Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.018
Filtrar
1.
Neuropsychopharmacology ; 49(6): 1014-1023, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38368493

RESUMO

In the central nervous system, noradrenaline transmission controls the degree to which we are awake, alert, and attentive. Aberrant noradrenaline transmission is associated with pathological forms of hyper- and hypo-arousal that present in numerous neuropsychiatric disorders often associated with dysfunction in serotonin transmission. In vivo, noradrenaline regulates the release of serotonin because noradrenergic input drives the serotonin neurons to fire action potentials via activation of excitatory α1-adrenergic receptors (α1-AR). Despite the critical influence of noradrenaline on the activity of dorsal raphe serotonin neurons, the source of noradrenergic afferents has not been resolved and the presynaptic mechanisms that regulate noradrenaline-dependent synaptic transmission have not been described. Using an acute brain slice preparation from male and female mice and electrophysiological recordings from dorsal raphe serotonin neurons, we found that selective optogenetic activation of locus coeruleus terminals in the dorsal raphe was sufficient to produce an α1-AR-mediated excitatory postsynaptic current (α1-AR-EPSC). Activation of inhibitory α2-adrenergic receptors (α2-AR) with UK-14,304 eliminated the α1-AR-EPSC via presynaptic inhibition of noradrenaline release, likely via inhibition of voltage-gated calcium channels. In a subset of serotonin neurons, activation of postsynaptic α2-AR produced an outward current through activation of GIRK potassium conductance. Further, in vivo activation of α2-AR by systemic administration of clonidine reduced the expression of c-fos in the dorsal raphe serotonin neurons, indicating reduced neural activity. Thus, α2-AR are critical regulators of serotonin neuron excitability.


Assuntos
Núcleo Dorsal da Rafe , Locus Cerúleo , Receptores Adrenérgicos alfa 2 , Neurônios Serotoninérgicos , Transmissão Sináptica , Animais , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/fisiologia , Núcleo Dorsal da Rafe/metabolismo , Masculino , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/fisiologia , Feminino , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Camundongos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Optogenética , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Camundongos Endogâmicos C57BL , Norepinefrina/metabolismo , Camundongos Transgênicos
2.
Cell Tissue Res ; 387(2): 303-314, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34837110

RESUMO

Renal α2-adrenoceptors have been reported to play a role in the regulation of urinary output, renin secretion, and water and sodium excretion in the kidneys. However, the distribution of α2-adrenoceptor subtypes in the kidneys remains unclear. In this study, we aimed to investigate the localization of α2-adrenoceptor subtypes in rat kidneys using 8-week-old Sprague-Dawley rats. Immunofluorescence imaging revealed that both α2A- and α2B-adrenoceptors were expressed in the basolateral, but not apical, membrane of the epithelial cells of the proximal tubules. We also found that α2A- and α2B-adrenoceptors were not expressed in the glomeruli, collecting ducts, or the descending limb of the loop of Henle and vasa recta. In contrast, α2C-adrenoceptors were found to be localized in the glomeruli and lumen of the cortical and medullary collecting ducts. These results suggest that noradrenaline may act on the basement membrane of the proximal tubules through α2A- and α2B-adrenoceptors. Moreover, noradrenaline may be involved in the regulation of glomerular filtration and proteinuria through the induction of morphological changes in mesangial cells and podocytes via α2C-adrenoceptors. In the collecting ducts, urinary noradrenaline may regulate morphological changes of the microvilli through α2C-adrenoceptors. Our findings provide an immunohistochemical basis for understanding the cellular targets of α2-adrenergic regulation in the kidneys. This may be used to devise therapeutic strategies targeting α2-adrenoceptors.


Assuntos
Receptores Adrenérgicos alfa 2 , Roedores , Animais , Rim , Norepinefrina , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/fisiologia
3.
Sci Rep ; 11(1): 21645, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34737352

RESUMO

Vagus nerve stimulation (VNS) paired with rehabilitation training is emerging as a potential treatment for improving recovery of motor function following stroke. In rats, VNS paired with skilled forelimb training results in significant reorganization of the somatotopic cortical motor map; however, the mechanisms underlying this form of VNS-dependent plasticity remain unclear. Recent studies have shown that VNS-driven cortical plasticity is dependent on noradrenergic innervation of the neocortex. In the central nervous system, noradrenergic α2 receptors (α2-ARs) are widely expressed in the motor cortex and have been critically implicated in synaptic communication and plasticity. In current study, we examined whether activation of cortical α2-ARs is necessary for VNS-driven motor cortical reorganization to occur. Consistent with previous studies, we found that VNS paired with motor training enlarges the map representation of task-relevant musculature in the motor cortex. Infusion of α2-AR antagonists into M1 blocked VNS-driven motor map reorganization from occurring. Our results suggest that local α2-AR activation is required for VNS-induced cortical reorganization to occur, providing insight into the mechanisms that may underlie the neuroplastic effects of VNS therapy.


Assuntos
Córtex Motor/metabolismo , Plasticidade Neuronal/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Córtex Motor/fisiologia , Movimento/fisiologia , Norepinefrina , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos , Receptores Adrenérgicos alfa 2/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Reabilitação do Acidente Vascular Cerebral/métodos , Nervo Vago/metabolismo , Nervo Vago/fisiologia , Estimulação do Nervo Vago/métodos
4.
Behav Brain Res ; 414: 113513, 2021 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-34358575

RESUMO

Male Japanese quail produce high-frequency crow vocalizations to attract females during the breeding season. The nucleus of intercollicularis (ICo) is the midbrain vocal center in birds and electrical stimulation of the ICo produces calls that include crowing. Noradrenaline plays a significant role in sexual behavior but the contribution of noradrenaline in the control of courtship vocalizations in quail has not been well established. Using dose-dependent intracerebroventricular injection of clonidine, an α2-adrenergic receptor-specific agonist, crowing vocalization was immediately suppressed. At the same time as crow suppression by clonidine there was a reduction of immediate early gene, zenk mRNA, in the ICo; no zenk mRNA expression was detected in the dorsomedial division of the nucleus. Using histochemistry, we determined that the ICo receives noradrenergic innervation and expresses α2A-adrenergic receptor mRNA. Taken together, these data suggest that noradrenaline regulates courtship vocalization in quail, possibly via the α2A-adrenergic receptor expressed on ICo neurons.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Corte , Mesencéfalo/metabolismo , Norepinefrina/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Comportamento Sexual Animal/fisiologia , Vocalização Animal/fisiologia , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Clonidina/farmacologia , Coturnix , Masculino , Mesencéfalo/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Vocalização Animal/efeitos dos fármacos
5.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34298984

RESUMO

While much of biomedical research since the middle of the twentieth century has focused on molecular pathways inside the cell, there is increasing evidence that extracellular signaling pathways are also critically important in health and disease. The neuromodulators norepinephrine (NE), serotonin (5-hydroxytryptamine, 5HT), dopamine (DA), acetylcholine (ACH), and melatonin (MT) are extracellular signaling molecules that are distributed throughout the brain and modulate many disease processes. The effects of these five neuromodulators on Alzheimer's disease (AD) are briefly examined in this paper, and it is hypothesized that each of the five molecules has a u-shaped (or Janus-faced) dose-response curve, wherein too little or too much signaling is pathological in AD and possibly other diseases. In particular it is suggested that NE is largely functionally opposed to 5HT, ACH, MT, and possibly DA in AD. In this scenario, physiological "balance" between the noradrenergic tone and that of the other three or four modulators is most healthy. If NE is largely functionally opposed to other prominent neuromodulators in AD, this may suggest novel combinations of pharmacological agents to counteract this disease. It is also suggested that the majority of cases of AD and possibly other diseases involve an excess of noradrenergic tone and a collective deficit of the other four modulators.


Assuntos
Doença de Alzheimer/fisiopatologia , Neurotransmissores/antagonistas & inibidores , Norepinefrina/fisiologia , Transmissão Sináptica/fisiologia , Adrenérgicos/administração & dosagem , Adrenérgicos/uso terapêutico , Neurônios Adrenérgicos/fisiologia , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Animais , Química Encefálica , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Melatonina/uso terapêutico , Camundongos , Modelos Neurológicos , Neurotransmissores/fisiologia , Nootrópicos/farmacologia , Nootrópicos/uso terapêutico , Norepinefrina/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Ratos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Proteínas tau/metabolismo
6.
Biol Pharm Bull ; 43(8): 1226-1234, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32741943

RESUMO

Attention deficit/hyperactivity disorder (AD/HD) is a neurodevelopmental disorder characterized by inattention, hyperactivity, and impulsivity. In patients with AD/HD, a decrease in the total and rapid eye movement (REM) sleep times has been observed. We have previously reported that mice with REM sleep deprivation-induced stress (REMSD) may show the hyperactivity- and inattention-like symptoms of AD/HD. However, in this model, impulsivity has not yet been investigated. Impulsivity and anxiety-related behaviors are evaluated by the elevated plus maze test (EPM). In this study, we investigated whether REMSD causes changes in the EPM and expression of alpha2A-adrenoceptors in the hippocampus and frontal cortex in a mouse model. Mice were deprived of REM sleep intermittently using the small-platform method (20 h/d) for 3 d. The time spent in the open arm and the expression levels of alpha2A-adrenoceptor in the hippocampus were significantly increased and decreased, respectively, by the REMSD. The time spent in the open arm was significantly limited by oxymetazoline (an alpha2A-adrenoceptor agonist), methylphenidate, and atomoxetine, which are clinically used to treat AD/HD. Moreover, the positive effects of oxymetazoline were attenuated by yohimbine and BRL44408, which are selective alpha2- and alpha2A-adrenoceptor antagonists, respectively. These results suggest that the increase in the time spent in the open arm induced by REMSD may serve as a model of impulsivity in AD/HD. Furthermore, the REMSD eliciting impulsivity-like behavior and the low-levels of anxiety may be linked to alpha2A-adrenoceptor signaling, as indicated by a decrease in alpha2A-adrenoceptor signaling, particularly in the mouse hippocampus.


Assuntos
Ansiedade/etiologia , Transtorno do Deficit de Atenção com Hiperatividade/etiologia , Modelos Animais de Doenças , Hipocampo/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Privação do Sono/complicações , Sono REM/fisiologia , Animais , Teste de Labirinto em Cruz Elevado , Comportamento Impulsivo/efeitos dos fármacos , Masculino , Camundongos , Receptores Adrenérgicos alfa 2/análise
7.
Clin Sci (Lond) ; 134(14): 1957-1971, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32643759

RESUMO

Acute respiratory distress syndrome (ARDS) is a severe condition with high morbidity and mortality and few interventions. The role of sympathetic stress in the pathogenesis of ARDS has attracted recent research attention. Blockade of α-2 or α2A-adrenoceptor (α2A-AR) has been shown to attenuate lung injury induced by lipopolysaccharide (LPS) in rats. However, the mechanism is unclear. We confirmed the role of α2A-AR in ARDS using knockout mice and alveolar macrophages following LPS stimulation to assess the underlying mechanisms. We found that α2A-AR deficiency decreased the permeability of the alveolar capillary barrier in ARDS mice and suppressed lung inflammation by reducing inflammatory cell infiltration and the production of TNF-α, interleukin (IL)-6, and CXCL2/MIP-2. LPS stimulation decreased NF-κB activation in lung tissues of α2A-AR deficient mice and increased norepinephrine concentrations. In vitro, we found that norepinephrine inhibited the production of TNF-α, IL-6, and CXCL2/MIP-2 and promoted the secretion of IL-10 from LPS-stimulated murine alveolar macrophages. Blockade of α2A-AR by a specific antagonist further inhibited the production of TNF-α, IL-6, and IL-10. Furthermore, norepinephrine down-regulated NF-κB activation in stimulated alveolar macrophages. Altogether, these results suggest that α2A-AR deficiency ameliorates lung injury by increasing norepinephrine concentrations in lung tissues and inhibiting the activation of alveolar macrophages.


Assuntos
Pulmão/metabolismo , Macrófagos Alveolares/fisiologia , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 2/fisiologia , Síndrome do Desconforto Respiratório/imunologia , Animais , Permeabilidade Capilar , Linhagem Celular , Modelos Animais de Doenças , Lipopolissacarídeos , Pulmão/imunologia , Ativação de Macrófagos , Masculino , Camundongos Knockout , Infiltração de Neutrófilos , Síndrome do Desconforto Respiratório/metabolismo
8.
Exp Brain Res ; 238(5): 1293-1303, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32322928

RESUMO

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


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Fenômenos Eletrofisiológicos/fisiologia , Fibras Nervosas Mielinizadas/fisiologia , Neurônios Aferentes/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Corno Dorsal da Medula Espinal/fisiologia , Transmissão Sináptica/fisiologia , Animais , Animais Recém-Nascidos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Vias Neurais/fisiologia , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
9.
Drug Alcohol Depend ; 209: 107946, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32145665

RESUMO

BACKGROUND AND PURPOSE: Kratom is a coffee-like plant containing compounds that cause opioid and stimulant effects. The most prevalent bioactive alkaloid of kratom is mitragynine (MG). Opioid effects of MG are apparent (e.g. antinociception and nanomolar affinity for µ, κ and δ opioid receptors), but effects encompassing interactions with additional systems, such as adrenergic and dopaminergic, remain undefined. Given that enhanced adrenergic transmission is a mechanism common to most first-line neuropathic pain medications, we tested the hypothesis that MG reduces chemotherapy-induced neuropathic pain through a mechanism involving α-adrenoceptor activation. METHODS: Rats were injected once with oxaliplatin (6 mg/kg IP) to induce allodynia and then treated with MG (0, 1, 5, 10 mg/kg IP) for 5-7 days. To investigate receptor mechanisms, a fixed dose of MG (5 mg/kg IP) was injected with yohimbine (5 mg/kg IP, α2-adrenoceptor antagonist), prazosin (5 mg/kg IP, α1-adrenoceptor antagonist), or naltrexone (5 mg/kg IP, opioid antagonist). KEY RESULTS: MG (5, 10 mg/kg) dose-dependently reduced mechanical sensitivity in oxaliplatin-injected rats. Anti-allodynic effects of MG were completely inhibited by yohimbine, and significantly reduced by prazosin and naltrexone. MG produced modest hyperlocomotion but only at a dose (30 mg/kg) higher than those required to reduce allodynia. CONCLUSION AND IMPLICATION: The finding that MG reduced neuropathic pain through a mechanism requiring active α-adrenoceptors indicates that the pharmacological profile of MG includes activation of adrenergic, as well as opioid, systems.


Assuntos
Antineoplásicos/toxicidade , Mitragyna , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Receptores Adrenérgicos alfa 2/fisiologia , Alcaloides de Triptamina e Secologanina/uso terapêutico , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Relação Dose-Resposta a Droga , Masculino , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oxaliplatina/toxicidade , Ratos , Ratos Sprague-Dawley , Alcaloides de Triptamina e Secologanina/farmacologia , Ioimbina/farmacologia
10.
J Am Soc Nephrol ; 31(4): 783-798, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32086277

RESUMO

BACKGROUND: Increased nerve activity causes hypertension and kidney disease. Recent studies suggest that renal denervation reduces BP in patients with hypertension. Renal NE release is regulated by prejunctional α2A-adrenoceptors on sympathetic nerves, and α2A-adrenoceptors act as autoreceptors by binding endogenous NE to inhibit its own release. However, the role of α2A-adrenoceptors in the pathogenesis of hypertensive kidney disease is unknown. METHODS: We investigated effects of α2A-adrenoceptor-regulated renal NE release on the development of angiotensin II-dependent hypertension and kidney disease. In uninephrectomized wild-type and α2A-adrenoceptor-knockout mice, we induced hypertensive kidney disease by infusing AngII for 28 days. RESULTS: Urinary NE excretion and BP did not differ between normotensive α2A-adrenoceptor-knockout mice and wild-type mice at baseline. However, NE excretion increased during AngII treatment, with the knockout mice displaying NE levels that were significantly higher than those of wild-type mice. Accordingly, the α2A-adrenoceptor-knockout mice exhibited a systolic BP increase, which was about 40 mm Hg higher than that found in wild-type mice, and more extensive kidney damage. In isolated kidneys, AngII-enhanced renal nerve stimulation induced NE release and pressor responses to a greater extent in kidneys from α2A-adrenoceptor-knockout mice. Activation of specific sodium transporters accompanied the exaggerated hypertensive BP response in α2A-adrenoceptor-deficient kidneys. These effects depend on renal nerves, as demonstrated by reduced severity of AngII-mediated hypertension and improved kidney function observed in α2A-adrenoceptor-knockout mice after renal denervation. CONCLUSIONS: Our findings reveal a protective role of prejunctional inhibitory α2A-adrenoceptors in pathophysiologic conditions with an activated renin-angiotensin system, such as hypertensive kidney disease, and support the concept of sympatholytic therapy as a treatment.


Assuntos
Hipertensão Renal/etiologia , Hipertensão Renal/prevenção & controle , Nefrite/etiologia , Nefrite/prevenção & controle , Receptores Adrenérgicos alfa 2/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Transmissão Sináptica/fisiologia , Angiotensina II , Animais , Modelos Animais de Doenças , Hipertensão Renal/fisiopatologia , Camundongos , Camundongos Knockout , Nefrite/fisiopatologia , Simpatectomia
11.
Pharmacol Biochem Behav ; 188: 172835, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31805289

RESUMO

Patients with anxiety disorders and posttraumatic stress disorder (PTSD) exhibit exaggerated fear responses and noradrenergic dysregulation. Fear-related responses to α2-adrenergic challenge were therefore studied in DxH C3H/HeJ-like recombinant inbred (C3HLRI) mice, which are a DBA/2J-congenic strain selectively bred for a high fear-sensitized startle (H-FSS). C3HLRI mice showed an enhanced acoustic startle response and immobility in the forced swim test compared to DBA/2J controls. The α2-adrenoceptor antagonist yohimbine (Yoh; 5.0 mg/kg) induced an anxiogenic and the α2-adrenoceptor agonist clonidine (Clon; 0.1 mg/kg) an anxiolytic effect in the open field (OF) in C3HLRI but not DBA/2J mice. In auditory fear-conditioning, Yoh (5.0 mg/kg)-treated C3HLRI mice showed higher freezing during fear recall and extinction learning than DBA/2J mice, and a higher ceiling for the Yoh-induced deficit in fear extinction. No strain differences were observed in exploration-related anxiety/spatial learning or the Clon-induced (0.1 mg/kg) corticosterone surge. A global analysis of the behavioral profile of the two mouse strains based on observed and expected numbers of significant behavioral outcomes indicated that C3HLRI mice showed significantly more often fear- and stress-related PTSD-like behaviors than DBA/2J controls. The analysis of the robustness of significant outcomes based on false discovery rate (FDR) thresholds confirmed significant differences for the strain-Yoh-interactions in the OF center and periphery, the Yoh-induced general extinction deficit, strain differences in conditioned fear levels, and at the dose of 5.0 mg/kg for the Yoh-induced ceiling in freezing levels among others. The current findings are consistent with previous observations showing alterations in the central noradrenergic system of C3HLRI mice (Browne et al., 2014, Stress 17:471-83). Based on their behavioral profile and response to α2-adrenergic stimulation, C3HLRI mice are a valuable genetic model for studying adrenergic mechanisms of anxiety disorders and potentially also of PTSD.


Assuntos
Estimulação Acústica/métodos , Antagonistas de Receptores Adrenérgicos alfa 2/toxicidade , Medo/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Reflexo de Sobressalto/fisiologia , Estimulação Acústica/efeitos adversos , Animais , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/efeitos dos fármacos , Medo/psicologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos DBA , Reflexo de Sobressalto/efeitos dos fármacos , Especificidade da Espécie , Ioimbina/toxicidade
12.
BMC Pharmacol Toxicol ; 20(1): 70, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31753018

RESUMO

BACKGROUND: Dexmedetomidine was proved to mitigate bupivacaine-induced cardiotoxicity but mechanism of this ability is still unclear. This study was designed to investigate the direct effects of dexmedetomidine on cardiotoxicity induced by bupivacaine on Langendorff rat heart preparation and the role of alpha 2 adrenoceptors in this process was explored. METHODS: Hearts of rat were isolated, mounted on a Langendorff system. Five experimental groups were assessed after 10 min Krebs-Henseleit buffer (KHB) infusions as follow: (1) Group Con, only KHB was perfused; (2) Group Dex, KHB was perfused for 5 min, then dexmedetomidine (10 nmol/L) was added; (3) Group Bupi, KHB was perfused for 25 min, then bupivacaine (50 µmol/L) was added; (4) Group Bupi + Dex, KHB was perfused for 5 min, then the dexmedetomidine (10 nmol/L) was added for 20 min, at last a mixture of KHB + dexmedetomidine + bupivacaine were perfused; (5) Group Bupi + Dex + Yoh, a combination of KHB + yohimbine (alpha 2 adrenoceptor antagonists, 1 µmol/L) was perfusion for 5 min, then dexmedetomidine (10 nmol/L) was added for 20 min, at last a mixture of KHB + yohimbine + dexmedetomidine + bupivacaine was perfused. The experimental perfusion was maintained for 35 min in group Con and group Dex, and the experimental perfusion was sustained until asystole in the other three groups. RESULTS: Compared with group Bupi, dexmedetomidine significantly increased the time to first arrhythmia (P <  0.001) and time to asystole (P <  0.001) in group Bupi + Dex. In addition, dexmedetomidine also significantly increased the time to 25, 50 and 75% reductions in heart rate (P <  0.001) and the time to 25, 50 and 75% reductions in rate-pressure product (P <  0.001) in group Bupi + Dex. Dexmedetomidine increased the cardiac tissue bupivacaine content when asystole (Bupi + Dex vs. Bupi, 58.5 ± 6.3 vs. 46.8 ± 5.6 nmol/g, P = 0.003). The benefit of dexmedetomidine on bupivacaine-induced cardiotoxicity were not eliminated by yohimbine. CONCLUSIONS: Dexmedetomidine could delay the occurrence of bupivacaine-induced arrhythmia and asystole in the isolated rat hearts, but the alpha 2 adrenoceptors were not involved in this process.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Analgésicos não Narcóticos/farmacologia , Anestésicos Locais/toxicidade , Bupivacaína/toxicidade , Dexmedetomidina/farmacologia , Tolerância a Medicamentos , Coração/efeitos dos fármacos , Animais , Cardiotoxicidade/fisiopatologia , Cardiotoxicidade/prevenção & controle , Coração/fisiologia , Técnicas In Vitro , Masculino , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/fisiologia
13.
Bull Exp Biol Med ; 167(5): 603-605, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31606802

RESUMO

The study examined the effect of clonidine hydrochloride (10-9-10-5 М) on electrical activity of working cardiomyocytes in rat right atrium. Stimulation of α2-adrenergic receptor with clonidine changed electrical activity of the heart. All tested concentrations of the agonist lengthened the action potential and decreased the firing rate of cardiomyocytes in a dose-dependent manner. The maximum effects of clonidine were observed at concentration of 10-5 М.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Clonidina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais não Endogâmicos , Relação Dose-Resposta a Droga , Coração/efeitos dos fármacos , Coração/fisiologia , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Ratos , Técnicas de Cultura de Tecidos
14.
Eur J Neurosci ; 50(11): 3772-3785, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31430399

RESUMO

The locus coeruleus (LC) contains the majority of central noradrenergic neurons sending wide projections throughout the entire CNS. The LC is considered to be essential for multiple key brain functions including arousal, attention and adaptive stress responses as well as higher cognitive functions and memory. Electrophysiological studies of LC neurons have identified several characteristic functional features such as low-frequency pacemaker activity with broad action potentials, transient high-frequency burst discharges in response to salient stimuli and an apparently homogeneous inhibition of firing by activation of somatodendritic α2 autoreceptors (α2AR). While stress-mediated plasticity of the α2AR response has been described, it is currently unclear whether different LC neurons projecting to distinct axonal targets display differences in α2AR function. Using fluorescent beads-mediated retrograde tracing in adult C57Bl6/N mice, we compared the anatomical distributions and functional in vitro properties of identified LC neurons projecting either to medial prefrontal cortex, hippocampus or cerebellum. The functional in vitro analysis of LC neurons confirmed their mostly uniform functional properties regarding action potential generation and pacemaker firing. However, we identified significant differences in tonic and evoked α2AR-mediated responses. While hippocampal-projecting LC neurons were partially inhibited by endogenous levels of norepinephrine and almost completely silenced by application of saturating concentrations of the α2 agonist clonidine, prefrontal-projecting LC neurons were not affected by endogenous levels of norepinephrine and only partially inhibited by saturating concentrations of clonidine. Thus, we identified a limited α2AR control of electrical activity for prefrontal-projecting LC neurons indicative of functional heterogeneity in the LC-noradrenergic system.


Assuntos
Autorreceptores/fisiologia , Axônios/fisiologia , Dendritos/fisiologia , Locus Cerúleo/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Autorreceptores/antagonistas & inibidores , Axônios/química , Axônios/efeitos dos fármacos , Dendritos/química , Dendritos/efeitos dos fármacos , Locus Cerúleo/química , Locus Cerúleo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos
15.
Toxins (Basel) ; 11(7)2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31288453

RESUMO

Oxaliplatin is a chemotherapeutic agent used for metastatic colon and other advanced cancers. Most common side effect of oxaliplatin is peripheral neuropathy, manifested in mechanical and cold allodynia. Although the analgesic effect of bee venom has been proven to be effective against oxaliplatin-induced peripheral neuropathy, the effect of its major component; melittin has not been studied yet. Thus, in this study, we investigated whether melittin has an analgesic effect on oxaliplatin-induced allodynia. Intraperitoneal single injection of oxaliplatin (6 mg/kg) induced mechanical and cold allodynia, resulting in increased withdrawal behavior in response to von Frey filaments and acetone drop on hind paw. Subcutaneous melittin injection on acupoint ST36 (0.5 mg/kg) alleviated oxaliplatin-induced mechanical and cold allodynia. In electrophysiological study, using spinal in vivo extracellular recording, it was shown that oxaliplatin-induced hyperexcitation of spinal wide dynamic range neurons in response to peripheral stimuli, and melittin administration inhibited this neuronal activity. In behavioral assessment, analgesic effect of melittin was blocked by intrathecal α1- and α2- adrenergic receptor antagonists administration. Based on these results, we suggest that melittin could be used as an analgesic on oxaliplatin-induced peripheral neuropathy, and that its effect is mediated by activating the spinal α1- and α2-adrenergic receptors.


Assuntos
Analgésicos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Meliteno/uso terapêutico , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Analgésicos/farmacologia , Animais , Antineoplásicos , Temperatura Baixa , Hiperalgesia/induzido quimicamente , Idazoxano/farmacologia , Meliteno/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Oxaliplatina , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Prazosina/farmacologia , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiologia , Tato
16.
Neurochem Int ; 129: 104490, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31226280

RESUMO

Multiple sclerosis (MS) is a chronic autoimmune inflammatory disease of the central nervous system (CNS) affecting more than 2.5 million individuals worldwide. In the present study, myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) mice were treated with adenosine receptor A2A antagonist SCH58261 at different periods of EAE development. The administration of SCH58261 at 11-28 days post-immunization (d.p.i.) with MOG improved the neurological deficits. This time window corresponds to the therapeutic time window for MS treatment. SCH58261 significantly reduced the CNS neuroinflammation including reduced local infiltration of inflammatory cells, demyelination, and the numbers of macrophage/microglia in the spinal cord. Importantly, SCH58261 ameliorated the EAE-induced neurobehavioral deficits. By contrast, the SCH58261 treatment was ineffective when administered at the beginning of the onset of EAE (i.e., 1-10 d.p.i). The identification of the effective therapeutic window of A2A receptor antagonist provide insight into the role of A2A receptor signaling in EAE, and support SCH58261 as a candidate for the treatment of MS in human.


Assuntos
Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Encefalomielite Autoimune Experimental/prevenção & controle , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Pirimidinas/uso terapêutico , Receptores Adrenérgicos alfa 2/fisiologia , Triazóis/uso terapêutico , Antagonistas do Receptor A2 de Adenosina/administração & dosagem , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Regulação para Baixo/efeitos dos fármacos , Esquema de Medicação , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Interferon gama/biossíntese , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Bainha de Mielina/patologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/toxicidade , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Quadriplegia/etiologia , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Triazóis/administração & dosagem , Triazóis/farmacologia
17.
Neuroscience ; 406: 176-185, 2019 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-30872164

RESUMO

Low frequency stimulation (LFS) has anticonvulsant effect and may restore the ability of long-term potentiation (LTP) to the epileptic brain. The mechanisms of LFS have not been completely determined. Here, we showed that LTP induction was impaired following in vitro epileptiform activity (EA) in hippocampal slices, but application of LFS prevented this impairment. Then, we investigated the involvement of α-adrenergic receptors in this effect of LFS. EA was induced by increasing the extracellular K+ concentration to 12 mM and EPSPs were recorded from CA1 neurons in whole cell configuration. EA increased EPSP amplitude from 6.9 ±â€¯0.7 mV to 9.6 ±â€¯0.6 mV. For LTP induction, the Schaffer collaterals were stimulated by high frequency stimulation (HFS; two trains of 100 pulses, 100 Hz at the interval of 20 s). The application of HFS resulted in 40.9 ±â€¯2.3% increase in the amplitude of EPSPs. However, following EA, HFS could not produce any significant changes in EPSP amplitude. Administration of LFS (1 Hz, 900 pulses) to Schaffer collaterals at the beginning of EA restored LTP induction to the hippocampal slices and HFS increased the EPSPs amplitude up to 41.7 ±â€¯3.1% of baseline. When slices were perfused by prazosin (α1-adrenergic receptor antagonist; 10 µM) before and during LFS application, LFS improvement on LTP induction was reduced significantly. Perfusion of slices by yohimbine (α2-adrenergic receptor antagonist; 5 µM) had no effect on LFS action. Therefore, it may be concluded that following epileptiform activity, LFS can improve the impairment of LTP generation through α1, but not α2, adrenergic receptor activity.


Assuntos
Hipocampo/fisiologia , Plasticidade Neuronal/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Convulsões/fisiopatologia , Sinapses/fisiologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Estimulação Elétrica , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Convulsões/prevenção & controle , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
18.
Inflammopharmacology ; 27(6): 1155-1167, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30850920

RESUMO

Previous reports suggest flavonoids as potent analgesic compounds. Based on these observations, the present study investigated the antinociceptive action of flavonol, 3', 4'-dimethoxy flavonol, 6, 3'-dimethoxy flavonol, 7, 2'-dimethoxy flavonol, and 7, 3'-dimethoxy flavonol and the possible mechanisms involved in these effects. The antinociceptive effect of the investigated compounds in doses of 25, 50, 100, and 200 mg/kg was evaluated in male Swiss albino mice using the acetic acid test, formalin-induced nociception, and hot water tail immersion test. The role of opioid, tryptaminergic, adrenergic, dopaminergic, GABAergic, and K+ATP channels in producing the antinociceptive effect was also studied using appropriate interacting agents. Treatment with flavonol and dimethoxy flavonols resulted in a significant reduction in the number of abdominal constrictions in the acetic acid test, a significant inhibition of the paw-licking/biting response time in both the phases of formalin nociception and also a significant increase in mean reaction time in the hot water tail immersion test. These observations revealed the antinociceptive effect of dimethoxy flavonols. The role of opioid, serotonergic (5HT3), and dopaminergic system was identified in the antinociceptive effect of flavonol and all dimethoxy derivatives investigated. In addition, the role of GABAergic, K+ATP channel, and α-2 adrenergic mechanisms were also observed in the antinociceptive action of some of the investigated compounds. The present study identified the antinociceptive effect of flavonol and dimethoxy flavonols in mice acting through different neuronal pathways.


Assuntos
Flavonóis/farmacologia , Analgésicos/farmacologia , Animais , Bicuculina/farmacologia , Formaldeído/farmacologia , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Canais de Potássio/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Dopaminérgicos/fisiologia , Receptores 5-HT3 de Serotonina/fisiologia , Ioimbina/farmacologia
19.
Pharmacol Ther ; 197: 179-190, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30703415

RESUMO

α2-adrenoceptor (α2-AR) isoforms, abundant in sympathetic synapses and noradrenergic neurons of the central nervous system, are integral in the presynaptic feed-back loop mechanism that moderates norepinephrine surges. We recently identified that postsynaptic α2-ARs, found in the myocellular sarcolemma, also contribute to a muscle-delimited feedback control capable of attenuating mobilization of intracellular Ca2+ and myocardial contractility. This previously unrecognized α2-AR-dependent rheostat is able to counteract competing adrenergic receptor actions in cardiac muscle. Specifically, in ventricular myocytes, nitric oxide (NO) and cGMP are the intracellular messengers of α2-AR signal transduction pathways that gauge the kinase-phosphatase balance and manage cellular Ca2+ handling preventing catecholamine-induced Ca2+ overload. Moreover, α2-AR signaling counterbalances phospholipase C - PKC-dependent mechanisms underscoring a broader cardioprotective potential under sympathoadrenergic and angiotensinergic challenge. Recruitment of such tissue-specific features of α2-AR under sustained sympathoadrenergic drive may, in principle, be harnessed to mitigate or prevent cardiac malfunction. However, cardiovascular disease may compromise peripheral α2-AR signaling limiting pharmacological targeting of these receptors. Prospective cardiac-specific gene or cell-based therapeutic approaches aimed at repairing or improving stress-protective α2-AR signaling may offer an alternative towards enhanced preservation of cardiac muscle structure and function.


Assuntos
Coração/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Sarcolema/fisiologia , Animais , Retroalimentação Fisiológica , Cardiopatias/tratamento farmacológico , Cardiopatias/fisiopatologia , Humanos
20.
Addict Biol ; 24(3): 509-521, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29480583

RESUMO

Effects of stress on the reward system are well established in the literature. Although previous studies have revealed that stress can reinstate extinguished addictive behaviors related to cocaine, the effects of stress on the rewarding memory of cocaine are not fully understood. Here, we provide evidence that stress potentiates the expression of rewarding memory of cocaine via the activation of brainstem-reward circuitry using a cocaine-induced conditioned place preference (CPP) paradigm combined with restraint stress in rats. The rats exposed to 30-minute restraint stress immediately before posttest exhibited significantly larger CPP scores compared with non-stressed rats. Intra-laterodorsal tegmental nucleus (LDT) microinjection of a ß or α2 adrenoceptor antagonist attenuated the stress-induced enhancement of cocaine CPP. Consistent with this observation, intra-LDT microinjection of a ß or α2 adrenoceptor agonist before posttest increased cocaine CPP. Additionally, intra-ventral tegmental area (VTA) microinjection of antagonists for the muscarinic acetylcholine, nicotinic acetylcholine or glutamate receptors attenuated the stress-induced enhancement of cocaine CPP. Finally, intra-medial prefrontal cortex (mPFC) microinjection of a D1 receptor antagonist also reduced the stress-induced enhancement of cocaine CPP. These findings suggest a mechanism wherein the LDT is activated by noradrenergic input from the locus coeruleus, leading to the activation of VTA dopamine neurons via both cholinergic and glutamatergic transmission and the subsequent excitation of the mPFC to enhance the memory of cocaine-induced reward value.


Assuntos
Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Memória/efeitos dos fármacos , Recompensa , Neurônios Adrenérgicos/efeitos dos fármacos , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Tronco Encefálico/efeitos dos fármacos , Condicionamento Psicológico/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Idazoxano/análogos & derivados , Idazoxano/farmacologia , Masculino , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Adrenérgicos beta/fisiologia , Restrição Física , Estresse Psicológico/fisiopatologia , Tegmento Mesencefálico/efeitos dos fármacos , Timolol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA