Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
1.
Life Sci ; 293: 120304, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35016879

RESUMO

The present study aimed to investigate the role of α and ß-adrenergic receptors (ßARs) in mediation or modulation of the dexamethasone-induced nephrotoxicity by using different pharmacological interventions. Nephrotoxicity was induced by subcutaneous injection of dexamethasone (10 mg/kg) for 7 days in Wistar albino rats. Eight groups were used: control; dexamethasone; carvedilol; phenylephrine; carvedilol and phenylephrine; propranolol; doxazosin; propranolol and doxazosin. At the end of experiment, rats were euthanized and blood, urine and kidney samples were collected. Serum and urinary creatinine and urinary total protein levels were measured. Also, the renal tissue levels of diacylglycerol (DAG); Akt kinase activity, malondialdehyde (MDA), NADPH oxidase 2 (NOX2), transforming growth factor-ß (TGF-ß), Wnt3A and ß-catenin were recorded. Furthermore, histopathological and ß-arrestin2-immunohistochemical examinations of renal tissues were performed. Results: Dexamethasone induced glomerular damage, proteinuria, renal oxidative stress and upregulated the renal Wnt/ß-arrestin2/ß-catenin pathway and the profibrotic signals. Blocking the α1 and ßARs by carvedilol reduced the dexamethasone-induced nephrotoxicity. Pre-injection of phenylephrine did not reduce the reno-protective action of carvedilol. Blocking the ßARs only by propranolol reduced the dexamethasone-induced nephrotoxicity to the same extent of carvedilol group. Blocking the α1ARs only by doxazosin reduced dexamethasone-induced nephrotoxicity to a higher extent than other treatments. However, combined use of propranolol and doxazosin did not synergize the reno-protective effects of doxazosin. Conclusion: Dexamethasone induces nephrotoxicity, possibly, by upregulating the Wnt/ß-arrestin2/ß-catenin pathway. Blocking either α1ARs or ßARs can effectively protect against the dexamethasone-induced nephrotoxicity. However, combined blocking of α1ARs and ßARs does not synergize the reno-protective effects.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Dexametasona/toxicidade , Receptores Adrenérgicos/metabolismo , Via de Sinalização Wnt/fisiologia , beta-Arrestina 2/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapêutico , Animais , Anti-Inflamatórios/toxicidade , Carvedilol/farmacologia , Carvedilol/uso terapêutico , Masculino , Fenilefrina/farmacologia , Fenilefrina/uso terapêutico , Ratos , Ratos Wistar , Via de Sinalização Wnt/efeitos dos fármacos
2.
Nitric Oxide ; 118: 17-25, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34718145

RESUMO

AIMS: Patients with type 2 diabetes mellitus (T2DM) have reduced vasodilatory responses during exercise partially attributable to low nitric oxide (NO) levels. Low NO contributes to greater α-adrenergic mediated vasoconstriction in contracting skeletal muscle. We hypothesized boosting NO bioavailability via 8wks of active beetroot juice (BRA, 4.03 mmol nitrate, 0.29 mmol nitrite, n = 19) improves hyperemia, via reduced α-mediated vasoconstriction, during handgrip exercise relative to nitrate/nitrite-depleted beetroot juice (BRP, n = 18) in patients with T2DM. METHODS: Forearm blood flow (FBF) and vascular conductance (FVC) were calculated at rest and during handgrip exercise (20%max, 20contractions·min-1). Phenylephrine (α1-agonist) and dexmedetomidine (α2-agonist) were infused intra-arterially during independent trials to determine the influence of α-mediated vasoconstriction on exercise hyperemia. Vasoconstriction was quantified as the percent-reduction in FVC during α-agonist infusion, relative to pre-infusion, as well as the absolute change in %FVC during exercise relative to the respective rest trial (magnitude of sympatholysis). RESULTS: ΔFBF (156 ± 69 to 175 ± 73 ml min-1) and ΔFVC (130 ± 54 to 156 ± 63 ml min-1·100 mmHg-1, both P < 0.05) during exercise were augmented following BRA, but not BRP (P = 0.96 and 0.51). Phenylephrine-induced vasoconstriction during exercise was blunted following BRA (-17.1 ± 5.9 to -12.6 ± 3.1%, P < 0.01), but not BRP (P = 0.58) supplementation; the magnitude of sympatholysis was unchanged by either (beverage-by-time P = 0.15). BRA supplementation reduced dexmedetomidine-induced vasoconstriction during exercise (-23.3 ± 6.7 to -19.7 ± 5.2%) and improved the corresponding magnitude of sympatholysis (25.3 ± 11.4 to 34.4 ± 15.5%, both P < 0.05). CONCLUSIONS: BRA supplementation improves the hyperemic and vasodilatory responses to exercise in patients with T2DM which appears to be attributable to reduced α-adrenergic mediated vasoconstriction in contracting skeletal muscle.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Exercício Físico/fisiologia , Nitratos/farmacologia , Nitritos/farmacologia , Vasoconstrição/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Idoso , Beta vulgaris/química , Dexmedetomidina/farmacologia , Suplementos Nutricionais , Feminino , Sucos de Frutas e Vegetais , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fenilefrina/farmacologia , Raízes de Plantas/química
3.
Am J Physiol Heart Circ Physiol ; 321(5): H905-H919, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34506227

RESUMO

Alzheimer's disease (AD) has long been considered a brain-specific dementia syndrome. However, in recent decades, the occurrence of cardiovascular (CV) disease in the progression of AD has been confirmed by increasing epidemiological evidence. In this study, we conducted an in-depth cardiovascular characterization of a humanized amyloid precursor protein (APP) overexpressing mouse model (hAPP23+/-), which overexpresses the Swedish mutation (KM670/671NL). At the age of 6 mo, hAPP23+/- mice had a lower survival, lower body weight, and increased corticosterone and VMA levels compared with C57BL/6 littermates. Systolic blood pressure was increased in hAPP23+/- animals compared with C57BL/6 littermates, but diastolic blood pressure was not statistically different. Pulse pressure remained unchanged but abdominal and carotid pulse-wave velocity (aPWV and cPWV) were increased in hAPP23+/- compared with C57BL/6 mice. Echocardiography showed no differences in systolic or diastolic cardiac function. Ex vivo evaluation of vascular function showed decreased adreno receptor dependent vasoconstriction of hAPP23+/- aortic segments, although the isobaric biomechanics of the aortic wall were similar to C57BL/6 aortic segments. In conclusion, hAPP23+/- mice exhibited high serum corticosterone levels, elevated systolic blood pressure, and increased arterial stiffness in vivo. However, ex vivo aortic stiffness of hAPP23+/- aortic segments was not changed and vascular reactivity to α1-adrenoceptor stimulation was attenuated. These findings highlight the need for more frequent assessment of circulating stress hormone levels and PWV measurements in daily clinical practice for people at risk of AD.NEW & NOTEWORTHY We showed that male amyloid precursor protein (APP) transgenic mice have higher circulating stress hormone levels. As a result, higher systolic blood pressure and pulse-wave velocity were measured in vivo in addition to a smaller α-adrenergic receptor-dependent contraction upon ex vivo stimulation with phenylephrine. Our findings highlight the need for more frequent assessment of circulating stress hormone levels and PWV measurements in daily clinical practice for people at risk of Alzheimer's disease.


Assuntos
Doença de Alzheimer/sangue , Precursor de Proteína beta-Amiloide/metabolismo , Aorta Torácica/metabolismo , Pressão Arterial , Corticosterona/sangue , Rigidez Vascular , Vasoconstrição , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiopatologia , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Receptores Adrenérgicos alfa 1/metabolismo , Regulação para Cima , Vasoconstrição/efeitos dos fármacos
4.
Exp Eye Res ; 211: 108760, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34487726

RESUMO

Little is known about the relationship between stimulation of lacrimal gland (LG) tear protein secretion by parasympathetic versus sympathetic nerves, particularly whether the spectrum of tear proteins evoked through each innervation pathway varies. We have previously shown that activity and abundance of cathepsin S (CTSS), a cysteine protease, is greatly increased in tears of Sjögren's syndrome (SS) patients and in tears from the male NOD mouse of autoimmune dacryoadenitis that recapitulates SS-associated dry eye disease. Beyond the increased synthesis of CTSS detected in the diseased NOD mouse LG, increased tear CTSS secretion in NOD mouse tears was recently linked to increased exocytosis from a novel endolysosomal secretory pathway. Here, we have compared secretion and trafficking of CTSS in healthy mouse LG acinar cells stimulated with either the parasympathetic acetylcholine receptor agonist, carbachol (CCh), or the sympathetic α1-adrenergic agonist, phenylephrine (PE). In situ secretion studies show that PE significantly increases CTSS activity and protein in tears relative to CCh stimulation by 1.2-fold (***, p = 0.0009) and ∼5-fold (*, p-0.0319), respectively. A similar significant increase in CTSS activity with PE relative to CCh is observed when cultured LGAC are stimulated in vitro. CCh stimulation significantly elevates intracellular [Ca2+], an effect associated with increases in the size of Rab3D-enriched vesicles consistent with compound fusion, and subsequently decreases in their intensity of labeling consistent with their exocytosis. PE stimulation induces a lower [Ca2+] response and has minimal effects on Rab3D-enriched SV diameter or the intensity of Rab3D-enriched SV labeling. LG deficient in Rab3D exhibit a higher sensitivity to PE stimulation, and secrete more CTSS activity. Significant increases in the colocalization of endolysosomal vesicle markers (Lamp1, Lamp2, Rab7) with the subapical actin suggestive of fusion of endolysosomal vesicles at the apical membrane occur both with CCh and PE stimulation, but PE demonstrates increased colocalization. In conclusion, the α1-adrenergic agonist, PE, increases CTSS secretion into tears through a pathway independent of the exocytosis of Rab3D-enriched mature SV, possibly representing an alternative endolysosomal secretory pathway.


Assuntos
Células Acinares/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Catepsinas/metabolismo , Aparelho Lacrimal/efeitos dos fármacos , Fenilefrina/farmacologia , Via Secretória/efeitos dos fármacos , Lágrimas/metabolismo , Células Acinares/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Carbacol/farmacologia , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Modelos Animais de Doenças , Feminino , Inativação Gênica , Aparelho Lacrimal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , beta-N-Acetil-Hexosaminidases/metabolismo , Proteínas rab3 de Ligação ao GTP/genética
5.
Pharmacol Res Perspect ; 9(4): e00799, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34355529

RESUMO

Highly selective drugs offer a way to minimize side-effects. For agonist ligands, this could be through highly selective affinity or highly selective efficacy, but this requires careful measurements of intrinsic efficacy. The α1-adrenoceptors are important clinical targets, and α1-agonists are used to manage hypotension, sedation, attention deficit hypersensitivity disorder (ADHD), and nasal decongestion. With 100 years of drug development, there are many structurally different compounds with which to study agonist selectivity. This study examined 62 α-agonists at the three human α1-adrenoceptor (α1A, α1B, and α1D) stably expressed in CHO cells. Affinity was measured using whole-cell 3 H-prazosin binding, while functional responses were measured for calcium mobilization, ERK1/2-phosphorylation, and cAMP accumulation. Efficacy ratios were used to rank compounds in order of intrinsic efficacy. Adrenaline, noradrenaline, and phenylephrine were highly efficacious α1-agonists at all three receptor subtypes. A61603 was the most selective agonist and its very high α1A-selectivity was due to selective α1A-affinity (>660-fold). There was no evidence of Gq-calcium versus ERK-phosphorylation biased signaling at the α1A, α1B, or α1D-adrenoceptors. There was little evidence for α1A calcium versus cAMP biased signaling, although there were suggestions of calcium versus cAMP bias the α1B-adrenoceptor. Comparisons of the rank order of ligand intrinsic efficacy suggest little evidence for selective intrinsic efficacy between the compounds, with perhaps the exception of dobutamine which may have some α1D-selective efficacy. There seems plenty of scope to develop affinity selective and intrinsic efficacy selective drugs for the α1-adrenoceptors in future.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo , Animais , Células CHO , Cálcio/metabolismo , Cricetulus , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Ligantes , Fosforilação/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/genética
6.
Neuropharmacology ; 196: 108702, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34246685

RESUMO

A growing body of literature implicates noradrenergic (NE) signaling in the modulation of ethanol consumption. However, relatively few studies have detailed specific brain pathways that mediate NE-associated binge-like ethanol consumption. To begin to fill this gap in the literature, male and female C57BL6/J and TH-ires-cre mice underwent pharmacological and chemogenetic testing, respectively, in combination with "drinking in the dark" procedures to model binge-like consumption of ethanol or sucrose solutions. First, we showed that intraperitoneal administration of the NE reuptake inhibitor, reboxetine, blunted binge-like ethanol intake in C57BL6/J mice. Chemogenetic activation of locus coeruleus (LC) tyrosine hydroxylase (TH)-expressing neurons blunted binge-like ethanol intake regardless of sex. Chemogenetic activation of LC projections to the lateral hypothalamus (LH), a region implicated in ethanol consumption, blunted binge-like ethanol drinking without altering sucrose intake in ethanol-experienced or ethanol-naïve mice. In C57BL/6 J mice, LH-targeted microinfusion of an α1-adrenergic receptor (AR) agonist blunted binge-like ethanol intake across both sexes, while LH infusion of a ß-AR agonist blunted binge-like ethanol intake in females exclusively. Finally, in mice with high baseline ethanol intake both an α1- AR agonist and an α-2 AR antagonist blunted binge-like ethanol intake. The present results provide novel evidence that increased NE tone in a circuit arising from the LC and projecting to the LH reduces binge-like ethanol drinking in mice, and may represent a novel approach to treating binge or heavy drinking prior to the development of dependence. This article is part of the special Issue on "Neurocircuitry Modulating Drug and Alcohol Abuse".


Assuntos
Inibidores da Captação Adrenérgica/farmacologia , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Depressores do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Região Hipotalâmica Lateral/metabolismo , Locus Cerúleo/metabolismo , Norepinefrina/metabolismo , Reboxetina/farmacologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Feminino , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/fisiopatologia , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/fisiopatologia , Masculino , Camundongos , Vias Neurais , Tirosina 3-Mono-Oxigenase
7.
Biochem Pharmacol ; 188: 114560, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33844984

RESUMO

Emerging evidence suggests that G protein coupled receptor 55 (GPR55) may influence adrenoceptor function/activity in the cardiovascular system. Whether this reflects direct interaction (dimerization) between receptors or signalling crosstalk has not been investigated. This study explored the interaction between GPR55 and the alpha 1A-adrenoceptor (α1A-AR) in the cardiovascular system and the potential to influence function/signalling activities. GPR55 and α1A-AR mediated changes in both cardiac and vascular function was assessed in male wild-type (WT) and GPR55 homozygous knockout (GPR55-/-) mice by pressure volume loop analysis and isolated vessel myography, respectively. Dimerization of GPR55 with the α1A-AR was examined in transfected Chinese hamster ovary-K1 (CHO-K1) cells via Bioluminescence Resonance Energy Transfer (BRET). GPR55 and α1A-AR mediated signalling (extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation) was investigated in neonatal rat ventricular cardiomyocytes using AlphaScreen proximity assays. GPR55-/- mice exhibited both enhanced pressor and inotropic responses to A61603 (α1A-AR agonist), while in isolated vessels, A61603 induced vasoconstriction was attenuated by a GPR55-dependent mechanism. Conversely, GPR55-mediated vasorelaxation was not altered by pharmacological blockade of α1A-ARs with tamsulosin. While cellular studies demonstrated that GPR55 and α1A-AR failed to dimerize, pharmacological blockade of GPR55 altered α1A-AR mediated signalling and reduced ERK1/2 phosphorylation. Taken together, this study provides evidence that GPR55 and α1A-AR do not dimerize to form heteromers, but do interact at the signalling level to modulate the function of α1A-AR in the cardiovascular system.


Assuntos
Multimerização Proteica/fisiologia , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Receptores de Canabinoides/deficiência , Receptores de Canabinoides/genética , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Animais Recém-Nascidos , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Gravidez , Multimerização Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
8.
Eur J Pharmacol ; 900: 174055, 2021 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-33775645

RESUMO

In patients with obstructive jaundice, the cardiovascular system exhibits hypotension and vascular hyporeactivity. Most norepinephrine is taken up through the neuronal norepinephrine transporter (NET), which is implicated in cardiovascular diseases. A previous study demonstrated that pharmacological NET inhibition could increase resting blood pressure. However, the role of NETs in vascular hyporeactivity induced by obstructive jaundice is poorly understood. This study used the NET inhibitor nisoxetine and a rat model of bile duct ligation (BDL) to investigate whether NET is associated with BDL-induced vascular hyporeactivity. Rats were injected with nisoxetine via the tail vein for 7 consecutive days after BDL. Samples of the superior cervical sympathetic ganglion (SCG) and thoracic aortic rings were processed for investigations. Our results showed that NET expression in the SCG was significantly increased after BDL. Nisoxetine prevented the augmentation of NET expression, increased α1-adrenoceptor activation, and enhanced the weakened contractile responses of thoracic aortic rings after BDL. Our study demonstrates that nisoxetine plays a protective role in BDL-induced vascular hyporeactivity through increased α1-adrenoceptor activation in rats.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiopatologia , Catecolaminas , Icterícia Obstrutiva/tratamento farmacológico , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/antagonistas & inibidores , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Ductos Biliares , Bilirrubina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Gânglios Simpáticos/efeitos dos fármacos , Icterícia Obstrutiva/fisiopatologia , Ligadura , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley
9.
Am J Physiol Heart Circ Physiol ; 320(5): H2112-H2129, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33606584

RESUMO

Electronic cigarette (e-cig) vaping (ECV) has been proposed as a safer alternative to tobacco cigarette smoking (TCS); however, this remains controversial due to a lack of long-term comparative studies. Therefore, we developed a chronic mouse exposure model that mimics human vaping and allows comparison with TCS. Longitudinal studies were performed to evaluate alterations in cardiovascular function with TCS and ECV exposure durations of up to 60 wk. For ECV, e-cig liquid with box-mod were used and for TCS, 3R4F-cigarettes. C57/BL6 male mice were exposed 2 h/day, 5 days/wk to TCS, ECV, or air control. The role of vape nicotine levels was evaluated using e-cig-liquids with 0, 6, or 24 mg/mL nicotine. Following 16-wk exposure, increased constriction to phenylephrine and impaired endothelium-dependent and endothelium-independent vasodilation were observed in aortic segents, paralleling the onset of systemic hypertension, with elevations in systemic vascular resistance. Following 32 wk, TCS and ECV induced cardiac hypertrophy. All of these abnormalities further increased out to 60 wk of exposure, with elevated heart weight and aortic thickness along with increased superoxide production in vessels and cardiac tissues of both ECV and TCS mice. While ECV-induced abnormalities were seen in the absence of nicotine, these occurred earlier and were more severe with higher nicotine exposure. Thus, long-term vaping of e-cig can induce cardiovascular disease similar to TCS, and the severity of this toxicity increases with exposure duration and vape nicotine content.NEW & NOTEWORTHY A chronic mouse exposure model that mimics human e-cigarette vaping and allows comparison with tobacco cigarette smoking was developed and utilized to perform longitudinal studies of alterations in cardiovascular function. E-cigarette exposure led to the onset of cardiovascular disease similar to that with tobacco cigarette smoking. Impaired endothelium-dependent and endothelium-independent vasodilation with increased adrenergic vasoconstriction were observed, paralleling the onset of systemic hypertension and subsequent cardiac hypertrophy. This cardiovascular toxicity was dependent on exposure duration and nicotine dose.


Assuntos
Aorta/efeitos dos fármacos , Doenças Cardiovasculares/induzido quimicamente , Nicotina/administração & dosagem , Vaping/efeitos adversos , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Aorta/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Doenças Cardiovasculares/fisiopatologia , Sistemas Eletrônicos de Liberação de Nicotina , Masculino , Camundongos , Fenilefrina/farmacologia , Fatores de Tempo , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
10.
Br J Anaesth ; 126(3): 652-664, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33483132

RESUMO

BACKGROUND: Immunosuppression after surgery is associated with postoperative complications, mediated in part by catecholamines that exert anti-inflammatory effects via the ß-adrenergic receptor. Phenylephrine, generally regarded as a selective α-adrenergic agonist, is frequently used to treat perioperative hypotension. However, phenylephrine may impair host defence through ß-adrenergic affinity. METHODS: Human leukocytes were stimulated with lipopolysaccharide (LPS) in the presence or absence of phenylephrine and α- and ß-adrenergic antagonists. C57BL/6J male mice received continuous infusion of phenylephrine (30-50 µg kg-1 min-1 i.v.) or saline via micro-osmotic pumps, before LPS administration (5 mg kg-1 i.v.) or caecal ligation and puncture (CLP). Twenty healthy males were randomised to a 5 h infusion of phenylephrine (0.5 µg kg-1 min-1) or saline before receiving LPS (2 ng kg-1 i.v.). RESULTS: In vitro, phenylephrine enhanced LPS-induced production of the anti-inflammatory cytokine interleukin (IL)-10 (maximum augmentation of 93%) while attenuating the release of pro-inflammatory mediators. These effects were reversed by pre-incubation with ß-antagonists, but not α-antagonists. Plasma IL-10 levels were higher in LPS-challenged mice infused with phenylephrine, whereas pro-inflammatory mediators were reduced. Phenylephrine infusion increased bacterial counts after CLP in peritoneal fluid (+42%, P=0.0069), spleen (+59%, P=0.04), and liver (+35%, P=0.09). In healthy volunteers, phenylephrine enhanced the LPS-induced IL-10 response (+76%, P=0.0008) while attenuating plasma concentrations of pro-inflammatory mediators including IL-8 (-15%, P=0.03). CONCLUSIONS: Phenylephrine exerts potent anti-inflammatory effects, possibly involving the ß-adrenoreceptor. Phenylephrine promotes bacterial outgrowth after surgical peritonitis. Phenylephrine may therefore compromise host defence in surgical patients and increase susceptibility towards infection. CLINICAL TRIAL REGISTRATION: NCT02675868 (Clinicaltrials.gov).


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Peritonite/imunologia , Fenilefrina/farmacologia , Complicações Pós-Operatórias/imunologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Citocinas/sangue , Humanos , Laboratórios , Leucócitos/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peritonite/metabolismo , Fenilefrina/administração & dosagem , Complicações Pós-Operatórias/metabolismo , Pesquisa Translacional Biomédica
11.
Pharmacol Rep ; 73(1): 255-260, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32860192

RESUMO

BACKGROUND: The spleen is a reservoir for circulating blood cells, and can contract to expel them. METHODS: We have investigated the adrenoceptors involved in isometric contractions of rat spleen produced by noradrenaline (NA) and the α1-adrenoceptor agonist phenylephrine (Phe). RESULTS: Contractions to NA were antagonized by both the α1-adrenoceptor antagonist prazosin (10-8 M) and the α2-adrenoceptor antagonist yohimbine (10-6M), and the combination produced further shifts in NA potency. Contractions to Phe were antagonized by prazosin (10-8 M) which caused a marked parallel shift in the concentration-response curve. High non-selective concentrations of the α1D-adrenoceptor antagonist BMY7378 (10-6 M), the α1A-adrenoceptor antagonist RS100329 ((3 × 10-8 M), and the putative α1B-adrenoceptor antagonist cyclazosin (10-8 M) also produced parallel shifts in the Phe concentration-response curve. BMY7378 at the selective concentration of 3 × 10-8 M had no effect on responses to Phe, but RS100329 in the selective concentration of 3 × 10-9 M produced a marked shift in the effects of high concentrations of Phe. Hence, antagonists in concentrations that block both α1A- and α1B-adrenoceptors produce approximately parallel shifts in Phe potency. CONCLUSIONS: Contractions of rat spleen to adrenergic agonists involve α2- and α1B-adrenoceptors, with a lesser role for α1A-adrenoceptors. This confirms the suggestion that smooth muscle contractions commonly involve multiple subtypes.


Assuntos
Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Baço/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Relação Dose-Resposta a Droga , Masculino , Contração Muscular/efeitos dos fármacos , Norepinefrina/farmacologia , Fenilefrina/farmacologia , Piperazinas/farmacologia , Prazosina/farmacologia , Quinazolinas/farmacologia , Quinoxalinas/farmacologia , Ratos , Ratos Wistar , Timina/farmacologia , Ioimbina/farmacologia
12.
Exp Neurol ; 335: 113517, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33132201

RESUMO

Chronic intermittent hypoxia (CIH) is a model for obstructive sleep apnea. The paraventricular nucleus (PVN) of the hypothalamus has been suggested to contribute to CIH-induced exaggerated cardiorespiratory reflexes, sympathoexcitation and hypertension. This may occur, in part, via activation of the dense catecholaminergic projections to the PVN that originate in the brainstem. However, the contribution of norepinephrine (NE) and activation of its alpha-adrenergic receptors (α-ARs) in the PVN after CIH exposure is unknown. We hypothesized CIH would increase the contribution of catecholaminergic input. To test this notion, we determined the expression of α-AR subtypes, catecholamine terminal density, and synaptic properties of PVN parvocellular neurons in response to α-AR activation in male Sprague-Dawley normoxic (Norm) and CIH exposed rats. CIH decreased mRNA for α1d and α2b AR. Dopamine-ß-hydroxylase (DßH) terminals in the PVN were similar between groups. NE and the α1-AR agonist phenylephrine (PE) increased sEPSC frequency after Norm but not CIH. Block of α1-ARs with prazosin alone did not alter sEPSCs after either Norm or CIH but did prevent agonist augmentation of sEPSC frequency following normoxia. These responses to NE were mimicked by PE during action potential block suggesting presynaptic terminal alterations in CIH. Altogether, these results demonstrate that α1-AR activation participates in neuronal responses in Norm, but are attenuated after CIH. These results may provide insight into the cardiovascular, respiratory and autonomic nervous systems alterations in obstructive sleep apnea.


Assuntos
Hipóxia/fisiopatologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Receptores Adrenérgicos alfa , Síndromes da Apneia do Sono/fisiopatologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Catecolaminas/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Masculino , Neurônios , Norepinefrina/farmacologia , Fenilefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sinapses/metabolismo
13.
Int J Mol Sci ; 21(24)2020 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-33321735

RESUMO

The autonomic nervous system, consisting of sympathetic and parasympathetic branches, plays an important role in regulating metabolic homeostasis. The sympathetic nervous system (SNS) regulates hepatic lipid metabolism by regulating adrenergic receptor activation, resulting in the stimulation of hepatic very-low-density lipoprotein-triglyceride (TG) production in vivo. However, only a few studies on the relationship between SNS and hepatic steatosis have been reported. Here, we investigate the effect of adrenergic receptor agonists on hepatic steatosis in mice fed a high-fat diet (HFD). The α-adrenergic receptor agonist phenylephrine (10 mg/kg/d) or the ß-adrenergic receptor agonist isoproterenol (30 mg/kg/d) was coadministered with HFD to male mice. After five weeks, hepatic steatosis, TG levels, and hepatic fat metabolism-related biomarkers were examined. HFD treatment induced hepatic steatosis, and cotreatment with phenylephrine, but not isoproterenol, attenuated this effect. Phenylephrine administration upregulated the mRNA levels of hepatic peroxisome proliferator-activated receptor alpha and its target genes (such as carnitine palmitoyltransferase 1) and increased hepatic ß-hydroxybutyrate levels. Additionally, phenylephrine treatment increased the expression of the autophagosomal marker LC3-II but decreased that of p62, which is selectively degraded during autophagy. These results indicate that phenylephrine inhibits hepatic steatosis through stimulation of ß-oxidation and autophagy in the liver.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Fígado Gorduroso/metabolismo , Isoproterenol/farmacologia , Fígado/efeitos dos fármacos , Fenilefrina/farmacologia , Ácido 3-Hidroxibutírico/metabolismo , Animais , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/etiologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Triglicerídeos/metabolismo
14.
Sci Rep ; 10(1): 20115, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33208818

RESUMO

Patulin (PAT) is a common mycotoxin contaminant of apple products linked to impaired metabolic and kidney function. Adenosine monophosphate activated protein kinase (AMPK), abundantly expressed in the kidney, intercedes metabolic changes and renal injury. The alpha-1-adrenergic receptors (α1-AR) facilitate Epinephrine (Epi)-mediated AMPK activation, linking metabolism and kidney function. Preliminary molecular docking experiments examined potential interactions and AMPK-gamma subunit 3 (PRKAG3). The effect of PAT exposure (0.2-2.5 µM; 24 h) on the AMPK pathway and α1-AR was then investigated in HEK293 human kidney cells. AMPK agonist Epi determined direct effects on the α1-AR, metformin was used as an activator for AMPK, while buthionine sulphoximine (BSO) and N-acetyl cysteine (NAC) assessed GSH inhibition and supplementation respectively. ADRA1A and ADRA1D expression was determined by qPCR. α1-AR, ERK1/2/MAPK and PI3K/Akt protein expression was assessed using western blotting. PAT (1 µM) decreased α1-AR protein and mRNA and altered downstream signalling. This was consistent in cells stimulated with Epi and metformin. BSO potentiated the observed effect on α1-AR while NAC ameliorated these effects. Molecular docking studies performed on Human ADRA1A and PRKAG3 indicated direct interactions with PAT. This study is the first to show PAT modulates the AMPK pathway and α1-AR, supporting a mechanism of kidney injury.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Patulina/farmacologia , Receptores Adrenérgicos alfa 1/genética , Proteínas Quinases Ativadas por AMP/química , Proteínas Quinases Ativadas por AMP/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Epinefrina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metformina/farmacologia , Simulação de Acoplamento Molecular , Patulina/química , Patulina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo
15.
Exp Dermatol ; 29(12): 1216-1224, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33015872

RESUMO

While human eccrine sweat glands respond to adrenergic agonists, there remains a paucity of information on the factors modulating this response. Thus, we assessed the relative contribution of α- and ß-adrenergic sweating during a heat exposure and as a function of individual factors of sex and training status. α- and ß-adrenergic sweating was assessed in forty-eight healthy young men (n = 35) and women (n = 13) including endurance-trained (n = 12) and untrained men (n = 12) under non-heat exposure (temperate, 25°C; n = 17) and heat exposure (hot, 35°C; n = 48) conditions using transdermal iontophoresis of phenylephrine (α-adrenergic agonist) and salbutamol (ß-adrenergic agonist) on the ventral forearm, respectively. Adrenergic sweating was also measured after iontophoretic administration of atropine (muscarinic receptor antagonist) or saline (control) to evaluate how changes in muscarinic receptor activity modulate the adrenergic response to a heat exposure (n = 12). α- and ß-adrenergic sweating was augmented in hot compared with temperate conditions (both P ≤ .014), albeit the relative increase was greater in ß (~5.4-fold)- as compared to α (~1.5-fold)-adrenergic-mediated sweating response. However, both α- and ß-adrenergic sweating was abolished by atropinization (P = .001). Endurance-trained men showed an augmentation in α- (P = .043) but not ß (P = .960)-adrenergic sweating as compared to untrained men. Finally, a greater α- and ß-adrenergic sweating response (both P ≤ .001) was measured in habitually active men than in women. We show that heat exposure augments α-and ß-adrenergic sweating differently via mechanisms associated with altered muscarinic receptor activity. Sex and training status modulate this response.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Albuterol/farmacologia , Fenilefrina/farmacologia , Condicionamento Físico Humano/fisiologia , Sudorese/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Agonistas de Receptores Adrenérgicos beta 2/administração & dosagem , Albuterol/administração & dosagem , Atropina/farmacologia , Feminino , Antebraço , Temperatura Alta , Humanos , Iontoforese , Masculino , Agonistas Muscarínicos/farmacologia , Antagonistas Muscarínicos/farmacologia , Fenilefrina/administração & dosagem , Pilocarpina/farmacologia , Fatores Sexuais , Sudorese/fisiologia , Adulto Jovem
16.
Hypertension ; 76(4): 1195-1207, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32862711

RESUMO

The hypoxic fetus is at greater risk of cardiovascular demise during a challenge, but the reasons behind this are unknown. Clinically, progress has been hampered by the inability to study the human fetus non-invasively for long period of gestation. Using experimental animals, there has also been an inability to induce gestational hypoxia while recording fetal cardiovascular function as the hypoxic pregnancy is occurring. We use novel technology in sheep pregnancy that combines induction of controlled chronic hypoxia with simultaneous, wireless recording of blood pressure and blood flow signals from the fetus. Here, we investigated the cardiovascular defense of the hypoxic fetus to superimposed acute hypotension. Pregnant ewes carrying singleton fetuses surgically prepared with catheters and flow probes were randomly exposed to normoxia or chronic hypoxia from 121±1 days of gestation (term ≈145 days). After 10 days of exposure, fetuses were subjected to acute hypotension via fetal nitroprusside intravenous infusion. Underlying in vivo mechanisms were explored by (1) analyzing fetal cardiac and peripheral vasomotor baroreflex function; (2) measuring the fetal plasma catecholamines; and (3) establishing fetal femoral vasoconstrictor responses to the α1-adrenergic agonist phenylephrine. Relative to controls, chronically hypoxic fetal sheep had reversed cardiac and impaired vasomotor baroreflex function, despite similar noradrenaline and greater adrenaline increments in plasma during hypotension. Chronic hypoxia markedly diminished the fetal vasopressor responses to phenylephrine. Therefore, we show that the chronically hypoxic fetus displays markedly different cardiovascular responses to acute hypotension, providing in vivo evidence of mechanisms linking its greater susceptibility to superimposed stress.


Assuntos
Barorreflexo/fisiologia , Hipóxia Fetal/fisiopatologia , Hipotensão/fisiopatologia , Resistência Vascular/fisiologia , Vasoconstrição/fisiologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Catecolaminas/sangue , Feminino , Hipóxia Fetal/sangue , Hemodinâmica , Hipotensão/sangue , Hipotensão/induzido quimicamente , Nitroprussiato , Fenilefrina/farmacologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Ovinos , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
17.
Eur J Pharmacol ; 885: 173423, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32750368

RESUMO

In a cell line, stably expressing α1A-adrenoceptors fused to the mCherry red fluorescent protein, noradrenaline, methoxamine, and oxymetazoline induced concentration-dependent increases in intracellular calcium. All of these agents increase α1A-adrenoceptor phosphorylation and internalization. Transient co-expression of these receptors with Rab proteins tagged with the enhanced Green Fluorescent Protein was employed to estimate α1A-adrenoceptor-Rab interaction using Förster Resonance Energy Transfer. Noradrenaline and methoxamine increased α1A-adrenoceptor interaction with Rab5 and Rab7 but did not modify it with Rab9. Oxymetazoline induced adrenoceptor interaction with Rab5 and Rab9 and only an insignificant increase in Rab7 signal. Phorbol myristate acetate increased α1A-adrenoceptor interaction with Rab5 and Rab9 but did not modify it with Rab7. The agonists and the active phorbol ester, all of which induce receptor phosphorylation and internalization, favor receptor interaction with Rab5, i.e., association with early endosomes. Cell stimulation with phorbol myristate acetate induced the α1A-adrenoceptors to interact with the late endosomal marker, Rab9, suggesting that the receptors are directed to slow recycling endosomes once they have transited to the Trans-Golgi network to be retrieved to the plasma membrane. The agonists noradrenaline and methoxamine likely induce a faster recycling and might direct some of the adrenoceptors toward degradation and/or very slow recycling to the plasma membrane. Oxymetazoline produced a mixed pattern of interaction with the Rab proteins. These data indicate that α1A-adrenoceptor agonists can trigger different vesicular traffic and receptor fates within the cells.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Ésteres de Forbol/farmacologia , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Proteínas rab de Ligação ao GTP/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular , Endossomos/efeitos dos fármacos , Humanos , Proteínas Luminescentes , Metoxamina/farmacologia , Norepinefrina/farmacologia , Oximetazolina/farmacologia , Fosforilação , Acetato de Tetradecanoilforbol/farmacologia , Proteínas rab5 de Ligação ao GTP/efeitos dos fármacos , Rede trans-Golgi/efeitos dos fármacos
18.
Neurourol Urodyn ; 39(7): 1930-1938, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32609936

RESUMO

AIMS: Urofacial syndrome (UFS) is an autosomal recessive disease characterized by detrusor contraction against an incompletely dilated outflow tract. This dyssynergia causes dribbling incontinence and incomplete voiding. Around half of individuals with UFS have biallelic mutations of HPSE2 that encodes heparanase 2, a protein found in pelvic ganglia and bladder nerves. Homozygous Hpse2 mutant mice have abnormal patterns of nerves in the bladder body and outflow tract, and also have dysfunctional urinary voiding. We hypothesized that bladder neurophysiology is abnormal Hpse2 mutant mice. METHODS: Myography was used to study bladder bodies and outflow tracts isolated from juvenile mice. Myogenic function was analyzed after chemical stimulation or blockade of key receptors. Neurogenic function was assessed by electrical field stimulation (EFS). Muscarinic receptor expression was semi-quantified by Western blot analysis. RESULTS: Nitrergic nerve-mediated relaxation of precontracted mutant outflow tracts was significantly decreased vs littermate controls. The contractile ability of mutant outflow tracts was normal as assessed by KCl and the α1-adrenoceptor agonist phenylephrine. EFS of mutant bladder bodies induced significantly weaker contractions than controls. Conversely, the muscarinic agonist carbachol induced significantly stronger contractions of bladder body than controls. CONCLUSIONS: The Hpse2 model of UFS features aberrant bladder neuromuscular physiology. Further work is required to determine whether similar aberrations occur in patients with UFS.


Assuntos
Glucuronidase/genética , Bexiga Urinaria Neurogênica/genética , Bexiga Urinaria Neurogênica/fisiopatologia , Doenças Urológicas/genética , Doenças Urológicas/fisiopatologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Carbacol/farmacologia , Estimulação Elétrica , Facies , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Agonistas Muscarínicos/farmacologia , Contração Muscular/efeitos dos fármacos , Mutação/genética , Óxido Nítrico/fisiologia , Fenilefrina/farmacologia , Cloreto de Potássio/farmacologia , Receptores Muscarínicos/biossíntese , Receptores Muscarínicos/genética , Urodinâmica
19.
Eur J Pharmacol ; 883: 173283, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32619676

RESUMO

Diabetes and hypertension can coexist and exacerbate each other. In the early stages of diabetes, there is a decreased vascular response of the sympathetic nervous system (SNS), probably due to lower expression of α1-adrenoceptors; however, it is unclear how diabetes in advanced stages changes the functionality of the SNS, especially the expression of α1-adrenoceptors. Thus, the aim of this work was to analyse the functional response to phenylephrine, a selective α1-adrenoceptor agonist, and the expression of α1-adrenoceptors in chronic diabetes and hypertension. Male SHR and WKY rats aged 10-12 weeks were administered either streptozotocin (60 mg/kg i.p.) or a vehicle (control group). Eight weeks after administration, dose-response curves to phenylephrine were generated and the gene and protein expression of α1-adrenoceptor subtypes (α1A-, α1B- and α1D-adrenoceptors) in the heart and aorta were measured. The response to phenylephrine was diminished in hypertensive rats and in normotensive diabetic rats. The coexistence of both diabetes and hypertension produced an even smaller response to phenylephrine than that observed for each condition separately. In the heart and aorta of diabetic rats, no changes in α1A-, α1B- or α1D-adrenoceptor mRNA expression were observed; however, protein expression was increased, mainly for the α1D-adrenoceptor. Hypertension increased mRNA and protein expression of α1-adrenoceptors in a tissue-dependent manner. The coexistence of both diabetes and hypertension produced differences in the regulation of mRNA and protein expression (increase or decrease) in both the heart and aorta. In conclusion, diabetes, hypertension and the coexistence of both pathologies impairs the in vivo response to phenylephrine. However, the differences in α1A-, α1B- and α1D-adrenoceptor expression cannot explain the reduced response to the agonist. This should be further explored in future experiments.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Aorta/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Hipertensão/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Fenilefrina/farmacologia , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Animais , Aorta/metabolismo , Glicemia/metabolismo , Pressão Sanguínea , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Miócitos Cardíacos/metabolismo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo
20.
Int J Mol Sci ; 21(11)2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32485850

RESUMO

The multifunctional tissue transglutaminase has been demonstrated to act as α1-adrenergic receptor-coupled G protein with GTPase activity in several cell types. To explore further the pathophysiological significance of this function we investigated the in vivo effects of the α1-adrenergic receptor agonist phenylephrine comparing responses in wild type and TG2-/- mice. Injection of phenylephrine, but not a beta3-adrenergic agonist (CL-316,243), resulted in the long-term decline of the respiratory exchange ratio and lower lactate concentration in TG2-/- mice indicating they preferred to utilize fatty acids instead of glucose as fuels. Measurement of tail blood pressure revealed that the vasoconstrictive effect of phenylephrine was milder in TG2-/- mice leading to lower levels of lactate dehydrogenase (LDH) isoenzymes in blood. LDH isoenzyme patterns indicated more damage in lung, liver, kidney, skeletal, and cardiac muscle of wild type mice; the latter was confirmed by a higher level of heart-specific CK-MB. Our data suggest that TG2 as an α1-adrenergic receptor-coupled G protein has important regulatory functions in alpha1-adrenergic receptor-mediated metabolic processes and vascular functions.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transglutaminases/metabolismo , Vasoconstrição , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , Dioxóis/farmacologia , Ácidos Graxos/metabolismo , Proteínas de Ligação ao GTP/genética , Glucose/metabolismo , Rim/metabolismo , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Fenilefrina/farmacologia , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...