studies have shown that BLA pyramidal neurons receive GABAergic input from two distinct sources: local interneurons and a cluster of GABAergic cells termed lateral paracapsular (LPCS) interneurons. It is noteworthy that whereas EtOH enhances local GABAergic synapses via a presynaptic increase in GABA release, EtOH potentiation of LPCS inhibition is mediated via a distinct mechanism that requires adrenoceptor (AR) activation. Here, we sought to further characterize the interaction between the AR system and EtOH enhancement of LPCS GABAergic synapses by using in vitro electrophysiology techniques in male Sprague-Dawley rats. Exogenous norepinephrine (NE) enhanced LPCS-evoked inhibitory postsynaptic currents (eIPSCs) via the activation of ß-ARs, because this effect was blocked by propranolol. EtOH potentiation of LPCS eIPSCs was also blocked by propranolol and significantly reduced by NE pretreatment, suggesting that NE and EtOH may enhance LPCS inhibition via a common mechanism. EtOH enhancement of LPCS eIPSCs was significantly reduced by a selective ß1-, but not ß2- or ß3-, AR antagonist, and both EtOH and NE potentiation of LPCS IPSCs was blocked by postsynaptic disruption of cAMP signaling. These data suggest that EtOH enhances LPCS synapses via a postsynaptic ß1-AR, cAMP-dependent cascade. Because enhancement of LPCS inhibition can reduce anxiety-like behaviors, these findings shed light on a novel mechanism that may play a role in some of the anxiolytic effects of EtOH that are thought to contribute to the development and progression of alcoholism.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Receptores Adrenérgicos beta 1/fisiologia , Sinapses/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas In Vitro , Masculino , Norepinefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
18.
Can J Physiol Pharmacol ; 90(4): 407-14, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22448877

RESUMO

Circulating autoantibodies directed against the 2nd extracellular loop (EL-2) of ß(1)-adrenoceptors (ß(1)-AABs) have been detected in the serum of patients with various cardiovascular pathologies. ß(1)-AABs induce agonistic, positive inotropic effects via ß(1)-adrenoceptors (ß(1)ARs). In the mammalian heart, ß(1)-AR can exist in 2 distinct activated configurations (the so-called high- and low-affinity states). The aim of the present study was to investigate whether the action of ß(1)-AAB is dependent on the affinity state of ß(1)AR in isolated ventricular cardiomyocytes of adult Wistar rats. Immunoglobulin G (IgG) containing ß(1)-AAB obtained from animals immunized with a peptide corresponding to the EL-2 of human ß(1)-AR, caused a dose-dependent increase in cell shortening. Isoproterenol-induced inotropy was significantly reduced in cardiomyocytes that had been preincubated with IgG containing ß(1)-AAB and in cardiomyocytes isolated from immunized rats. The negative effects of preincubation with IgG containing ß(1)-AAB on the response to isoproterenol was inhibited in the presence of bisoprolol. CGP 12177A and pindolol-induced inotropy was not affected by IgG preincubation or immunization. No detectable inotropic effect of cell shortening was obtained with IgG containing ß(1)-AAB in the presence of propranolol and 3-isobutyl-1-methylxanthine. The present study demonstrates that ß(1)-AABs have no agonist/antagonist-like effects upon low-affinity state ß(1)-ARs. This result indicates that ß(1)-AABs recognize and stabilize the high-affinity state, but are unable to stabilize and (or) induce the low-affinity state receptor.


Assuntos
Autoanticorpos/farmacologia , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Animais , Autoanticorpos/imunologia , Bisoprolol/farmacologia , Isoproterenol/antagonistas & inibidores , Isoproterenol/farmacologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Pindolol/farmacologia , Propanolaminas/farmacologia , Propranolol/farmacologia , Ratos , Ratos Wistar , Receptores Adrenérgicos beta 1/imunologia , Receptores Adrenérgicos beta 1/metabolismo
19.
Geroscience ; 44(1): 329-348, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34608562

RESUMO

Our past study showed that coronary arterioles isolated from adipose-derived stromal vascular fraction (SVF)-treated rats showed amelioration of the age-related decrease in vasodilation to beta-adrenergic receptor (ß-AR) agonist and improved ß-AR-dependent coronary flow and microvascular function in a model of advanced age. We hypothesized that intravenously (i.v.) injected SVF improves coronary microvascular function in aged rats by re-establishing the equilibrium of the negative regulators of the internal adrenergic signaling cascade, G-protein receptor kinase 2 (GRK2) and G-alpha inhibitory (Gαi) proteins, back to youthful levels. Female Fischer-344 rats aged young (3 months, n = 24), old (24 months, n = 26), and old animals that received 1 × 107 green fluorescent protein (GFP+) SVF cells (O + SVF, n = 11) 4 weeks prior to sacrifice were utilized. Overnight urine was collected prior to sacrifice for catecholamine measurements. Cardiac samples were used for western blotting while coronary arterioles were isolated for pressure myography studies, immunofluorescence staining, and RNA sequencing. Coronary microvascular levels of the ß1 adrenergic receptor are decreased with advancing age, but this decreased expression was rescued by SVF treatment. Aging led to a decrease in phosphorylated GRK2 in cardiomyocytes vs. young control with restoration of phosphorylation status by SVF. In vessels, there was no change in genetic transcription (RNAseq) or protein expression (immunofluorescence); however, inhibition of GRK2 (paroxetine) led to improved vasodilation to norepinephrine in the old control (OC) and O + SVF, indicating greater GRK2 functional inhibition of ß1-AR in aging. SVF works to improve adrenergic-mediated vasodilation by restoring the ß1-AR population and mitigating signal cascade inhibitors to improve vasodilation.


Assuntos
Envelhecimento , Terapia Baseada em Transplante de Células e Tecidos , Envelhecimento/patologia , Animais , Circulação Coronária , Feminino , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Microcirculação , Ratos , Receptores Adrenérgicos beta 1/fisiologia , Vasodilatação
20.
Am J Physiol Renal Physiol ; 301(1): F71-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21389089

RESUMO

During nephrogenesis, renin expression shifts from large renal arteries toward smaller vessels in a defined spatiotemporal pattern, finally becoming restricted to the juxtaglomerular position. Chronic stimulation in adult kidneys leads to a recruitment of renin expression in the upstream vasculature. The mechanisms that control this characteristic switch-on and switch-off in the immature and adult kidney are not well-understood. Previous studies in mice with juxtaglomerular cell-specific deletion of the adenylyl cyclase-stimulatory G protein Gsα suggested that signaling along the cAMP pathway plays an essential role for renin expression during nephrogenesis and in the adult kidney. To identify the Gsα-dependent receptor that might be involved in activating this pathway, the present studies were performed to compare renin expression in wild types with that in mice with targeted deletions of ß(1) and ß(2)-adrenoceptors. The sympathetic nervous system is an important regulator of the renin system in the adult kidney so that activation of ß-adrenenoceptors may also participate in the activation of renin expression along the developing arterial tree and in upstream vasculature in adulthood. Compared with wild-types, renin expression was found to be significantly lower at all developmental stages in the kidneys of ß(1)/ß(2) Adr(-/-) mice. Three-dimensional analysis showed reduced renin expression in all segments of the vascular tree in mutants and a virtual absence of renin expression in the large arcuate arteries. Adult mutant kidneys showed the typical upstream renin expression after chronic stimulation. Tyrosine hydroxylase staining in fetal and postnatal kidneys revealed that sympathetic innervation of renin-producing cells occurs early in fetal development. Our data indicate that genetic disruption of ß-adrenergic receptors reduces basal renin expression along the developing preglomerular tree and in adult kidneys. Furthermore, ß-adrenergic receptor input is critical for the expression of renin in large renal vessels during early fetal development.


Assuntos
Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Rim/embriologia , Rim/metabolismo , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Renina/biossíntese , Animais , Arteríolas/metabolismo , Feminino , Desenvolvimento Fetal , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Renina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA