Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Crit Care Med ; 43(9): e332-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25962080

RESUMO

OBJECTIVE: Preliminary experimental data suggest that selective ß1-blockers may improve ex vivo cardiac function in animal sepsis. Currently, the effects of esmolol on in vivo cardiac function and on vascular function are unknown. The present study was designed to examine the effects of the ß1-selective blocker esmolol on myocardial and vascular function in peritonitis-induced septic rats and to explore the inflammatory pathways involved in this process. DESIGN: Randomized animal study. SETTING: University research laboratory. SUBJECTS: Male Wistar rats. INTERVENTIONS: Four hours after cecal ligation and puncture, Wistar rats were randomly allocated to the following groups: control, esmolol, norepinephrine (started at 18 hr after the surgery), and esmolol (started at 4 hr after the surgery) + norepinephrine (started at 18 hr after the surgery). Assessment at 18 hours after surgery was focused on cardiac contractility and vascular ex vivo function. Cardiac and vascular protein expressions of nuclear factor κB and endothelial nitric oxide synthase/Akt/inducible nitric oxide synthase pathways were assessed by Western blotting. MEASUREMENTS AND MAIN RESULTS: When compared with sham-operated animals, cecal ligation and puncture animals developed hypotension, cardiac depression, and vascular hyporesponsiveness to vasopressor treatment. Esmolol infusion increased cardiac contractility and restored mesenteric vasoreactivity. This effect was associated with a decrease in nuclear factor κB activation, an increase in Akt and endothelial nitric oxide synthase phosphorylation, and a decrease in inducible nitric oxide synthase expression both at the cardiac and vessel level. Esmolol infusion was also associated with an up-regulation in α1-vascular adrenoreceptors. CONCLUSION: Adjunction of selective ß1-blockade to standard septic shock management enhances intrinsic cardiac contractility and vascular responsiveness to catecholamines. These protective cardiovascular effects are likely predominantly attributed to the anti-inflammatory effect of esmolol.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Propanolaminas/farmacologia , Choque Séptico/tratamento farmacológico , Choque Séptico/fisiopatologia , Vasoconstritores/farmacologia , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Hemodinâmica , Mediadores da Inflamação/metabolismo , Masculino , NF-kappa B/biossíntese , Óxido Nítrico Sintase Tipo III/metabolismo , Norepinefrina/farmacologia , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 1/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
2.
Neurochem Res ; 40(8): 1747-57, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26148532

RESUMO

Rapid eye movement sleep (REMS) modulates Na-K ATPase activity and maintains brain excitability. REMS deprivation (REMSD)-associated increased Na-K ATPase activity is mediated by noradrenaline (NA) acting on α1-adrenoceptor (AR) in the brain. It was shown that NA-induced increased Na-K ATPase activity was due to allosteric modulation as well as increased turnover of the enzyme. Although the former has been studied in detail, our understanding on the latter was lacking, which we have studied. Male Wistar rats were REMS deprived for 4-days by classical flower-pot method; suitable control experiments were conducted. In another set, α1-AR antagonist prazosin (PRZ) was i.p. injected 48 h REMSD onward. At the end of experiments rats were sacrificed by cervical dislocation and brains were removed. Synaptosomes prepared from the brains were used to estimate Na-K ATPase activity as well as protein expressions of different isoforms of the enzyme subunits using western blot. REMSD significantly increased synaptosomal Na-K ATPase activity and that was due to differential increase in the expressions of α1-, α2- and α3-isoforms, but not that of ß1- and ß2-isoforms. PRZ reduced the REMSD-induced increased Na-K ATPase activity and protein expressions. We also observed that the increased Na-K ATPase subunit expression was not due to enhanced mRNA synthesis, which suggests the possibility of post-transcriptional regulation. Thus, the findings suggest that REMSD-associated increased Na-K ATPase activity is due to elevated level of α-subunit of the enzyme and that is induced by NA acting on α1-AR mediated mRNA-stabilization.


Assuntos
Encéfalo/enzimologia , Norepinefrina/farmacologia , Subunidades Proteicas/biossíntese , Receptores Adrenérgicos alfa 1/biossíntese , Privação do Sono/enzimologia , Sono REM/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Masculino , Norepinefrina/fisiologia , Ratos , Ratos Wistar , Sono REM/efeitos dos fármacos
3.
Am J Physiol Renal Physiol ; 307(11): F1238-48, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25339698

RESUMO

The homeostatic control of blood pressure hinges upon the delicate balance between prohypertensinogenic and antihypertensinogenic systems. D1-like dopamine receptors [dopamine D1 and D5 receptors (D1Rs and D5Rs, respectively)] and the α1A-adrenergic receptor (α1A-AR) are expressed in the renal proximal tubule and engender opposing effects on Na(+) transport, i.e., natriuresis (via D1Rs and D5Rs) or antinatriuresis (via α1A-ARs). We tested the hypothesis that the D1R/D5R regulates the α1A-AR. D1-like dopamine receptors coimmunoprecipitated, colocalized, and cofractionated with α1A-ARs in lipid rafts in immortalized human renal proximal tubule cells. Long-term treatment with the D1R/D5R agonist fenoldopam resulted in decreased D1R and D5R expression but increased α1A-AR abundance in the plasma membrane. Short-term fenoldopam treatment stimulated the translocation of Na(+)-K(+)-ATPase from the plasma membrane to the cytosol that was partially reversed by an α1A-AR agonist, which by itself induced Na(+)-K(+)-ATPase translocation from the cytosol to the plasma membrane. The α1A-AR-specific agonist A610603 also minimized the ability of fenoldopam to inhibit Na(+)-K(+)-ATPase activity. To determine the interaction among D1Rs, D5Rs, and α1A-ARs in vivo, we used phenylephrine and A610603 to decrease Na(+) excretion in several D1-like dopamine receptor knockout mouse strains. Phenylephrine and A61603 treatment resulted in a partial reduction of urinary Na(+) excretion in wild-type mice and its abolition in D1R knockout, D5R knockout, and D1R-D5R double-knockout mice. Our results demonstrate the ability of the D1-like dopamine receptors to regulate the expression and activity of α1A-AR. Elucidating the intricacies of the interaction among these receptors is crucial for a better understanding of the crosstalk between anti- and pro-hypertensive systems.


Assuntos
Túbulos Renais Proximais/metabolismo , Receptores Adrenérgicos alfa 1/biossíntese , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/fisiologia , Animais , Biotinilação , Pressão Sanguínea/fisiologia , Linhagem Celular , Membrana Celular/metabolismo , Humanos , Túbulos Renais Proximais/citologia , Camundongos , Camundongos Knockout , Receptores de Dopamina D5/metabolismo , Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
4.
Am J Physiol Heart Circ Physiol ; 302(8): H1614-24, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22307672

RESUMO

We examined α(1A)-adrenergic receptor (AR) mediation of preconditioning in a novel α(1A)-AR cardiac transgenic (TG) rat model (α(1A)-TG). Compared with nontransgenic littermates (NTLs), in conscious α(1A)-TG rats, heart rate was reduced, contractility [left ventricle (LV) +dP/dt, ejection fraction, end-systolic elastance] was significantly enhanced, and triple product (LV systolic wall stress × LV +dP/dt × heart rate) was unchanged. However, infarct size (IS)/area at risk (AAR) in response to ischemia-reperfusion (30 min coronary occlusion/3 h reperfusion) was reduced to 35 ± 4.6% in α(1A)-TGs vs. 52 ± 2.2% in NTLs (P < 0.05). Second window preconditioning reduced IS/AAR in NTLs to 29 ± 2.7% but did not afford further protection in α(1A)-TGs. In contrast, with first window preconditioning, IS/AAR was reduced to similar levels in both α(1A)-TGs (12 ± 1.4%) and NTLs (10 ± 1.1%). In untreated α(1A)-TGs, cardioprotection was associated with enhanced myocardial phosphorylated (p)-mitogen/extracellular signal-regulated kinase (MEK), p-extracellular signal-regulated kinase (ERK), and inducible nitric oxide synthase (iNOS) at the protein level, along with a 1.3-fold increase in total nitric oxide synthase activity like in second window preconditioning. Affymetrix microarrays revealed that few genes (4.6% of 3,172 upregulated; 8.8% of 3,498 downregulated) showed directionally similar changes in α(1A)-TGs vs. NTLs subjected to second window preconditioning. Thus, second, but not first, window cardioprotection is evident in α(1A)-TGs in the absence of ischemic preconditioning and is mediated by iNOS activation associated with MEK/ERK phosphorylation. Transcriptionally, however, second window preconditioning is considerably more complex than α(1A)-TG preconditioning, with the alteration of thousands of additional genes affording no further protection than that already available in α(1A)-TG rats.


Assuntos
Precondicionamento Isquêmico Miocárdico , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos alfa 1/biossíntese , Animais , Apoptose/fisiologia , Ecocardiografia , Expressão Gênica/fisiologia , Frequência Cardíaca/fisiologia , Hemodinâmica/fisiologia , Marcação In Situ das Extremidades Cortadas , Masculino , Análise em Microsséries , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Contração Miocárdica/fisiologia , Infarto do Miocárdio/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/metabolismo , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/fisiologia , Consumo de Oxigênio/fisiologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Adrenérgicos alfa 1/genética
5.
Can J Physiol Pharmacol ; 90(1): 1-12, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22149309

RESUMO

We investigated the effect of inter-renal aortic coarctation on the function and expression of vascular α(1A)- and α(1D)-adrenoceptors and plasma angiotensin II (ATII) in rats. Male Wistar rats, either sham operated (SO), or with aortic coarctation for 7 (AC7) and 14 days (AC14) were used for agonist-induced pressor responses in vehicle (physiological saline)- and antagonist-treated anesthetized animals, immunoblot analysis (α(1A)- and α(1D)-adrenoceptor in aorta and caudal arteries), and immunoassay (plasma ATII). The α(1D)-adrenoceptor antagonist, BMY-7378 (BMY) blocked noradrenaline-induced responses in the order SO > AC7 â‰« AC14; in contrast, the α(1A)-adrenoceptor antagonist RS-100329 (RS), produced a marginal shift to the right of the dose-response curve to noradrenaline, along with a strong decrease of the maximum pressor effect in the order SO > AC7 = AC14. The potency of the α(1A)-adrenoceptor agonist A-61603 increased in rats with AC14, and responses were inhibited by RS in the order AC14 > AC7 > SO. In aorta, α(1D)-adrenoceptor protein increased in AC7 and decreased in AC14; α(1A)-adrenoreceptor protein increased in the caudal artery of AC7 and returned to control values in AC14. Plasma ATII increased in AC7 and AC14, compared with SO rats. These results suggest an early and direct relationship between ATII and α(1D)-adrenoreceptors in the development of hypertension in this experimental model.


Assuntos
Coartação Aórtica/metabolismo , Coartação Aórtica/fisiopatologia , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/fisiopatologia , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/fisiologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Angiotensina II/sangue , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/fisiologia , Coartação Aórtica/sangue , Coartação Aórtica/complicações , Relação Dose-Resposta a Droga , Hipertensão Renovascular/sangue , Hipertensão Renovascular/complicações , Imidazóis/farmacologia , Masculino , Norepinefrina/antagonistas & inibidores , Norepinefrina/farmacologia , Piperazinas/farmacologia , Ratos , Ratos Wistar , Tetra-Hidronaftalenos/farmacologia , Timina/farmacologia , Timina/fisiologia
6.
Genetika ; 48(11): 1328-35, 2012 Nov.
Artigo em Russo | MEDLINE | ID: mdl-23297488

RESUMO

Using a large amount of breeding material, the idea of D. K. Belyaev on the role of selection in the appearance of new behavioral and neuronal forms was confirmed. Experiments were performed using rats of the GC (genetics + catatonia) strain, which are prone to passive defensive reactions of cataleptic freezing. At the current breeding stage, elevation of the proportion of so-called nervous animals was demonstrated, both with respect to the expression of such reactions and their frequency. At this breeding stage, in the brains of GC rats, the mRNA levels of alpha1A- and alpha2A-adrenoreceptor genes were determined. A decrease of alpha1A-adrenoreceptor gene expression in the midbrain and medulla oblongata, along with elevation of alpha2A-adrenoreceptor gene expression in the frontal cortex was observed. It was suggested that changes in the expression of alpha-adrenoreceptor genes could be caused by an increase in the proportion of nervous animals and could contribute to the akinetic behavioral component in GC rats.


Assuntos
Comportamento Animal , Encéfalo/metabolismo , Catatonia/metabolismo , Regulação da Expressão Gênica , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 2/biossíntese , Animais , Encéfalo/fisiopatologia , Cruzamento , Catatonia/imunologia , Catatonia/fisiopatologia , Modelos Animais de Doenças , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 2/genética
7.
J Pharmacol Exp Ther ; 338(2): 648-57, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21571945

RESUMO

Catecholamines released from the sympathetic nervous system in response to stress or injury affect expression of inflammatory cytokines generated by immune cells. α(1)-Adrenergic receptors (ARs) are expressed on innate immune cell populations, but their subtype expression patterns and signaling characteristics are not well characterized. Primary human monocytes, a human monocytic cell line, and monocyte-derived macrophage cells were used to measure expression of the proinflammatory mediator interleukin (IL)-1ß responding to lipopolysaccharide (LPS) in the presence or absence of α(1)-AR activation. Based on our previous findings, we hypothesized that α(1)-AR stimulation on innate immune cells positively regulates LPS-initiated IL-1ß production. IL-1ß production in response to LPS was synergistically higher for both monocytes and macrophages in the presence of the selective α(1)-AR agonist (R)-(-)-phenylephrine hydrochloride (PE). This synergistic IL-1ß response could be blocked with a selective α(1)-AR antagonist as well as inhibitors of protein kinase C (PKC). Radioligand binding studies characterized a homogenous α(1B)-AR subtype population on monocytes, which changed to a heterogeneous receptor subtype expression pattern when differentiated to macrophages. Furthermore, increased p38 mitogen-activated protein kinase (MAPK) activation was observed only with concurrent PE and LPS stimulation, peaking after 120 and 30 min in monocytes and macrophages, respectively. Blocking the PKC/p38 MAPK signaling pathway in both innate immune cell types inhibited the synergistic IL-1ß increase observed with concurrent PE and LPS treatments. This study characterizes α(1)-AR subtype expression on both human monocyte and macrophage cells and illustrates a mechanism by which increased IL-1ß production can be modulated by α(1)-AR input.


Assuntos
Macrófagos/metabolismo , Monócitos/metabolismo , Subunidades Proteicas/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Receptor 4 Toll-Like/fisiologia , Regulação para Cima/imunologia , Adulto , Diferenciação Celular/imunologia , Linhagem Celular Transformada , Células Cultivadas , Humanos , Imunidade Inata , Mediadores da Inflamação/fisiologia , Interleucina-1beta/biossíntese , Interleucina-1beta/sangue , Lipopolissacarídeos/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Monócitos/imunologia , Monócitos/patologia , Subunidades Proteicas/biossíntese , Subunidades Proteicas/sangue , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/sangue , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/sangue
8.
Nat Med ; 6(12): 1388-94, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11100125

RESUMO

Progress toward elucidating the function of alpha1B-adrenergic receptors (alpha1BARs) in the central nervous system has been constrained by a lack of agonists and antagonists with adequate alpha1B-specificity. We have obviated this constraint by generating transgenic mice engineered to overexpress either wild-type or constitutively active alpha1BARs in tissues that normally express the receptor, including the brain. All transgenic lines showed granulovacular neurodegeneration, beginning in alpha1B-expressing domains of the brain and progressing with age to encompass all areas. The degeneration was apoptotic and did not occur in non-transgenic mice. Correspondingly, transgenic mice showed an age-progressive hindlimb disorder that was parkinsonian-like, as demonstrated by rescue of the dysfunction by 3, 4-dihydroxyphenylalanine and considerable dopaminergic-neuronal degeneration in the substantia nigra. Transgenic mice also had a grand mal seizure disorder accompanied by a corresponding dysplasia and neurodegeneration of the cerebral cortex. Both behavioral phenotypes (locomotor impairment and seizure) could be partially rescued with the alpha1AR antagonist terazosin, indicating that alpha1AR signaling participated directly in the pathology. Our results indicate that overstimulation of alpha1BAR leads to apoptotic neurodegeneration with a corresponding multiple system atrophy indicative of Shy-Drager syndrome, a disease whose etiology is unknown.


Assuntos
Apoptose , Atrofia/etiologia , Doenças Neurodegenerativas/etiologia , Receptores Adrenérgicos alfa 1/biossíntese , Fatores Etários , Animais , Córtex Cerebral/patologia , Membro Posterior/patologia , Camundongos , Camundongos Transgênicos , Doença de Parkinson/etiologia , Fenótipo , Receptores Adrenérgicos alfa 1/genética , Convulsões/etiologia , Substância Negra/patologia
9.
Biomed Pharmacother ; 141: 111941, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34328102

RESUMO

BACKGROUND: Hypertension is a leading risk factor for developing kidney disease. Current single-target antihypertensive drugs are not effective for hypertensive nephropathy, in part due to its less understood mechanism of pathogenesis. We recently showed that QiShenYiQi (QSYQ), a component-based cardiovascular Chinese medicine, is also effective for ischemic stroke. Given the important role of the brain-heart-kidney axis in blood pressure control, we hypothesized that QSYQ may contribute to blood pressure regulation and kidney protection in Dahl salt-sensitive hypertensive rats. METHODS: The therapeutic effects of QSYQ on blood pressure and kidney injury in Dahl salt-sensitive rats fed with high salt for 9 weeks were evaluated by tail-cuff blood pressure monitoring, renal histopathological examination and biochemical indicators in urine and serum. RNA-seq was conducted to identify QSYQ regulated genes in hypertensive kidney, and RT-qPCR, immunohistochemistry, and Western blotting analysis were performed to verify the transcriptomics results and validate the purposed mechanisms. RESULTS: QSYQ treatment significantly decreased blood pressure in Dahl salt-sensitive hypertensive rats, alleviated renal tissue damage, reduced renal interstitial fibrosis and collagen deposition, and improved renal physiological function. RNA-seq and subsequent bioinformatic analysis showed that the expression of ADRA1D and SIK1 genes were among the most prominently altered by QSYQ in salt-sensitive hypertensive rat kidney. RT-qPCR, immunohistochemistry and Western blotting results confirmed that the mRNA and protein expression levels of alpha-1D adrenergic receptor (ADRA1D) in the kidney tissue of the QSYQ-treated rats were markedly down-regulated, while the mRNA and protein levels of salt inducible kinase 1 (SIK1) were significantly increased. CONCLUSION: QSYQ not only lowered blood pressure, but also alleviated renal damage via reducing the expression of ADRA1D and increasing the expression of SIK1 in the kidney of Dahl salt-sensitive hypertensive rats.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/metabolismo , Nefrite/tratamento farmacológico , Nefrite/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Receptores Adrenérgicos alfa 1/biossíntese , Cloreto de Sódio na Dieta/toxicidade , Animais , Medicamentos de Ervas Chinesas/farmacologia , Expressão Gênica , Masculino , Ratos , Ratos Endogâmicos Dahl
10.
Mol Brain ; 14(1): 55, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33726812

RESUMO

Our previous study showed the intrinsic ability of descending noradrenergic neurons projecting from the locus coeruleus to the spinal dorsal horn (SDH) to suppress itch-related behaviors. Noradrenaline and α1A-adrenaline receptor (α1A-AR) agonist increase inhibitory synaptic inputs onto SDH interneurons expressing gastrin-releasing peptide receptors, which are essential for itch transmission. However, the contribution of α1A-ARs expressed in SDH inhibitory interneurons to itch-related behavior remains to be determined. In this study, RNAscope in situ hybridization revealed that Adra1a mRNA is expressed in SDH inhibitory interneurons that are positive for Slc32a1 mRNA (known as vesicular GABA transporter). Mice with conditional knock-out of α1A-ARs in inhibitory interneurons (Vgat-Cre;Adra1aflox/flox mice) exhibited an increase in scratching behavior when induced by an intradermal injection of chloroquine, but not compound 48/80, which are known as models of histamine-independent and dependent itch, respectively. Furthermore, knockout of inhibitory neuronal α1A-ARs in the SDH using the CRISPR-Cas9 system also increased the scratching behavior elicited by chloroquine but not compound 48/80. Our findings demonstrated for the first time that α1A-ARs in SDH inhibitory interneurons contribute to the regulation of itch signaling with preference for histamine-independent itch.


Assuntos
Cloroquina/toxicidade , Interneurônios/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células do Corno Posterior/fisiologia , Prurido/fisiopatologia , Receptores Adrenérgicos alfa 1/fisiologia , Animais , Sistemas CRISPR-Cas , Feminino , Edição de Genes , Técnicas de Inativação de Genes , Masculino , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Prurido/induzido quimicamente , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/deficiência , Receptores Adrenérgicos alfa 1/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/biossíntese , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , p-Metoxi-N-metilfenetilamina/farmacologia
11.
World Neurosurg ; 152: e321-e331, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34062300

RESUMO

OBJECTIVE: Previous studies have shown that deep brain stimulation (DBS) can improve the level of consciousness of comatose patients with traumatic brain injuries (TBIs). However, the most suitable targets for DBS are unknown, and the mechanisms underlying recovery remain to be determined. The aim of the present study was to assess the effects of lateral hypothalamic area-DBS (LHA-DBS) in comatose rats with TBIs. METHODS: A total of 55 Sprague-Dawley rats were randomly assigned to 5 groups: the control group, TBI group, stimulated (TBI+LHA-DBS) group, antagonist (TBI+SB334867+LHA-DBS) group, and antagonist control (TBI+saline+LHA-DBS) group. The rats in the control group had undergone a sham operation and anesthesia, without coma induction. Coma was induced using a free-fall drop method. The rats in the stimulated group received bilateral LHA stimulation (frequency, 200 Hz; voltage, 2-4 V; pulse width, 0.1 ms) for 1 hour, with 5-minute intervals between subsequent stimulations, which were applied alternately to the left and right sides of the lateral hypothalamus. The comatose rats in the antagonist group received an intracerebroventricular injection with an orexins receptor type 1 (OX1R) antagonist (SB334867) and then received LHA-DBS. A I-VI consciousness scale and electroencephalography were used to assess the level of consciousness in each group of rats after LHA-DBS. Western blotting and immunofluorescence were used to detect OX1R expression in the LHA and α1-adrenoceptor (α1-AR) subtype and gamma-aminobutyric acid ß receptor (GABABR) expression in the prefrontal cortex. RESULTS: In the TBI, stimulated, antagonist, and antagonist control groups, 5, 10, 6, and 9 rats were awakened. The electroencephalographic readings indicated that the proportion of δ waves was lower in the stimulated group than in the TBI and antagonist groups (P < 0.05). Western blotting and immunofluorescence analysis showed that OX1R expression was greater in the stimulated group than in the TBI group (P < 0.05). The expression of α1-AR was also greater in the stimulated group than in the TBI and antagonist groups (P < 0.05). In contrast, the GABABR levels in the stimulated group were lower than those in the TBI and antagonist groups (P < 0.05). A statistically significant difference was found between the antagonist and antagonist control groups. CONCLUSIONS: Taken together, these results suggest that LHA-DBS promotes the recovery of consciousness in comatose rats with TBIs. Upregulation of α1-AR expression and downregulation of GABABR expression in the prefrontal cortex via the orexins and OX1R pathways might be involved in the wakefulness-promoting effects of LHA-DBS.


Assuntos
Lesões Encefálicas Traumáticas/psicologia , Lesões Encefálicas Traumáticas/cirurgia , Coma/psicologia , Coma/cirurgia , Estimulação Encefálica Profunda/métodos , Região Hipotalâmica Lateral/cirurgia , Orexinas/genética , Receptores Adrenérgicos alfa 1/biossíntese , Receptores de GABA/biossíntese , Transdução de Sinais/genética , Vigília , Anestesia , Animais , Benzoxazóis/farmacologia , Estado de Consciência/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Eletroencefalografia , Feminino , Lateralidade Funcional , Injeções Intraventriculares , Masculino , Naftiridinas/farmacologia , Receptores de Orexina/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ureia/análogos & derivados , Ureia/farmacologia
12.
FASEB J ; 23(10): 3564-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19564249

RESUMO

Activation of the heterotrimeric G protein Gq causes cardiomyocyte hypertrophy in vivo and in cell culture models. Hypertrophic responses induced by pressure or volume overload are exacerbated by increased Gq activity and ameliorated by Gq inhibition. Gq activates phospholipase Cbeta (PLCbeta) subtypes, resulting in generation of the intracellular messengers inositol(1,4,5)tris-phosphate [Ins(1,4,5)P(3)] and sn-1,2-diacylglycerol (DAG), which regulate intracellular Ca(2+) and conventional protein kinase C subtypes, respectively. Gq can also signal independently of PLCbeta, and the involvement of either Ins(1,4,5)P(3) or DAG in cardiomyocyte hypertrophy has not been unequivocally established. Overexpression of one splice variant of PLCbeta1, specifically PLCbeta1b, in neonatal rat cardiomyocytes causes increased cell size, elevated protein/DNA ratio, and heightened expression of the hypertrophy-related marker gene, atrial natriuretic peptide. The other splice variant, PLCbeta1a, had no effect. Expression of a 32-aa C-terminal PLCbeta1b peptide, which competes with PLCbeta1b for sarcolemmal association, prevented PLC activation and eliminated hypertrophic responses initiated by Gq or Gq-coupled alpha(1)-adrenergic receptors. In contrast, a PLCbeta1a C-terminal peptide altered neither PLC activity nor cellular hypertrophy. We conclude that hypertrophic responses initiated by Gq are mediated specifically by PLCbeta1b. Preventing PLCbeta1b association with the sarcolemma may provide a useful therapeutic target to limit hypertrophy.


Assuntos
Cardiomegalia/enzimologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/biossíntese , Miócitos Cardíacos/enzimologia , Fosfolipase C beta/biossíntese , Receptores Adrenérgicos alfa 1/biossíntese , Agonistas de Receptores Adrenérgicos alfa 1 , Animais , Cardiomegalia/patologia , Células Cultivadas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Miócitos Cardíacos/patologia , Fosfolipase C beta/genética , Ratos , Ratos Sprague-Dawley
13.
Investig Clin Urol ; 61(3): 297-303, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32377606

RESUMO

Purpose: Many patients with benign prostatic hyperplasia require treatment for persistent storage symptoms, even when the obstruction is successfully relieved by surgery. Previous studies identified a characteristic increase in α1D-adrenoceptor levels in the bladder in a bladder outlet obstruction (BOO) model. Here, we investigated the expression of α1-adrenoceptor subtypes in the bladder after relief of partial BOO (pBOO) in a rat model. Materials and Methods: A total of 60 female Sprague-Dawley rats were randomly divided into three groups (sham-operated, pBOO, and pBOO relief groups), and the expression of α1-adrenoceptor subtypes in the urothelium and detrusor muscle tissues was examined by western blot. Results: The expression of the α1D-adrenoceptor was significantly higher in the urothelium and detrusor muscle tissue of the pBOO and pBOO relief groups than in the corresponding tissue of the sham-operated group. Additionally, the α1A-adrenoceptor was predominant in the sham-operated group but significantly decreased in the urothelium in the pBOO group. No significant differences were found in α1A-adrenoceptor levels in detrusor muscle or whole bladder. Conclusions: Our results showed that α1D-adrenoceptor levels were consistently increased with pBOO, even after relief, suggesting that the α1D-adrenoceptor might be a cause of persistent storage symptoms after relief of pBOO.


Assuntos
Receptores Adrenérgicos alfa 1/biossíntese , Obstrução do Colo da Bexiga Urinária/metabolismo , Bexiga Urinária/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Obstrução do Colo da Bexiga Urinária/cirurgia
14.
J Pharmacol Exp Ther ; 330(3): 892-901, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19498105

RESUMO

2-((R-5-chloro-4-methoxymethyl-indan-1-yl)-1H-imidazole (PF-3774076) is a central nervous system (CNS) penetrant, potent, selective, partial agonist at the human alpha1(A)-adrenoceptor, demonstrating efficacy and selectivity in a range of binding and functional assays. In vivo, PF-3774076 increases peak urethral pressure in anesthetized female dogs in a dose-dependent manner, inducing changes in both the proximal and distal portions of the urethra via a central mechanism of action. The profile of this compound suggests that a CNS penetrant partial agonist at the alpha1(A)-adrenoceptor may offer significant benefit in stress urinary incontinence (SUI). However, despite partial agonism at the alpha1(A)-adrenoceptor and selectivity over alpha1(B)- and alpha1(D)-adrenoceptors, PF-3774076 did not offer the necessary degree of separation over cardiovascular events when assessed in in vivo models of cardiovascular function. This may be due to activation of both peripheral and central alpha1(A)-adrenoceptors. These data indicate that although central, partial alpha1(A)-agonists may offer significant benefit in the treatment of SUI, it may not be possible to achieve the desired level of urethral selectivity over cardiovascular events with this class of agent.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1 , Agonistas alfa-Adrenérgicos/farmacologia , Imidazóis/farmacologia , Uretra/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , DNA Complementar/biossíntese , DNA Complementar/genética , Cães , Feminino , Frequência Cardíaca/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Humanos , Ensaio Radioligante , Receptores Adrenérgicos alfa 1/biossíntese , Sulfonamidas/farmacologia , Telemetria , Uretra/metabolismo
15.
Epilepsia ; 50 Suppl 1: 59-64, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19125850

RESUMO

PURPOSE: The role of alpha 1b-adrenergic receptor (alpha 1b-AR) in relation with neuronal degeneration, drug addiction, and seizure susceptibility has recently emerged. In particular, mice that overexpress alpha 1b-AR undergo spontaneous epileptic seizures and progressive neuronal loss in a variety of brain areas. Therefore, one should expect that the blockade of alpha 1b-AR leads to anticonvulsant and neuroprotective effects. However, the lack of alpha 1b-AR antagonists does not allow testing of this hypothesis. METHODS: The development of alpha 1b-AR knockout (KO) mice led us to measure seizure susceptibility and neurodegeneration following systemic excitotoxins in these mice. RESULTS: We found that alpha 1b-AR KO mice are markedly resistant to kainate- and pilocarpine-induced seizures. Moreover, when marked seizure duration and severity are obtained by doubling the dose of chemoconvulsants in alpha 1b-AR KO, neuronal degeneration never occurs. CONCLUSIONS: These data indicate that alpha 1b-AR per se plays a fundamental role in the mechanisms responsible for seizure onset, severity, and duration, whereas the brain damage observed in alpha 1b-AR-overexpressing mice is likely to be a secondary phenomenon. In fact, the absence of alpha 1b-AR confers resistance to neurotoxicity induced by seizures/chemoconvulsants. These data, although confirming a pivotal role of alpha 1b-AR in modulating seizure threshold and neuronal death, offer a novel target, which may be used to develop novel anticonvulsants and neuroprotective agents.


Assuntos
Epilepsia/genética , Epilepsia/prevenção & controle , Predisposição Genética para Doença , Receptores Adrenérgicos alfa 1/deficiência , Receptores Adrenérgicos alfa 1/genética , Animais , Encéfalo/patologia , Epilepsia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidades Proteicas/biossíntese , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/fisiologia , Índice de Gravidade de Doença
16.
Toxicology ; 259(3): 97-106, 2009 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-19428949

RESUMO

The harmful effects of exposure to benzo[alpha]pyrene (B[alpha]P), which is a neurotoxic pollutant, on mammalian neurodevelopment and/or behaviour as yet remain widely unclear. In the present investigation, we evaluated the impact of the lactational exposure to B[alpha]P on postnatal development of pups and behaviour of young mice. The neurobiological effects of B[alpha]P during lactation were also evaluated on pups' brain. Here, we found that lactational exposure to B[alpha]P at 2 and 20mg/kg affects the neuromaturation of pups by significantly decreasing their reflex as highlighted in surface righting reflex and negative geotaxis tests. However, we noted a significant increase in muscular strength of lactationally B[alpha]P mg/kg-exposed pups, which was probably due to the impact of the exposure to this toxic compound on body weight gain. At the pup stage, lactational exposure to B[alpha]P also provoked a neurobiological change, which was assessed by determination of neuronal receptor gene expression. Indeed, a significant reduction in gene expression of 5HT(1A) receptors in pups exposed to B[alpha]P through lactation was found in comparison to controls. Additionally, attenuation in the expression of MOR(1) mRNA was observed, but statistically significant only in animals receiving the higher dose. Neither the expression levels of ADRA(1D) nor GABA(A) mRNA were altered. Interestingly, the harmful effects of lactational exposure to B[alpha]P on behaviour and cognitive function were still found despite a long post-weaning period. Young mice whose mothers were exposed to B[alpha]P displayed a disinhibition behaviour towards the aversive spaces of the elevated plus maze. Furthermore, a significant increase of spontaneous alternation in the Y-maze was observed, but only in young mice whose mothers were orally exposed to the lower dose of B[alpha]P. Our results suggest a close link between the neurobiological change highlighted in pups' brain and the different behavioural disturbances observed during postnatal development period until young adult stage.


Assuntos
Comportamento Animal/efeitos dos fármacos , Benzo(a)pireno/toxicidade , Encéfalo/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Exposição Materna/efeitos adversos , Animais , Animais Recém-Nascidos , Ansiedade/induzido quimicamente , Benzo(a)pireno/farmacocinética , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Poluentes Ambientais/farmacocinética , Feminino , Lactação , Masculino , Memória/efeitos dos fármacos , Camundongos , Atividade Motora/efeitos dos fármacos , Distribuição Aleatória , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/genética , Receptores de GABA-A/biossíntese , Receptores de GABA-A/genética , Receptores Opioides mu/biossíntese , Receptores Opioides mu/genética , Receptores de Serotonina/biossíntese , Receptores de Serotonina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Neurourol Urodyn ; 28(4): 349-55, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19034955

RESUMO

AIMS: To explore possible changes in expression and/or function of alpha(1)- and beta-adrenoceptor subtypes as a cause for bladder dysfunction in a rat model of bladder outlet obstruction (BOO). METHODS: BOO was induced in rats by partial urethral ligature. Contraction and relaxation experiments were performed with isolated bladder strips from BOO, sham-operated and non-operated (control) rats 7 days after BOO induction. mRNA expression of alpha(1)- and beta-adrenoceptor subtypes was assessed by quantitative real-time PCR. RESULTS: Receptor-independent contraction or relaxation did not differ between BOO and sham rats. The alpha(1)-agonists methoxamine and A-61,603 caused only weak contraction without major differences between groups. Against KCl-induced tone, the beta-adrenoceptor agonists noradrenaline and isoprenaline caused similar relaxation in BOO and sham rats, whereas relaxation in response to the beta(3)-selective BRL 37,344 was attenuated. Against passive tension, noradrenaline induced relaxation in sham and control rats; in contrast, noradrenaline induced contraction at low concentrations and relaxation at high concentrations in BOO rats. The contraction component was abolished by the alpha(1)-antagonist prazosin. The mRNA expression of alpha(1D)-adrenoceptors was increased in BOO, whereas none of the other receptor mRNAs were up-regulated. CONCLUSIONS: In a rat BOO model, weak contraction responses to alpha(1)-agonists and relaxation responses to beta-agonists are not altered to a major extent. Nevertheless, relaxation responses to the endogenous agonist noradrenaline are turned into alpha(1)-adrenoceptor-mediated contraction responses in BOO, possibly due to an up-regulation of alpha(1D)-adrenoceptors.


Assuntos
Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos beta/biossíntese , Receptores Adrenérgicos beta/fisiologia , Obstrução do Colo da Bexiga Urinária/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Ligadura , Masculino , Dados de Sequência Molecular , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Relaxamento Muscular/efeitos dos fármacos , Relaxamento Muscular/fisiologia , Norepinefrina/farmacologia , Cloreto de Potássio/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Análise de Regressão , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Pharmacology ; 84(6): 367-76, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19923872

RESUMO

Vessels from pregnant animals show a blunted response to adrenergic agonists. In this work, we explored if pregnancy reduces alpha(1)-adrenergic receptor (alpha(1)-AR)-mediated vascular smooth muscle protein expression as well as agonist-induced contraction in rat aorta, and if angiotensin II (Ang II) levels during pregnancy are related to these changes. Female Wistar rats were divided randomly into nonpregnant (NP) and pregnant groups (first week: P1, second week: P2, third week: P3). Subsets of animals were treated with captopril (5 mg kg(-1) day(-1) for 7 days; n = 6). Phenylephrine (PHE) concentration-response curves were constructed (1 x 10(-9) to 3.1 x 10(-5) mol/l) and alpha(1A)-, alpha(1B)- and alpha(1D)-AR were measured in the aorta by immunoblot. Captopril decreased alpha-AR expression in the NP group. In contrast, pregnancy decreased alpha(1A)-, alpha(1B)- and alpha(1D)-AR levels and captopril prevented this reduction. PHE sensitivity was decreased in the thoracic and abdominal aorta in the P2 group, with no changes in E(max ), and E(max) was decreased in the abdominal aorta in all experimental groups. Our results suggest that Ang II levels during pregnancy are related to a decrease in aortic alpha(1)-AR expression. The physiological meaning of this finding remains to be established.


Assuntos
Angiotensina II/fisiologia , Aorta Abdominal/metabolismo , Aorta Torácica/metabolismo , Gravidez , Receptores Adrenérgicos alfa 1/biossíntese , Resistência Vascular , Agonistas de Receptores Adrenérgicos alfa 1 , Antagonistas de Receptores Adrenérgicos alfa 1 , Agonistas alfa-Adrenérgicos/farmacologia , Angiotensina II/metabolismo , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Torácica/efeitos dos fármacos , Western Blotting , Captopril/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Técnicas In Vitro , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fenilefrina/farmacologia , Gravidez/metabolismo , Ratos , Ratos Wistar , Resistência Vascular/efeitos dos fármacos
19.
Neuron ; 103(4): 702-718.e5, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31227310

RESUMO

The locus coeruleus (LC) supplies norepinephrine (NE) to the entire forebrain and regulates many fundamental brain functions. Studies in humans have suggested that strong LC activation might shift network connectivity to favor salience processing. To causally test this hypothesis, we use a mouse model to study the effect of LC stimulation on large-scale functional connectivity by combining chemogenetic activation of the LC with resting-state fMRI, an approach we term "chemo-connectomics." We show that LC activation rapidly interrupts ongoing behavior and strongly increases brain-wide connectivity, with the most profound effects in the salience and amygdala networks. Functional connectivity changes strongly correlate with transcript levels of alpha-1 and beta-1 adrenergic receptors across the brain, and functional network connectivity correlates with NE turnover within select brain regions. We propose that these changes in large-scale network connectivity are critical for optimizing neural processing in the context of increased vigilance and threat detection.


Assuntos
Conectoma , Locus Cerúleo/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Animais , Ansiedade/fisiopatologia , Clozapina/farmacologia , Corpo Estriado/metabolismo , Drogas Desenhadas/farmacologia , Dopamina/metabolismo , Comportamento Exploratório/fisiologia , Neuroimagem Funcional , Genes fos , Locus Cerúleo/efeitos dos fármacos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Norepinefrina/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos beta 1/biossíntese , Receptores Adrenérgicos beta 1/genética , Receptores de Droga/fisiologia , Teste de Desempenho do Rota-Rod , Regulação para Cima/efeitos dos fármacos
20.
Biochem Biophys Res Commun ; 368(4): 887-92, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18279663

RESUMO

The cardiogenic capacity of embryonic stem (ES) cells has been well-investigated. However, little is known about the development of adrenoceptor (AR) systems during the process of ES cell differentiation, which are critically important in cardiac physiology and pharmacology. In this present study, we investigated the expression profile of adrenoceptor subtypes, beta-adrenergic modulation of muscarinic receptors and adrenoceptor-related signaling in cardiomyocytes derived from ES cells (ESCMs). Reverse transcription-polymerase chain reaction revealed that undifferentiated mouse ES cells expressed alpha(1A)-, alpha(1B)-, alpha(1D)- and beta(2)-AR mRNA. However, beta(1)-AR was only expressed after vitamin C induction. The expressions of alpha(1A)-, alpha(1D)- and beta(1)-ARs increased significantly while alpha(1B)- and beta(2)-ARs showed no significant change during the differentiation process. Furthermore, we detected the expression of tyrosine hydroxylase. Both alpha(1)-AR and beta-AR could activate extracellular responsive kinase in ESCMs. Isoprenaline could inhibit the expression of M(2) muscarinic receptor protein. CGP20712A, a beta(1)-AR antagonist, up-regulated the expression of M(2) muscarinic receptor while ICI118551, a beta(2)-AR antagonist, showed no effect. These results indicated that functional adrenoceptors and tyrosine hydroxylase, a critical enzyme in catecholamine biosynthesis, were differentially expressed in ESCMs. Adrenoceptor-related signaling pathways and beta-adrenergic modulation of muscarinic receptors were established during differentiation.


Assuntos
Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos alfa 1/biossíntese , Animais , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Imidazóis/farmacologia , Isoproterenol/farmacologia , Camundongos , Fenilefrina/farmacologia , Prazosina/farmacologia , Propanolaminas/farmacologia , Propranolol/farmacologia , Receptor Muscarínico M2/biossíntese , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Adrenérgicos beta/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA