Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 923
Filtrar
1.
Med Oncol ; 41(4): 87, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38472423

RESUMEN

Liver cancer annually accounts for over 800,000 cases and 700,000 deaths worldwide. Hepatocellular carcinoma is responsible for over 80% of liver cancer cases. Due to ineffective treatment options and limited surgical interventions, hepatocellular carcinoma is notoriously difficult to treat. Nonetheless, drugs utilized for other medical conditions, such as the antihypertensive medication prazosin, the neuroleptic medication chlorpromazine, and the neuroleptic medication haloperidol, have gained attention for their potential anti-cancer effects. Therefore, this study used these medications for investigating toxicity to hepatocellular carcinoma while testing the adverse effects on a noncancerous liver cell line model THLE-2. After treatment, an XTT cell viability assay, cell apoptosis assay, reactive oxygen species (ROS) assay, apoptotic proteome profile, and western blot were performed. We calculated IC50 values for chlorpromazine and prazosin to have a molar range of 35-65 µM. Our main findings suggest the capability of both of these treatments to reduce cell viability and generate oxidative stress in HepG2 and THLE-2 cells (p value < 0.05). Haloperidol, however, failed to demonstrate any reduction in cell viability revealing no antitumor effect up to 100 µM. Based on our findings, a mechanism of cell death was not able to be established due to lack of cleaved caspase-3 expression. Capable of bypassing many aspects of the lengthy, costly, and difficult cancer drug approval process, chlorpromazine and prazosin deserve further investigation for use in conjunction with traditional chemotherapeutics.


Asunto(s)
Antineoplásicos , Antipsicóticos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Haloperidol/farmacología , Haloperidol/uso terapéutico , Clorpromazina/farmacología , Clorpromazina/uso terapéutico , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Prazosina/farmacología , Prazosina/uso terapéutico , Células Hep G2 , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral
2.
Toxicon ; 218: 57-65, 2022 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-36113683

RESUMEN

Rhinella marina toad is abundant in Brazil. Its poison contains cardiac glycosides called bufadienolides, which are extensively investigated for their bioactivity. Our aim was to characterize the vasoactivity of Rhinella marina poison (RmP) on the aorta of male Wistar rats. For this, the RmP was first collected and processed to obtain an alcoholic extract. To determine cardiovascular effects of RmP, we performed in vivo tests by administering RmP intravenously in doses of 0.1-0.8 mg/kg. Vascular reactivity was also performed through concentration-response curves to RmP (10 ng/mL to 200 µg/mL) in aortic segments with and without endothelium. RmP induced a concentration-dependent contraction in rat aorta which was partly endothelium-mediated. Nitric oxide contributes with this response in view that incubation with L-NAME increased the contractile response. Additionally, treatment with indomethacin [cyclooxygenase, (COX) inhibitor], nifedipine (L-type voltage-gated calcium channels blocker), and BQ-123 (ETA receptors antagonist) decreased maximum response, and ketanserin (5-HT2 receptors antagonist) decreased pEC50, suggesting active participation of these pathways in the contractile response. On the other hand, apocynin (NADPH oxidase inhibitor) did not alter contractility. Incubation with prazosin (α1-adrenergic receptor antagonist) abolished the contractile response, suggesting that the RmP-induced contraction is dependent on the adrenergic pathway. In the Na+/K+ ATPase protocol, a higher Emax was observed in the RmP experimental group, suggesting that RmP potentiated Na+/K+ATPase hyperpolarizing response. When this extract was injected (i.v.) in vivo, increase in blood pressure and decrease in heart rate were observed. The results were immediate and transitory, and occurred in a dose-dependent manner. Overall, these data suggest that the poison extract of R. marina toad has an important vasoconstrictor action and subsequent vasopressor effects, and its use can be investigated to some cardiovascular disorders.


Asunto(s)
Bufanólidos , Venenos , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/farmacología , Adrenérgicos/farmacología , Antagonistas Adrenérgicos/farmacología , Animales , Bufanólidos/toxicidad , Bufo marinus/metabolismo , Canales de Calcio , Endotelio Vascular , Hemodinámica , Indometacina/farmacología , Ketanserina/farmacología , Masculino , Metanol/farmacología , NADPH Oxidasas , NG-Nitroarginina Metil Éster , Nifedipino/farmacología , Óxido Nítrico/metabolismo , Prazosina/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Wistar , Serotonina/farmacología , Vasoconstrictores
3.
Chin J Physiol ; 65(4): 179-186, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36073566

RESUMEN

Bilateral nucleus tractus solitarii (NTS) lesions, possibly caused by enterovirus 71 infection, cause severe neurogenic hypertension, leading to acute heart failure (HF), pulmonary edema, and death within hours. Alpha-adrenergic blockers attenuate blood pressure and ameliorate HF and pulmonary edema, thereby prolonging survival time. However, the molecular mechanisms of these blockers are not clear. In this study, we investigated these mechanisms in a rat model of 6-hydroxydopamine (6-OHDA)-induced HF. Sprague-Dawley rats were treated with prazosin 10 min after the microinjection of 6-OHDA into the NTS. Immunohistochemistry and dihydroethidium (DHE) staining were used for analysis. In the cardiac tissue of 6-OHDA-induced HF, in situ expression of tumor necrosis factor-alpha (TNF-α), fibroblast growth factor-23 (FGF23), and FGF receptor 1 (FGFR1) increased, but in situ expression of Vitamin D receptor (VDR) decreased. DHE staining revealed several heart cells with high reactive oxygen species production. Prazosin treatment decreased TNF-α, FGF23, and FGFR1 expression in the heart of rats with 6-OHDA-induced HF. It also prevented cardiomyopathy caused by 6-OHDA-induced bilateral NTS lesions by inhibiting the FGF23-FGFR1 pathway and downregulating TNF-α expression. In situ, FGF23, FGFR1, VDR, superoxide, and TNF-α in the heart were found to be involved in acute HF in our rat model of 6-OHDA-induced bilateral NTS lesions. These findings are potentially useful for treating fatal enterovirus 71 infection-induced NTS lesions and HF.


Asunto(s)
Insuficiencia Cardíaca , Edema Pulmonar , Animales , Regulación hacia Abajo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos , Insuficiencia Cardíaca/tratamiento farmacológico , Oxidopamina , Prazosina/farmacología , Prazosina/uso terapéutico , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa
4.
Pflugers Arch ; 474(12): 1311-1321, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36131146

RESUMEN

Atrial fibrillation (AF) from elevated adrenergic activity may involve increased atrial L-type Ca2+ current (ICaL) by noradrenaline (NA). However, the contribution of the adrenoceptor (AR) sub-types to such ICaL-increase is poorly understood, particularly in human. We therefore investigated effects of various broad-action and sub-type-specific α- and ß-AR antagonists on NA-stimulated atrial ICaL. ICaL was recorded by whole-cell-patch clamp at 37 °C in myocytes isolated enzymatically from atrial tissues from consenting patients undergoing elective cardiac surgery and from rabbits. NA markedly increased human atrial ICaL, maximally by ~ 2.5-fold, with EC75 310 nM. Propranolol (ß1 + ß2-AR antagonist, 0.2 microM) substantially decreased NA (310 nM)-stimulated ICaL, in human and rabbit. Phentolamine (α1 + α2-AR antagonist, 1 microM) also decreased NA-stimulated ICaL. CGP20712A (ß1-AR antagonist, 0.3 microM) and prazosin (α1-AR antagonist, 0.5 microM) each decreased NA-stimulated ICaL in both species. ICI118551 (ß2-AR antagonist, 0.1 microM), in the presence of NA + CGP20712A, had no significant effect on ICaL in human atrial myocytes, but increased it in rabbit. Yohimbine (α2-AR antagonist, 10 microM), with NA + prazosin, had no significant effect on human or rabbit ICaL. Stimulation of atrial ICaL by NA is mediated, based on AR sub-type antagonist responses, mainly by activating ß1- and α1-ARs in both human and rabbit, with a ß2-inhibitory contribution evident in rabbit, and negligible α2 involvement in either species. This improved understanding of AR sub-type contributions to noradrenergic activation of atrial ICaL could help inform future potential optimisation of pharmacological AR-antagonism strategies for inhibiting adrenergic AF.


Asunto(s)
Canales de Calcio Tipo L , Miocitos Cardíacos , Norepinefrina , Receptores Adrenérgicos alfa , Receptores Adrenérgicos beta , Animales , Humanos , Conejos , Fibrilación Atrial/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Norepinefrina/farmacología , Norepinefrina/fisiología , Prazosina/farmacología , Receptores Adrenérgicos alfa 2 , Atrios Cardíacos/citología , Receptores Adrenérgicos beta/fisiología , Receptores Adrenérgicos alfa/fisiología , Antagonistas Adrenérgicos alfa/farmacología , Antagonistas Adrenérgicos beta/farmacología , Canales de Calcio Tipo L/fisiología
5.
Pharmacol Res Perspect ; 10(4): e00991, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35892277

RESUMEN

The relationship between cardiovascular diseases and iron disorders has gained increasing attention; however, the effects of hypotensive drugs on iron metabolic alterations in hypertension are not well understood. The purpose of this study was to investigate iron metabolic changes after prazosin treatment of spontaneously hypertensive rats (SHRs) and Wistar-Kyoto (WKY) rats. Our second objective was to examine the effects of hypertension and anti-hypertensive drugs on bone formation and resorption. SHRs and WKY rats were randomized into either prazosin-treated groups (WKY + PZ and SHR + PZ) or untreated groups (WKY and SHR). After 7 days of intragastric prazosin administration, the rats were sacrificed for analysis; blood samples and organs (the duodenum, liver, kidneys, spleen, and femur) were collected. Both WKY + PZ and SHR groups exhibited iron deficiency in the serum and liver. Prazosin increased the iron levels in the bone tissue of SHRs. Prazosin stimulated the expression of hepcidin mRNA in the liver of SHRs and inhibited the expression of this iron-regulatory hormone in WKY rats. FPN1 expression in the duodenum was increased significantly in SHRs, however markedly decreased after prazosin treatment. The expression of TLR4 and Ctsk was enhanced in the bone tissue of SHRs, whereas CLC-7 expression was inhibited. Both hypotension and hypertension can lead to iron deficiency. Treatment with prazosin restored iron homeostasis in SHRs. The inverse impacts of prazosin on hepatic hepcidin expression in SHRs versus WKY rats indicates differing iron regulatory mechanisms between hypertensive and normal animals. The osteoclast activity was found to be enhanced in SHRs. Further study is needed to address whether the changes in osteoblast and osteoclast activity in SHRs correlates with the effects on iron metabolism.


Asunto(s)
Hipertensión , Hierro , Prazosina , Animales , Hepcidinas/genética , Hipertensión/tratamiento farmacológico , Hierro/metabolismo , Prazosina/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
6.
Int J Mol Sci ; 23(8)2022 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-35457136

RESUMEN

In this study, we examined whether aortic contraction, induced by the alpha-2 adrenoceptor agonist dexmedetomidine, is involved in the transactivation of the epidermal growth factor receptor (EGFR) in isolated endothelium-denuded rat aortas. Additionally, we aimed to elucidate the associated underlying cellular mechanisms. The effects of the alpha-2 adrenoceptor inhibitor rauwolscine, EGFR tyrosine kinase inhibitor AG1478, Src kinase inhibitors PP1 and PP2, and matrix metalloproteinase inhibitor GM6001 on EGFR tyrosine phosphorylation and c-Jun NH2-terminal kinase (JNK) phosphorylation induced by dexmedetomidine in rat aortic smooth muscles were examined. In addition, the effects of these inhibitors on dexmedetomidine-induced contraction in isolated endothelium-denuded rat aorta were examined. Dexmedetomidine-induced contraction was inhibited by the alpha-1 adrenoceptor inhibitor prazosin, rauwolscine, AG1478, PP1, PP2, and GM6001 alone or by a combined treatment with prazosin and AG1478. AG1478 (3 × 10-6 M) inhibited dexmedetomidine-induced contraction in isolated endothelium-denuded rat aortas pretreated with rauwolscine. Dexmedetomidine-induced EGFR tyrosine and JNK phosphorylation were inhibited by rauwolscine, PP1, PP2, GM6001, and AG1478. Furthermore, dexmedetomidine-induced JNK phosphorylation reduced upon EGFR siRNA treatment. Therefore, these results suggested that the transactivation of EGFR associated with dexmedetomidine-induced contraction, mediated by the alpha-2 adrenoceptor, Src kinase, and matrix metalloproteinase, caused JNK phosphorylation and increased calcium levels.


Asunto(s)
Dexmedetomidina , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Aorta/metabolismo , Dexmedetomidina/farmacología , Receptores ErbB/metabolismo , Músculo Liso Vascular/metabolismo , Fosforilación , Prazosina/farmacología , Ratas , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Activación Transcripcional , Tirosina/metabolismo , Yohimbina/farmacología , Familia-src Quinasas/metabolismo
7.
Curr Drug Deliv ; 19(4): 508-517, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34238184

RESUMEN

BACKGROUND: The research and development of drugs for the treatment of central nervous system diseases faces many challenges at present. One of the most important questions to be answered is, how does the drug cross the blood-brain barrier to get to the target site for pharmacological action. Fluoxetine is widely used in clinical antidepressant therapy. However, the mechanism by which fluoxetine passes through the BBB also remains unclear. Under physiological pH conditions, fluoxetine is an organic cation with a relatively small molecular weight (<500), which is in line with the substrate characteristics of organic cation transporters (OCTs). Therefore, this study aimed to investigate the interaction of fluoxetine with OCTs at the BBB and BBB-associated efflux transporters. This is of great significance for fluoxetine to better treat depression. Moreover, it can provide a theoretical basis for clinical drug combination. METHODS: In vitro BBB model was developed using human brain microvascular endothelial cells (hCMEC/D3), and the cellular accumulation was tested in the presence or absence of transporter inhibitors. In addition, an in vivo trial was performed in rats to investigate the effect of OCTs on the distribution of fluoxetine in the brain tissue. Fluoxetine concentration was determined by a validated UPLC-MS/MS method. RESULTS: The results showed that amantadine (an OCT1/2 inhibitor) and prazosin (an OCT1/3 inhibitor) significantly decreased the cellular accumulation of fluoxetine (P <.001). Moreover, we found that N-methylnicotinamide (an OCT2 inhibitor) significantly inhibited the cellular uptake of 100 and 500 ng/mL fluoxetine (P <.01 and P <.05 respectively). In contrast, corticosterone (an OCT3 inhibitor) only significantly inhibited the cellular uptake of 1000 ng/mL fluoxetine (P <.05). The P-glycoprotein (P-gp) inhibitor, verapamil, and the multidrug resistance associated proteins (MRPs) inhibitor, MK571, significantly decreased the cellular uptake of fluoxetine. However, intracellular accumulation of fluoxetine was not significantly changed when fluoxetine was incubated with the breast cancer resistance protein (BCRP) inhibitor Ko143. Furthermore, in vivo experiments proved that corticosterone and prazosin significantly inhibited the brain-plasma ratio of fluoxetine at 5.5 h and 12 h, respectively. CONCLUSION: OCTs might play a significant role in the transport of fluoxetine across the BBB. In addition, P-gp, BCRP, and MRPs seemed not to mediate the efflux transport of fluoxetine.


Asunto(s)
Barrera Hematoencefálica , Fluoxetina , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Cromatografía Liquida , Corticosterona/metabolismo , Corticosterona/farmacología , Células Endoteliales/metabolismo , Fluoxetina/metabolismo , Fluoxetina/farmacología , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/farmacología , Prazosina/metabolismo , Prazosina/farmacología , Ratas , Espectrometría de Masas en Tándem
8.
Biol Pharm Bull ; 44(3): 458-460, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33642556

RESUMEN

The functional role of ATP released from sympathetic nerve terminals was examined in isolated guinea pig ventricular papillary muscles. The contractile force of papillary muscles was increased by field electrical stimulation of sympathetic nerve endings. This increase was attenuated by pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) or suramin, blockers of the P2X receptor, and was abolished by propranolol and prazosin. PPADS, suramin, and ATP affected neither the basal contractile force nor the positive inotropic effect of noradrenaline. These results provide functional evidence that ATP released from sympathetic nerve terminals enhances noradrenaline release and contributes to sympathetic nerve-induced inotropy.


Asunto(s)
Adenosina Trifosfato/fisiología , Retroalimentación Fisiológica , Músculos Papilares/fisiología , Sistema Nervioso Simpático , Función Ventricular , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Cobayas , Ventrículos Cardíacos , Masculino , Contracción Muscular , Norepinefrina/fisiología , Prazosina/farmacología , Propranolol/farmacología , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Suramina/farmacología
9.
J Pharmacol Sci ; 145(3): 228-240, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33602503

RESUMEN

Astrocytes are glial cells with numerous fine processes which are important for the functions of the central nervous system. The activation of ß-adrenoceptors induces process formation of astrocytes via cyclic AMP (cAMP) signaling. However, the role of α-adrenoceptors in the astrocyte morphology has not been elucidated. Here, we examined it by using cultured astrocytes from neonatal rat spinal cords and cortices. Exposure of these cells to noradrenaline and the ß-adrenoceptor agonist isoproterenol increased intracellular cAMP levels and induced the formation of processes. Noradrenaline-induced process formation was enhanced with the α1-adrenoceptor antagonist prazosin and α2-adrenoceptor antagonist atipamezole. Atipamezole also enhanced noradrenaline-induced cAMP elevation. Isoproterenol-induced process formation was not inhibited by the α1-adrenoceptor agonist phenylephrine but was inhibited by the α2-adrenoceptor agonist dexmedetomidine. Dexmedetomidine also inhibited process formation induced by the adenylate cyclase activator forskolin and the membrane-permeable cAMP analog dibutyryl-cAMP. Moreover, dexmedetomidine inhibited cAMP-independent process formation induced by adenosine or the Rho-associated kinase inhibitor Y27632. In the presence of propranolol, noradrenaline inhibited Y27632-induced process formation, which was abolished by prazosin or atipamezole. These results demonstrate that α-adrenoceptors inhibit both cAMP-dependent and -independent astrocytic process formation.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Receptores Adrenérgicos alfa/fisiología , Receptores Adrenérgicos beta/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Dexmedetomidina/farmacología , Imidazoles/farmacología , Isoproterenol/farmacología , Norepinefrina/farmacología , Prazosina/farmacología , Ratas Wistar , Transducción de Señal
10.
Vascul Pharmacol ; 136: 106810, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33181321

RESUMEN

ATP, norepinephrine and NPY are co-released by sympathetic nerves innervating arteries. ATP elicits vasoconstriction via activation of smooth muscle P2X receptors. The functional interaction between neuropeptide Y (NPY) and P2X receptors in arteries is not known. In this study we investigate the effect of NPY on P2X1-dependent vasoconstriction in mouse mesenteric arteries. Suramin or P2X1 antagonist NF449 abolished α,ß-meATP evoked vasoconstrictions. NPY lacked any direct vasoconstrictor effect but facilitated the vasoconstrictive response to α,ß-meATP. Mesenteric arteries expressed Y1 and Y4 receptors, but not Y2 or Y5. Y1 receptor inhibition (BIBO3304) reversed NPY facilitation of the α,ß-meATP-evoked vasoconstriction. L-type Ca2+ channel antagonism (nifedipine) had no effect on α,ß-meATP-evoked vasoconstrictions, but completely reversed NPY facilitation. Electrical field stimulation evoked sympathetic neurogenic vasoconstriction. Neurogenic responses were dependent upon dual α1-adrenergic (prazosin) and P2X1 (NF449) receptor activation. Y1 receptor antagonism partially reduced neurogenic vasoconstriction. Isolation of the P2X1 component by α1-adrenergic blockade allowed faciliatory effects of Y1 receptor activation to be explored. Y1 receptor antagonism reduced the P2X1 receptor component during neurogenic vasoconstriction. α1-adrenergic and P2X1 receptors are post-junctional receptors during sympathetic neurogenic vasoconstriction in mesenteric arteries. In conclusion, we have identified that NPY lacks a direct vasoconstrictor effect in mesenteric arteries but can facilitate vasoconstriction by enhancing the activity of P2X1, following activation by exogenous agonists or during sympathetic nerve stimulation. The mechanism of P2X1 facilitation by NPY involved activation of the NPY Y1 receptor and the L-type Ca2+ channel.


Asunto(s)
Arterias Mesentéricas/inervación , Neuropéptido Y/farmacología , Receptores de Neuropéptido Y/agonistas , Receptores Purinérgicos P2X1/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Bencenosulfonatos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Masculino , Ratones Endogámicos C57BL , Nifedipino/farmacología , Prazosina/farmacología , Receptores de Neuropéptido Y/metabolismo , Suramina/farmacología , Sistema Nervioso Simpático/metabolismo
11.
Psychopharmacology (Berl) ; 238(2): 475-486, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33150479

RESUMEN

RATIONALE AND OBJECTIVES: The reinforcement-enhancing effect (REE) of nicotine refers to the drug's ability to enhance the strength of other primary and conditioned reinforcers. The main aim was to investigate neuropharmacological mechanisms underlying nicotine's strengthening of a primary visual reinforcer (i.e., a light cue), using a subcutaneous (SC) dose previously shown to provide plasma nicotine levels associated with habitual smoking. METHODS: Adult male rats pressed an "active" lever to illuminate a brief cue light during daily 60-min sessions. Rats that showed a clear REE were tested with systemically administered pretreatment drugs followed by nicotine (0.1 mg/kg SC) or saline challenge, in within-subject counterbalanced designs. Pretreatments were mecamylamine (nicotinic, 0.1-1 mg/kg SC), SCH 39166 (D1-like dopaminergic, 0.003-0.2 mg/kg SC), naloxone (opioid, 1 and 5 mg/kg SC), prazosin (alpha1-adrenergic antagonist, 1 and 2 mg/kg IP), rimonabant (CB1 cannabinoid inverse agonist, 3 mg/kg IP), sulpiride (D2-like dopaminergic antagonist, 40 mg/kg SC), or propranolol (beta-adrenergic antagonist, 10 mg/kg IP). RESULTS: The nicotine REE was abolished by three antagonists at doses that did not impact motor output, i.e., mecamylamine (1 mg/kg), SCH 39166 (0.01 and 0.03 mg/kg), and naloxone (5 mg/kg). Prazosin and rimonabant both attenuated the nicotine REE, but rimonabant also suppressed responding more generally. The nicotine REE was not significantly altered by sulpiride or propranolol. CONCLUSIONS: In adult male rats, the reinforcement-enhancing effect of low-dose nicotine depends on nicotinic receptor stimulation and on neurotransmission via D1/D5 dopaminergic, opioid, alpha1-adrenergic, and CB1 cannabinoid receptors.


Asunto(s)
Nicotina/farmacología , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo , Refuerzo en Psicología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Benzazepinas/farmacología , Condicionamiento Operante , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Mecamilamina/farmacología , Prazosina/farmacología , Propranolol/farmacología , Ratas
12.
Am J Physiol Regul Integr Comp Physiol ; 320(2): R105-R116, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33175586

RESUMEN

Hypoxia tolerance in the vertebrate brain often involves chemical modulators that arrest neuronal activity to conserve energy. However, in intact networks, it can be difficult to determine whether hypoxia triggers modulators to stop activity in a protective manner or whether activity stops because rates of ATP synthesis are insufficient to support network function. Here, we assessed the extent to which neuromodulation or metabolic limitations arrest activity in the respiratory network of bullfrogs-a circuit that survives moderate periods of oxygen deprivation, presumably, by activating an inhibitory noradrenergic pathway. We confirmed that hypoxia and norepinephrine (NE) reduce network output, consistent with the view that hypoxia may cause the release of NE to inhibit activity. However, these responses differed qualitatively; hypoxia, but not NE, elicited a large motor burst and silenced the network. The stereotyped response to hypoxia persisted in the presence of both NE and an adrenergic receptor blocker that eliminates sensitivity to NE, indicating that noradrenergic signaling does not cause the arrest. Pharmacological inhibition of glycolysis and mitochondrial respiration recapitulated all features of hypoxia on network activity, implying that reduced ATP synthesis underlies the effects of hypoxia. Finally, activating modulatory mechanisms that dampen neuronal excitability when ATP levels fall, KATP channels and AMP-dependent protein kinase, did not resemble the hypoxic response. These results suggest that energy failure-rather than inhibitory modulation-silences the respiratory network during hypoxia and emphasize the need to account for metabolic limitations before concluding that modulators arrest activity as an adaptation for energy conservation in the nervous system.


Asunto(s)
Tronco Encefálico/fisiología , Metabolismo Energético/fisiología , Consumo de Oxígeno/fisiología , Rana catesbeiana/fisiología , Adenosina Trifosfato/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Desoxiglucosa/farmacología , Femenino , Humanos , Ácido Yodoacético/farmacología , Norepinefrina/farmacología , Prazosina/farmacología
13.
Cell Rep ; 32(2): 107905, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668257

RESUMEN

Cross-presentation of antigens by dendritic cells (DCs) is critical for initiation of anti-tumor immune responses. Yet, key steps involved in trafficking of antigens taken up by DCs remain incompletely understood. Here, we screen 700 US Food and Drug Administration (FDA)-approved drugs and identify 37 enhancers of antigen import from endolysosomes into the cytosol. To reveal their mechanism of action, we generate proteomic organellar maps of control and drug-treated DCs (focusing on two compounds, prazosin and tamoxifen). By combining organellar mapping, quantitative proteomics, and microscopy, we conclude that import enhancers undergo lysosomal trapping leading to membrane permeation and antigen release. Enhancing antigen import facilitates cross-presentation of soluble and cell-associated antigens. Systemic administration of prazosin leads to reduced growth of MC38 tumors and to a synergistic effect with checkpoint immunotherapy in a melanoma model. Thus, inefficient antigen import into the cytosol limits antigen cross-presentation, restraining the potency of anti-tumor immune responses and efficacy of checkpoint blockers.


Asunto(s)
Antineoplásicos/farmacología , Citosol/metabolismo , Endosomas/metabolismo , Inmunidad , Neoplasias/inmunología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antígenos/metabolismo , Transporte Biológico/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Citosol/efectos de los fármacos , Células Dendríticas/metabolismo , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Endosomas/efectos de los fármacos , Inmunidad/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/tratamiento farmacológico , Permeabilidad , Prazosina/farmacología , Quinazolinas/farmacología , Tamoxifeno/farmacología , beta-Lactamasas/metabolismo
15.
J Mol Neurosci ; 70(7): 1120-1129, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32133592

RESUMEN

The predator scent-induced (PSI) stress model is a rat model used to mimic post-traumatic stress disorder (PTSD) symptoms in humans. There is growing evidence that prazosin, which blocks α-1 and is approved by the FDA as an anti-hypertensive drug, can potentially be of use in the treatment of PTSD-related sleep disorders. The aim of this study was to investigate the role of prazosin treatment on behavioral parameters (freezing time, total transitions, and rearing frequency measured from the open field; anxiety index, total entries and time spent in open arms calculated from the elevated plus maze), apoptotic proteins and α-2c-AR in fear memory reconsolidation in the PSI stress rat model. We used western blot analysis to determine the effect of prazosin (0.5 mg/kg/ip) on α-2c-AR and apoptotic protein expression changes in the frontal cortex, hippocampus, and amygdala. It was determined that in the stress group, there was increased freezing time and anxiety index, and decreased rearing frequency, total transitions, total entries, and time spent in open arms compared to the control groups. Following PSI-stress, pro-apoptotic (bax) protein expression levels increased and α-2c AR and anti-apoptotic protein (bcl-2) levels decreased in investigated all brain regions. The majority of stress-induced changes were recovered with prazosin treatment. The results of our study may potentially be useful in understanding the effect of prazosin treatment, given the fact that the amygdala, frontal cortex, and hippocampus regions are affected for stress conditions.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Prazosina/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Proteína X Asociada a bcl-2/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Apoptosis , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Femenino , Masculino , Prazosina/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas , Ratas Wistar , Proteína X Asociada a bcl-2/genética
16.
Biomed Pharmacother ; 124: 109731, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31954876

RESUMEN

BACKGROUND: Prazosin, a non-selective α1-adrenoceptor and a selective α2B-adrenoceptor antagonist, is reported to possess anti-cancer activity in some types of cancer. The aim of this study was to investigate the effect of prazosin on acute myeloid leukemia (AML) and the underlying relevant mechanisms. METHODS: AML cell lines U937 and HL60 were treated with different concentration of prazosin (5, 10 and 15 µM), CCK8 and flow cytometry assays were performed to examine the effects of prazosin on cell viability, cell cycle distribution and apoptosis. Western blot assay was used to detect the expression of related proteins. RESULTS: We observed that prazosin inhibited cell viability of U937 and HL60 cells and induced the rate of apoptosis in a dose-dependent manner, as well as induced cell cycle arrest at G1 phase. The activation of PI3K/Akt/mTOR signaling pathway was significantly suppressed by prazosin via reducing the phosphorylation of Akt and mTOR. Moreover, by RNA-seq analysis, we found that the expression of tensin 1 (TNS1) was down-regulated by prazosin, and down-regulation of TNS1 could inhibit cell viability of U937 and HL60 cells, as well as induced cell apoptosis. The PI3K/Akt/mTOR signaling pathway was also suppressed by depletion of TNS1. Furthermore, up-regulation of TNS1 could reverse the effects of prazosin on viability and apoptosis in U937 and HL-60 cells, as well as the PI3K/Akt/mTOR signaling pathway. CONCLUSION: These results highlight an anti-cancer activity of prazosin on AML by inhibiting the PI3K/Akt/mTOR pathway and targeting TNS1.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Leucemia Mieloide Aguda/metabolismo , Prazosina/farmacología , Tensinas/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
17.
Nat Prod Res ; 34(23): 3369-3372, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30698035

RESUMEN

Oxalis pes-caprae L. is a plant of the Oxalidaceae family, from which several compounds have been previously identified. Recently, we showed that an Oxalis pes-caprae L. extract inhibits the vasopressor effect of noradrenaline. In this work we aimed to explore the mechanisms involved in this effect. The results confirmed that the flavonoid fraction present in the extract inhibits noradrenaline-induced contractions and that this effect is concentration-dependent. Also, a parallel shift to the right in the noradrenaline concentration-response curve was observed, suggesting a decrease in efficacy and also in potency. Together these results support the assumption that the extract could exert a non-competitive antagonism on the α-adrenergic receptors. However, experiments in the presence of competitive antagonists for α-adrenergic receptor sub-types (i.e. prazosin, yohimbine and phentolamine) showed that the effect may not be directly mediated by α-adrenergic receptors. Thus, the interaction of this extract with the adrenergic system remains to be confirmed.


Asunto(s)
Oxalidaceae/química , Extractos Vegetales/farmacología , Polifenoles/análisis , Arterias Torácicas/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Humanos , Persona de Mediana Edad , Norepinefrina/farmacología , Extractos Vegetales/química , Hojas de la Planta/química , Prazosina/farmacología , Receptores Adrenérgicos alfa/efectos de los fármacos , Receptores Adrenérgicos alfa/metabolismo , Yohimbina/farmacología
18.
Shock ; 53(4): 476-484, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31232864

RESUMEN

Severely burned patients suffer from a hypermetabolic syndrome that can last for years after the injury has resolved. The underlying cause of these metabolic alterations most likely involves the persistent elevated catecholamine levels that follow the surge induced by thermal injury. At the cellular level, endoplasmic reticulum (ER) stress in metabolic tissues is a hallmark observed in patients following burn injury and is associated with several detrimental effects. Therefore, ER stress could be the underlying cellular mechanism of persistent hypermetabolism in burned patients. Here, we show that catecholamines induce ER stress and that adreno-receptor blockers reduce stress responses in the HepG2 hepatocyte cell line. Our results also indicate that norepinephrine (NE) significantly induces ER stress in HepG2 cells and 3T3L1 mouse adipocytes. Furthermore, we demonstrate that the alpha-1 blocker, prazosin, and beta blocker, propranolol, block ER stress induced by NE. We also show that the effects of catecholamines in inducing ER stress are cell type-specific, as NE treatment failed to evoke ER stress in human fibroblasts. Thus, these findings reveal the mechanisms used by catecholamines to alter metabolism and suggest inhibition of the receptors utilized by these agents should be further explored as a potential target for the treatment of ER stress-mediated disease.


Asunto(s)
Catecolaminas/fisiología , Estrés del Retículo Endoplásmico/fisiología , Fibroblastos/fisiología , Células Hep G2/fisiología , Receptores Adrenérgicos alfa/fisiología , Receptores Adrenérgicos beta/fisiología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas Adrenérgicos beta/farmacología , Técnicas de Cultivo de Célula , Fibroblastos/efectos de los fármacos , Células Hep G2/efectos de los fármacos , Humanos , Prazosina/farmacología , Propranolol/farmacología
19.
Neuropharmacology ; 164: 107912, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31843397

RESUMEN

Post-traumatic stress disorder (PTSD) is a severe chronic mental illness that develops in individuals exposed to life-threatening trauma and is characterized by hyperarousal, flashbacks and nightmares. The serotonergic (5-HT) and noradrenergic (NE) systems are deeply involved in the pathogenesis of PTSD. We have previously reported a novel anxiolytic compound, ACH-000029, that modulates 5-HT and α1-adrenergic receptors and induces acute anxiolytic-like effects in rodents. Here, we investigated the potential of ACH-000029 to prevent anxiety-like behavior in the single prolonged stress (SPS) PTSD model. Mice were subjected to the SPS procedure, followed by a 7-day treatment with ACH-000029 and, for comparison, with the α1-adrenergic antagonist prazosin. Animals were behaviorally assessed using social interaction, elevated plus maze and open field tests. Interestingly, treatment with ACH-000029 but not with prazosin ameliorated the SPS-induced sociability impairment and anxiety-like behavior. The brain-wide c-fos mapping, used as a surrogate for brain activity, indicated the brain structures that were altered by SPS and putatively involved in the anxiolytic-like effect of ACH-000029. The SPS protocol produced long-lasting impairment of regions involved in stress-anxiety response, such as the amygdala, prefrontal cortex, globus pallidus and superior colliculus. ACH-000029 treatment reversed the SPS-induced c-fos changes in the globus pallidus, lateral septum and entorhinal cortex and exclusively modulated c-fos levels in subregions from the retrosplenial cortex, cerebellum, superior colliculus and ventromedial hypothalamus. These results support the hypothesis that the dual regulation of 5-HT and α1-adrenergic receptors is required to alleviate PTSD symptoms and suggest a possible role of ACH-000029 as a PTSD treatment.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Piperazinas/farmacología , Quinazolinas/farmacología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Serotoninérgicos/farmacología , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/psicología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Ansiolíticos/uso terapéutico , Química Encefálica/efectos de los fármacos , Genes fos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/uso terapéutico , Prazosina/farmacología , Quinazolinas/uso terapéutico , Serotoninérgicos/uso terapéutico , Interacción Social , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología
20.
Auton Neurosci ; 222: 102588, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31669796

RESUMEN

The vas deferens responds to a single electrical pulse with a biphasic contraction caused by cotransmitters ATP and noradrenaline. Removing Mg2+ (normally 1.2 mM) from the physiological salt solution (PSS) enhances the contraction. This study aimed to determine the effect of Mg2+ concentration on nerve cotransmitter-mediated contractions. Rat vasa deferentia were sequentially bathed in increasing (0, 1.2, 3 mM) or decreasing (3, 1.2, 0 mM) Mg2+ concentrations. At each concentration a single field pulse was applied, and the biphasic contraction recorded. Contractions to exogenous noradrenaline 10 µM and ATP 100 µM were also determined. The biphasic nerve-mediated contraction was elicited by ATP and noradrenaline as NF449 (10 µM) and prazosin (100 nM) completely prevented the respective peaks. Taking the contractions in normal PSS (Mg2+ 1.2 mM) as 100%, lowering Mg2+ to 0 mM enhanced the ATP peak to 170 ±â€¯7% and raising Mg2+ to 3 mM decreased it to 39 ±â€¯3%; the noradrenaline peak was not affected by lowering Mg2+ to 0 mM (97 ±â€¯3%) but was decreased to 63 ±â€¯4% in high Mg2+ (3 mM). Contractions to exogenous ATP, but not noradrenaline, were increased in Mg2+ 0 mM and both were inhibited with Mg2+ 3 mM. Changing Mg2+ concentration affects the contractions elicited by the cotransmitters ATP and noradrenaline. The greatest effects were to potentiate the contraction to ATP in Mg2+ 0 mM and to inhibit the contraction to both ATP and noradrenaline in high Mg2+. Future publications should clearly justify any decision to vary the magnesium concentration from normal (1.2 mM) values.


Asunto(s)
Adenosina Trifosfato/metabolismo , Sistema Nervioso Autónomo/fisiología , Fenómenos Electrofisiológicos/fisiología , Magnesio/farmacología , Contracción Muscular/fisiología , Norepinefrina/metabolismo , Conducto Deferente/fisiología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/metabolismo , Bencenosulfonatos/farmacología , Cationes Bivalentes/farmacología , Fenómenos Electrofisiológicos/efectos de los fármacos , Masculino , Contracción Muscular/efectos de los fármacos , Prazosina/farmacología , Ratas , Ratas Sprague-Dawley , Conducto Deferente/efectos de los fármacos , Conducto Deferente/inervación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA