Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(13)2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-39000340

RESUMO

Most α2-AR agonists derived from dexmedetomidine have few structural differences between them and have no selectivity for α2A/2B-AR or Gi/Gs, which can lead to side effects in drugs. To obtain novel and potent α2A-AR agonists, we performed virtual screening for human α2A-AR and α2B-AR to find α2A-AR agonists with higher selectivity. Compound P300-2342 and its three analogs significantly decreased the locomotor activity of mice (p < 0.05). Furthermore, P300-2342 and its three analogs inhibited the binding of [3H] Rauwolscine with IC50 values of 7.72 ± 0.76 and 12.23 ± 0.11 µM, respectively, to α2A-AR and α2B-AR. In α2A-AR-HEK293 cells, P300-2342 decreased forskolin-stimulated cAMP production without increasing cAMP production, which indicated that P300-2342 activated α2A-AR with coupling to the Gαi/o pathway but without Gαs coupling. P300-2342 exhibited no agonist but slight antagonist activities in α2B-AR. Similar results were obtained for the analogs of P300-2342. The docking results showed that P300-2342 formed π-hydrogen bonds with Y394, V114 in α2A-AR, and V93 in α2B-AR. Three analogs of P300-2342 formed several π-hydrogen bonds with V114, Y196, F390 in α2A-AR, and V93 in α2B-AR. We believe that these molecules can serve as leads for the further optimization of α2A-AR agonists with potentially few side effects.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2 , Simulação de Acoplamento Molecular , Receptores Adrenérgicos alfa 2 , Humanos , Animais , Células HEK293 , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Camundongos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , AMP Cíclico/metabolismo , Masculino , Ligação Proteica
2.
Artigo em Inglês | MEDLINE | ID: mdl-38916640

RESUMO

RATIONALE: The phenylalkylamine hallucinogen (-)-2,5-dimethoxy-4-methylamphetamine (DOM) exhibits an inverted U-shaped dose-response curve for both head twitch response (HTR) and locomotor activity in mice. Accumulated studies suggest that HTR and locomotor hyperactivity induced by DOM are mainly caused by the activation of serotonin 5-hydroxytryptamine 2 A receptor (5-HT2A receptor). However, the mechanisms underlying the biphasic dose response of HTR and locomotor activity induced by DOM, particularly at high doses, remain unclear. OBJECTIVES: The primary objective of this study is to investigate the modulation of 5-HT2A/2C/1A receptors in HTR and locomotor activity, while also exploring the potential receptor mechanisms underlying the biphasic dose response of DOM. METHODS: In this study, we employed pharmacological methods to identify the specific 5-HT receptor subtypes responsible for mediating the biphasic dose-response effects of DOM on HTR and locomotor activity in C57BL/6J mice. RESULTS: The 5-HT2A receptor selective antagonist (R)-[2,3-di(methoxy)phenyl]-[1-[2-(4-fluorophenyl)ethyl]piperidin-4-yl]methanol (M100907) (500 µg/kg, i.p.) fully blocked the HTR at every dose of DOM (0.615-10 mg/kg, i.p.) in C57BL/6J mice. M100907 (50 µg/kg, i.p.) decreased the locomotor hyperactivity induced by a low dose of DOM (0.625, 1.25 mg/kg, i.p.), but had no effect on the locomotor hypoactivity induced by a high dose of DOM (10 mg/kg) in C57BL/6J mice. The 5-HT2C antagonist 6-chloro-5-methyl-1-[(2-[2-methylpyrid-3yloxy]pyrid-5yl)carbamoyl]indoline (SB242084) (0.3, 1 mg/kg, i.p.) reduced the HTR induced by a dose of 2.5 mg/kg DOM, but did not affect the response to other doses. SB242084 (1 mg/kg, i.p.) significantly increased the locomotor activity induced by DOM (0.615-10 mg/kg, i.p.) in mice. The 5-HT1A antagonist N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]N-(2-pyridinyl) cyclohexane carboxamide maleate (WAY100635) (1 mg/kg, i.p.) increased both HTR and locomotor activity induced by DOM in mice. The 5-HT1A agonist 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) (1 mg/kg, i.p.) significantly reduced both the HTR and locomotor activity induced by DOM in mice. Additionally, pretreatment with the Gαi/o inhibitor PTX (0.25 µg/mouse, i.c.v.) enhanced the HTR induced by DOM and attenuated the effect of DOM on locomotor activity in mice. CONCLUSIONS: Receptor subtypes 5-HT2C and 5-HT1A are implicated in the inverted U-shaped dose-response curves of HTR and locomotor activity induced by DOM in mice. The biphasic dose-response function of HTR and locomotor activity induced by DOM has different mechanisms in mice.

3.
J Med Chem ; 67(13): 11435-11449, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38889119

RESUMO

Enhancing the selectivity of alpha2-adrenoceptor (α2A-AR) agonists remains an unresolved issue. Herein, we reported the design of an α2A-AR agonist using the conformation constraint method, beginning with medetomidine. The structure-activity relationship indicated that the 8-substituent of chromane derivatives exerted the most pronounced effect on α2A-AR agonistic activity. Compounds A9 and B9 were identified as the most promising, exhibiting EC50 values of 0.78 and 0.23 nM, respectively. Their selectivity indexes surpassed dexmedetomidine (DMED) by 10-80 fold. In vivo studies demonstrated that both A9 and B9 dose-dependently increased the loss of righting reflex in mice, with ED50 values of 1.54 and 0.138 mg/kg, respectively. Binding mode calculations and mutation studies suggested the indispensability of the hydrogen bond between ASP1283.32 and α2A-AR agonist. In particular, A9 and B9 showed no dual reverse pharmacological effect, a characteristic exhibited by DMED in α2A-AR activation.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2 , Cromanos , Desenho de Fármacos , Receptores Adrenérgicos alfa 2 , Animais , Receptores Adrenérgicos alfa 2/metabolismo , Cromanos/farmacologia , Cromanos/química , Cromanos/síntese química , Relação Estrutura-Atividade , Camundongos , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/química , Humanos , Conformação Molecular , Simulação de Acoplamento Molecular , Masculino
4.
Molecules ; 29(5)2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38474611

RESUMO

The α2A adrenergic receptor (α2A-AR) serves as a critical molecular target for sedatives and analgesics. However, α2A-AR ligands with an imidazole ring also interact with an imidazoline receptor as well as other proteins and lead to undesirable effects, motivating us to develop more novel scaffold α2A-AR ligands. For this purpose, we employed an ensemble-based ligand discovery strategy, integrating long-term molecular dynamics (MD) simulations and virtual screening, to identify new potential α2A-AR agonists with novel scaffold. Our results showed that compounds SY-15 and SY-17 exhibited significant biological effects in the preliminary evaluation of protein kinase A (PKA) redistribution assays. They also reduced levels of intracellular cyclic adenosine monophosphate (cAMP) in a dose-dependent manner. Upon treatment of the cells with 100 µM concentrations of SY-15 and SY-17, there was a respective decrease in the intracellular cAMP levels by 63.43% and 53.83%. Subsequent computational analysis was conducted to elucidate the binding interactions of SY-15 and SY-17 with the α2A-AR. The binding free energies of SY-15 and SY-17 calculated by MD simulations were -45.93 and -71.97 kcal/mol. MD simulations also revealed that both compounds act as bitopic agonists, occupying the orthosteric site and a novel exosite of the receptor simultaneously. Our findings of integrative computational and experimental approaches could offer the potential to enhance ligand affinity and selectivity through dual-site occupancy and provide a novel direction for the rational design of sedatives and analgesics.


Assuntos
Analgésicos , Receptores Adrenérgicos alfa 2 , Ligantes , Receptores Adrenérgicos alfa 2/metabolismo , Hipnóticos e Sedativos
5.
Eur J Pharmacol ; 957: 176038, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37657742

RESUMO

AIMS: (-)-2,5-dimethoxy-4-methylamphetamine (DOM) induces the head-twitch response (HTR) primarily by activating the serotonin 5-hydroxytryptamine 2A receptor (5-HT2A receptor) in mice. However, the mechanisms underlying 5-HT2A receptor activation and the HTR remain elusive. Gßγ subunits are a potential treatment target in numerous diseases. The present study investigated the mechanism whereby Gßγ subunits influence DOM-induced HTR. MAIN METHODS: The effects of the Gßγ inhibitor 3',4',5',6'-tetrahydroxyspiro[2-benzofuran-3,9'-xanthene]-1-one (gallein) and antagonistic peptide ßARKct (ß-adrenergic receptor kinase C-terminal fragment) on DOM-induced HTR were studied via an HTR test. The activation of the phospholipase C ß (PLCß)/inositol triphosphate (IP3)/calcium (Ca2+) signaling pathway and extracellular signal-regulated kinase (ERK) following Gßγ subunit inhibition was detected by western blotting, Homogeneous Time-Resolved Fluorescence (HTRF) inositol phosphate (IP1) assay and Fluorometric Imaging Plate Reader (FLIPR) calcium 6 assay. The Gßγ subunit-mediated regulation of cyclic adenosine monophosphate (cAMP) was assessed via a GloSensor™ cAMP assay. KEY FINDINGS: The Gßγ subunit inhibitors gallein and ßARKct reduced DOM-induced HTR in C57BL/6J mice. Like the 5-HT2A receptor-selective antagonist (R)-[2,3-di(methoxy)phenyl]-[1-[2-(4-fluorophenyl)ethyl]piperidin-4-yl]methanol (M100907), gallein inhibited PLCß phosphorylation (pPLCß), IP1 production, Ca2+ transients, ERK1/2 phosphorylation (pERK1/2) and cAMP accumulation induced by DOM in human embryonic kidney (HEK) 293T cells stably or transiently transfected with the human 5-HT2A receptor. Moreover, PLCß protein inhibitor 1-[6-[[(8R,9S,13S,14S,17S)-3-methoxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-17-yl]amino]hexyl]pyrrole-2,5-dione (U73122) (10 nmol/mouse), intracellular Ca2+ blocker 6-[6-[6-[5-acetamido-4,6-dihydroxy-2-(sulfooxymethyl)oxan-3-yl]oxy-2-carboxy-4-hydroxy-5-sulfooxyoxan-3-yl]oxy-2-(hydroxymethyl)-5-(sulfoamino)-4-sulfooxyoxan-3-yl]oxy-3,4-dihydroxy-5-sulfooxyoxane-2-carboxylic acid (heparin) (5 nmol/mouse), L-type Ca2+ channel blocker 3-O-(2-methoxyethyl) 5-O-propan-2-yl 2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3,5-dicarboxylate (nimodipine) (4 mg/kg), mitogen extracellular regulating kinase 1/2 (MEK1/2) inhibitor (Z)-3-amino-3-(4-aminophenyl)sulfanyl-2-[2-(trifluoromethyl)phenyl]prop-2-enenitrile (SL327) (30 mg/kg), and Gαs protein selective antagonist 4,4',4″,4‴-(Carbonylbis-(imino-5,1,3-benzenetriylbis(carbonylimino)))tetrakisbenzene-1,3-disulfonic acid (NF449) (10 nmol/mouse) reduced DOM-induced HTR in C57BL/6J mice. SIGNIFICANCE: The Gßγ subunits potentially mediate the HTR after 5-HT2A receptor activation via the PLCß/IP3/Ca2+/ERK1/2 and cAMP signaling pathways. Inhibitors targeting the Gßγ subunits potentially inhibit the hallucinogenic effects of 5-HT2A receptor agonists.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular , Receptor 5-HT2A de Serotonina , Humanos , Animais , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipase C beta , Cálcio , Transdução de Sinais
6.
Mol Pharmacol ; 100(2): 170-180, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34031190

RESUMO

Opioids play an important role in pain relief, but repeated exposure results in tolerance and dependence. To make opioids more effective and useful, research in the field has focused on reducing the tolerance and dependence for chronic pain relief. Here, we showed the effect of A20-binding inhibitor of nuclear factor-κB (ABIN-1) in modulating morphine function. We used hot-plate tests and conditioned place preference (CPP) tests to show that overexpression of ABIN-1 in the mouse brain attenuated morphine dependence. These effects of ABIN-1 are most likely mediated through the formation of ABIN-1-ß-arrestin2 complexes, which accelerate ß-arrestin2 degradation by ubiquitination. With the degradation of ß-arrestin2, ABIN-1 overexpression also decreased µ opioid receptor (MOR) phosphorylation and internalization after opioid treatment, affecting the ß-arrestin2-dependent signaling pathway to regulate morphine tolerance. Importantly, the effect of ABIN-1 on morphine tolerance was abolished in ß-arrestin2-knockout mice. Taken together, these results suggest that the interaction between ABIN-1 and ß-arrestin2 inhibits MOR internalization to attenuate morphine tolerance, revealing a novel mechanism for MOR regulation. Hence, ABIN-1 may be a therapeutic target to regulate MOR internalization, thus providing a foundation for a novel treatment strategy for alleviating morphine tolerance and dependence. SIGNIFICANCE STATEMENT: A20-binding inhibitor of nuclear factor-κB (ABIN-1) overexpression in the mouse brain attenuated morphine tolerance and dependence. The likely mechanism for this finding is that ABIN-1-ß-arrestin2 complex formation facilitated ß-arrestin2 degradation by ubiquitination. ABIN-1 targeted ß-arrestin2 to regulate morphine tolerance. Therefore, the enhancement of ABIN-1 is an important strategy to prevent morphine tolerance and dependence.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Encéfalo/metabolismo , Transtornos Relacionados ao Uso de Opioides/genética , Receptores Opioides mu/metabolismo , beta-Arrestina 2/metabolismo , Animais , Animais Geneticamente Modificados , Células CHO , Células , Cricetulus , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Transtornos Relacionados ao Uso de Opioides/metabolismo , Fosforilação , Proteólise , Ubiquitinação , beta-Arrestina 2/química , beta-Arrestina 2/genética
7.
Brain Res ; 1748: 147083, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32871137

RESUMO

Thienorphine hydrochloride is a new anti-relapse drug for opioid abusers that is currently in phase II clinical trial. In the present study, the antinociception, dependence, and signal transduction induced by thienorphine were examined. Thienorphine showed a potent antinociception effect in acetic acid-induced writhing test and formalin test. In the hot plate test and tail-flick test, thienorphine presented the typical partial opioid agonist character with a ceiling dose-response curve in addition to a bell-shaped curve. The hot plate test revealed that thienorphine induced approximately 50% of antinociception in µ receptor knockout (µ-KO) mice compared to wild-type controls (P < 0.05). The κ, δ selective antagonist nor-binaltorphimine (nor-BNI), and naltrindole decreased approximately 50-60% of theinorphine antinociception in µ-KO mice, respectively. The ORL1 receptor-selective antagonist J113397 did not affect theinorphine antinociception in µ-KO mice. Chronic treatment with thienorphine (1.5 mg/kg) induced some tolerance that was lower compared to buprenorphine or morphine addition. In contrast to buprenorphine or morphine, thienorphine did not lead to psychological dependence by conditioned place preference (CPP). The maximum inhibition of thienorphine on protein kinase A (PKA) activity was about 36%, 100%, 100%, and 12% in CHO-µ/κ/δ/ORL1-PKAcatEGFP cells, respectively. Similar results were observed in cyclic adenosine monophosphate (cAMP) accumulation inhibited by thienorphine in cells. Thienorphine significantly increased pERK1/2 in CHO-κ/δ-PKAcatEGFP cells. These results indicated that thienorphine induced analgesia through activation of κ- and δ-, partial activation of µ- opioid receptor without a bias between G-protein- and ß-arrestin-mediated pathways. Thienorphine might be used for antinociception with minimal adverse effects.


Assuntos
Analgésicos Opioides/uso terapêutico , Encéfalo/efeitos dos fármacos , Buprenorfina/análogos & derivados , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Analgésicos Opioides/farmacologia , Animais , Encéfalo/metabolismo , Buprenorfina/farmacologia , Buprenorfina/uso terapêutico , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Dor/genética , Dor/metabolismo , Ratos , Ratos Wistar , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
8.
Life Sci ; 252: 117676, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32304763

RESUMO

AIMS: Many µ-opioid receptor (MOR)-associated proteins can regulate the MOR signaling pathway. Using a bacterial two-hybrid screen, we found that the C-terminal of the MOR associated with heat shock protein 90 isoform ß (Hsp90ß). Here, we explored the effect of Hsp90ß on MOR signaling transduction and function. MAIN METHODS: The interaction of Hsp90ß with MOR was detected by co-immunoprecipitation and immunofluorescence. The effects of Hsp90ß on MOR signaling induced by opioids were studied in vitro and in vivo. The effects of the Hsp90ß inhibitor 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) on morphine tolerance and dependence were studied via a hot plate test and CPP test. KEY FINDINGS: Hsp90ß, instead of Hsp90α, interacted with the MOR in HEK293 cells and SH-SY5Y cells, and the interaction was augmented after morphine pretreatment. The interaction of Hsp90ß and MOR increased the inhibition of cAMP and decreased PKA activity under opioid treatment. The functional Hsp90ß-MOR complex also promoted the phosphorylation and internalization of the MOR induced by DAMGO in MOR-CHO cells. 17-AAG blocked Hsp90ß-MOR interactions and decreased the effect of Hsp90ß on the MOR signal transduction. In C57BL/6 mice, 17-AAG decreased morphine-induced acute anti-nociception in the hot plate test, with an increase in phosphorylated PKA and phosphorylated JNK and a decrease in phosphorylated CREB and phosphorylated ERK in murine brains. Chronic morphine treatment induced tolerance, and dependence was inhibited by 17-AAG co-administration. SIGNIFICANCE: Hsp90ß is a positive co-regulator of the MOR via the activation of a G-protein-dependent and ß-arrestin-dependent pathway. Hsp90ß has the potential to improve the pharmacologic profile of existing opiates. It is conceivable that in future clinical treatments, the Hsp90ß inhibitor, 17-AAG, could decrease the tolerance and dependence in cancer patients induced by opioids.


Assuntos
Analgésicos Opioides/farmacologia , Benzoquinonas/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Lactamas Macrocíclicas/farmacologia , Morfina/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Analgésicos Opioides/administração & dosagem , Animais , Células CHO , Cricetinae , Cricetulus , Tolerância a Medicamentos , Feminino , Células HEK293 , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Nociceptividade/efeitos dos fármacos , Receptores Opioides mu/metabolismo
9.
Eur J Pharmacol ; 831: 28-37, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29738700

RESUMO

Dexmedetomidine (DMED) is a potent and highly selective α2-adrenergic receptor agonist and is widely used for short-term sedation. However, the mechanism of DMED-induced sedation has not been deciphered. In the present study, we investigated the mechanism of Gαi and Gßγ subunits on DMED-induced sedation. An ED50 of DMED-induced loss of righting reflex (200.0nmol/kg) was increased to 375.0 or 433.3nmol/kg after pre-treatment with cAMP analog dbcAMP (50nmol/5 µl/mouse, i.c.v.) or the phosphodiesterase 4 inhibitor rolipram (100nmol/5 µl/mouse, i.c.v.). Conversely, the ED50 of DMED-induced LORR decreased to 113.6 or 136.5 nmol/kg after pre-treated with Gßγ subunit inhibitor M119 (100 mg/kg, i.p.) or gallein (100 mg/kg, i.p.) respectively. Administration of dbcAMP, rolipram, gallein or M119 alone had no effect on LORR. Gallein (10 µM) significantly inhibited forskolin-stimulated cAMP accumulation in α2A-AR -CHO cells. Compared with Gßγ subunit inhibitors or DMED alone, [Ca2+]i and pERK1/2 was significantly increased after co-administration with Gßγ subunit inhibitors and DMED. DbcAMP (5 µM) or rolipram (5 µM) alone had no effect on ERK1/2 phosphorylation, but decreased DMED-induced ERK1/2 phosphorylation after co-administration with DMED. Gßγ subunit inhibitor treatment increased DMED-induced phosphorylation of CREB, whereas dbcAMP or rolipram had no effect on pCREB induced by DMED. From our results we conclude that, Gßγ subunit may inhibit DMED-induced sedation through the cAMP and pERK1/2 pathway.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Comportamento Animal/efeitos dos fármacos , Estado de Consciência/efeitos dos fármacos , Dexmedetomidina/farmacologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/efeitos dos fármacos , Subunidades beta da Proteína de Ligação ao GTP/efeitos dos fármacos , Subunidades gama da Proteína de Ligação ao GTP/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Animais , Bucladesina/farmacologia , Células CHO , Cricetulus , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Masculino , Camundongos , Inibidores da Fosfodiesterase 4/farmacologia , Fosforilação , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Reflexo de Endireitamento/efeitos dos fármacos , Rolipram/farmacologia , Transdução de Sinais/efeitos dos fármacos
10.
Int Urol Nephrol ; 50(5): 983-991, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29368245

RESUMO

BACKGROUND: Podocyte damage exerts a key role in proteinuria. We have demonstrated that calcineurin-binding protein 1 (Cabin1) upregulated during podocyte injury, yet its function in podocyte is still unclear. METHODS: We established 5/6 nephrectomized rats and angiotensin II (AngII)-injured podocyte, as well as knocked down Cabin1 with siRNA in cultured podocytes. Rats were killed at 4 or 8 weeks after 5/6 nephrectomy. The localization of podocyte cytoskeleton was detected after immunofluorescence staining. Podocyte mitochondrial morphology was observed under electron microscopy. Podocyte mitochondrial transmembrane potential (MMP) was measured with MitoCapture kit. Cabin1 and cytochrome c protein expression were detected by western blot. RESULTS: Massive proteinuria, as well as obvious segmental glomerular sclerosis, was found in rats at 8 weeks after nephrectomy, accompanied with the disruption of synaptopodin. Moreover, mitochondria changed from large and ellipsoid shape to the small, long, and irregular shape in rats at 4 weeks after operation. At 8 weeks, mitochondria were swollen and cristae were remarkably dissolved. Compared to sham-operated rats, Cabin1 protein expression was obviously upregulated in rats at 8 weeks. AngII induced the decrease in MMP, as well as the overexpression of Cabin1 and cytochrome c protein in podocytes. Knocking down Cabin1 induced the disruption of F-actin and overexpression of cytochrome c (1.81 ± 0.21 in siRNA group vs. 0.86 ± 0.11 in negative control group). CONCLUSIONS: Knocking down Cabin1 induces the disruption of cytoskeleton and mitochondrial dysfunction in podocyte. Cabin1 could be a crucial factor in podocyte damage.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Citocromos c/metabolismo , Mitocôndrias/patologia , Mitocôndrias/fisiologia , Nefrectomia , Podócitos/patologia , Actinas/metabolismo , Angiotensina II/farmacologia , Animais , Proteínas Reguladoras de Apoptose/genética , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/genética , Técnicas de Silenciamento de Genes , Potenciais da Membrana/efeitos dos fármacos , Nefrectomia/métodos , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Proteinúria/genética , RNA Interferente Pequeno , Ratos , Regulação para Cima/efeitos dos fármacos , Vasoconstritores/farmacologia
11.
Mol Pharmacol ; 93(2): 36-48, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29237725

RESUMO

The µ-opioid receptor (MOR) is a Gi/o protein-coupled receptor that mediates analgesic, euphoric, and reward effects. Using a bacterial two-hybrid screen, we reported that the carboxyl tail of the rat MOR associates with A20-binding inhibitor of nuclear factor κB (ABIN-1). This interaction was confirmed by direct protein-protein binding and coimmunoprecipitation of MOR and ABIN-1 proteins in cell lysates. Saturation binding studies showed that ABIN-1 had no effect on MOR binding. However, the interaction of ABIN-1 and MOR inhibited the activation of G proteins induced by DAMGO ([d-Ala2,N-Me-Phe4,Gly5-ol]-Enkephalin). MOR phosphorylation, ubiquitination, and internalization induced by DAMGO were decreased in Chinese hamster ovary cells that coexpressed MOR and ABIN-1. The suppression of forskolin-stimulated adenylyl cyclase by DAMGO was also inhibited by the interaction of ABIN-1 with MOR. In addition, extracellular signal-regulated kinase activation was also negatively regulated by overexpression of ABIN-1. These data suggest that ABIN-1 is a negative coregulator of MOR activation, phosphorylation, and internalization in vitro. ABIN-1 also inhibited morphine-induced hyperlocomotion in zebrafish larvae (AB strain). By utilization of an antisense morpholino oligonucleotide (MO) gene knockdown technology, the ABIN-1 MO-injected zebrafish larvae showed a significant increase (approximately 60%) in distance moved compared with control MO-injected larvae after acute morphine treatment (P < 0.01). Taken together, ABIN-1 negatively regulates MOR function in vitro and in vivo.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Receptores Opioides mu/fisiologia , Analgésicos Opioides/farmacologia , Animais , Sítios de Ligação , Células CHO , Linhagem Celular Tumoral , Cricetulus , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endocitose , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Larva , Ligantes , Proteínas Luminescentes , Fosforilação , Ratos , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Transdução de Sinais , Radioisótopos de Enxofre/metabolismo , Ubiquitinação , Peixe-Zebra/crescimento & desenvolvimento
12.
Iran J Kidney Dis ; 11(6): 430-437, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29190603

RESUMO

INTRODUCTION: Calcineurin-binding protein 1 (Cabin1) interacts with calcineurin and p53, but its function in renal tubular epithelial cell (RTEC) is unclear. We established 5/6 nephrectomized rats and angiotensin II-induced injury to the RTECs in vitro, to observe the expression of Cabin1 during RTEC injury. MATERIALS AND METHODS: Sprague-Dawley rats were sacrificed at 4 and 8 weeks after 5/6 nephrectomy. Renal pathology and mitochondrial damage were detected by light and electrical microscope. The distribution of E-cadherin and α-smad were detected by indirect immunofluorescence staining. Cabin1 protein expression was detected by Western blot. RESULTS: Obvious tubulointerstitial fibrosis was found in the nephrectomized rats at 8 weeks after 5/6 nephrectomy, accompanied by the increasing levels of creatinine, as well as the disruption of E-cadherin and overexpression of α-smad in RTECs. Moreover, the mitochondria became swollen and mitochondrial cristae were disrupted and poorly defined in the RTECs. Compared to the sham-operated rats, Cabin1 protein expression was significantly increased at 8 weeks after 5/6 nephrectomy, while angiotensin II-induced Cabin1 protein expression significantly increased 48 hours after stimulation in normal rat kidney epithelial cells. CONCLUSIONS: Injury to the RTEC and Cabin1 protein overexpression occurred in a time-dependent manner both in vitro and in vivo. Cabin1 may become a potential molecular target in RTEC injury.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Células Epiteliais/metabolismo , Nefropatias/metabolismo , Túbulos Renais/metabolismo , Angiotensina II/toxicidade , Animais , Caderinas/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/ultraestrutura , Fibrose , Nefropatias/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/ultraestrutura , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Dilatação Mitocondrial , Nefrectomia , Ratos Sprague-Dawley , Proteínas Smad/metabolismo , Fatores de Tempo , Regulação para Cima
13.
Acta Pharmacol Sin ; 38(7): 977-989, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28502978

RESUMO

Opioid analgesics remain the first choice for the treatment of moderate to severe pain, but they are also notorious for their respiratory depression and addictive effects. This study focused on the pharmacology of a novel opioid receptor mixed agonist DPI-125 and attempted to elucidate the relationship between the δ-, µ- and κ-receptor potency ratio and respiratory depression and abuse liability. Five diarylmethylpiperazine compounds (DPI-125, DPI-3290, DPI-130, KUST202 and KUST13T02) were selected for this study. PKA fluorescence redistribution assays in CHO cells individually expressing δ-, µ- or κ-receptors were used to measure the agonist potency. The respiratory safety profiles were estimated in rats by the ratio of ED50 (pCO2 increase)/ED50 (antinociception). The abuse liability of DPI-125 was evaluated with a self-administration model in rhesus monkeys. The observed agonist potencies of DPI-125 for δ-, µ- and κ-opioid receptors were 4.29±0.36, 11.10±3.04, and 16.57±4.14 nmol/L, respectively. The other four compounds were also mixed agonists with varying potencies. DPI-125 exhibited a high respiratory safety profile, clearly related to its high δ-receptor potency. The ratio of the EC50 potencies for the µ- and δ-receptors was found to be positively correlated with the respiratory safety ratio. DPI-125 has similar potencies for µ- and κ-receptors, which is likely the reason for its reduced abuse potential. Our results demonstrate that the opioid receptor mixed agonist DPI-125 is safer and less addictive than traditional µ-agonist analgesics. These findings suggest that the development of δ>µâˆ¼κ opioid receptor mixed agonists is feasible, and such compounds could represent a promising class of potent analgesics with wider therapeutic windows.


Assuntos
Analgesia , Analgésicos Opioides/farmacologia , Dor/tratamento farmacológico , Piperazinas/farmacologia , Insuficiência Respiratória/tratamento farmacológico , Tiofenos/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/química , Animais , Células CHO , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Masculino , Conformação Molecular , Medição da Dor , Piperazinas/administração & dosagem , Piperazinas/química , Ratos , Ratos Wistar , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Relação Estrutura-Atividade , Tiofenos/administração & dosagem , Tiofenos/química
14.
Eur J Pharmacol ; 789: 144-151, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27445237

RESUMO

Opioid analgesics are widely used in moderate to severe pain including renal colic. Morphine is believed to cause spasm of ureter and affect the bladder contractions. Thienorphine is a partial opioid agonist that is a good candidate for the treatment of opioid dependence and pain. This study examined the effects of thienorphine on the guinea pig isolated ureter and bladder. The contractile amplitude of isolated ureter induced by KCl (40mM) was not influenced by thienorphine or buprenorphine, whereas morphine increased the amplitude of the isolated ureter. Thienorphine, buprenorphine or naloxone concentration-dependently antagonized the isolated ureter contraction induced by morphine. Thienorphine (1.0-32.0µM) or buprenorphine (1.0-32.0µM) had no effects on the spontaneous or acetylcholine (Ach) induced contractions of isolated bladder, but decreased the amplitude of the contractions of isolated bladder at 100µM concentration. Morphine (0.1-3.2mM) concentration dependently increased the spontaneous movement and Ach (1µM) induced contractions of isolated bladder. The mRNA levels of µ receptor in the ureter and bladder was as the same as that in the frontal cortex. In comparison, the mRNA levels of κ receptor, δ receptor and N/OFQ receptor was fewer than that in the frontal cortex. In summary, thienorphine has little influence on the guinea pig isolated ureter and bladder compared with morphine, which may result in a lack of adverse renal colic effects.


Assuntos
Analgésicos/farmacologia , Buprenorfina/análogos & derivados , Contração Muscular/efeitos dos fármacos , Ureter/efeitos dos fármacos , Ureter/fisiologia , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/fisiologia , Animais , Buprenorfina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Cobaias , Masculino , Cloreto de Potássio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Opioides/genética , Ureter/metabolismo , Bexiga Urinária/metabolismo , Receptor de Nociceptina
15.
Ren Fail ; 38(4): 564-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26905810

RESUMO

Podocyte injury is a vital factor, which induces massive proteinuria. Studies have shown that tacrolimus (TAC) protected podocyte via stabilizing cytoskeleton. Our latest study indicates that calcineurin binding protein 1 (Cabin1) undergoes nuclear translocation during podocytes injury. Whether TAC targets on Cabin1 during podocyte injury is still not clear. This study establishes non-immunological proteinuric model. To observe the effect of the treatment of TAC on Cabin1 expression in 5/6 nephrectomized rats. Sprague-Dawley rats were injected with TAC (0.2 mg/kg/day) for 4-8 weeks after 5/6 nephrectomy. Then, rats were sacrificed in the eighth week after operation, renal tissues were processed for morphological studies under light and electrical microscope. Cabin1 expression and distribution were detected by western blot and indirect immunofluorescence staining. In 5/6 nephrectomized rats, urinary protein excretion reached 90.2 ± 30.1 mg/24 h, glomerular sclerosis index and tubulointerstitial fibrosis score were significantly increased, and widespread of podocyte foot processes fusion was found. Moreover, Cabin1 protein expression was markedly increased, and its distribution became much more obviously in podocytes nuclei. In TAC treated rats, urinary protein excretion significantly decreased (44.9 ± 22.5 mg/24 h), glomerular sclerosis and tubulointerstitial fibrosis were alleviated, and podocyte foot processes fusion was inhibited. Furthermore, TAC alleviated the increased protein expression and abnormal distribution of Cabin1. In conclusion, TAC restores podocyte injury and stabilizes the expression of Cabin1. Cabin1 may become a new target to demonstrate the mechanism of TAC in podocyte injury.


Assuntos
Oligopeptídeos/biossíntese , Oligopeptídeos/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Tacrolimo/farmacologia , Animais , Masculino , Nefrectomia/métodos , Ratos , Ratos Sprague-Dawley
16.
J Recept Signal Transduct Res ; 36(2): 173-80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26400065

RESUMO

Calcineurin binding protein 1 (Cabin1) is a natural inhibitor of calcineurin (CN). Moreover, Cabin1 retards tumor cell apoptosis by regulating p53. This study was designed to observe the expression of Cabin1 during podocyte injury, as well as its relationship with p53. Sprague-Dawley rats were used for the establishment of 5/6 nephrectomized rat model. Sham-operated rats underwent ventral laparotomy without nephrectomy. Then, rats were sacrificed at 8 and 12 weeks after nephrectomy. WT-1, a podocyte nuclear protein, was used for indicating the localization of Cabin1 in glomeruli. As tacrolimus protects podocyte via inhibiting AngiotensinII (AngII) induced CN activation. Cultured podocytes were injured by AngII or restored by tacrolimus. The protein expression and localization was detected by western blot or immunofluorescence staining. Cabin1 was knocked down by siRNA in cultured podocytes. In 5/6 nephrectomized rats, the colocalization of Cabin1 and WT-1 became more obviously in podocyte nuclei. Cabin1 protein was markedly increased in rats at 8 and 12 weeks after nephrectomy, as well as in AngII injured podocytes at 48 h (0.99 ± 0.12 in AngII group versus 0.80 ± 0.16 in control group). Cabin1 and p53 colocalized in cultured podocyte nuclei, p53 expression was significantly decreased (0.21 ± 0.05 in siRNA group versus 0.31 ± 0.05 in negative control group) after Cabin1 was being knocked down. In conclusion, Cabin1 expression significantly increases during podocyte injury. Knockdown of Cabin1 induces p53 expression decrease in cultured podocyte. Cabin1 may provide a new target to investigate podocyte injury.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Glomérulos Renais/metabolismo , Podócitos/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Angiotensina II/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Calcineurina/biossíntese , Calcineurina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Glomérulos Renais/ultraestrutura , Nefrectomia , Podócitos/patologia , RNA Interferente Pequeno , Ratos , Tacrolimo/administração & dosagem , Proteína Supressora de Tumor p53/genética , Proteínas WT1/genética , Proteínas WT1/metabolismo
17.
Life Sci ; 143: 156-67, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26522050

RESUMO

AIMS: Opioid addiction is associated with long-term adaptive changes in the brain that involve protein expression. The carboxyl-terminal of the µ opioid receptor (MOR-C) is important for receptor signal transduction under opioid treatment. However, the proteins that interact with MOR-C after chronic morphine exposure remain unknown. The brain cDNA library of chronic morphine treatment rats was screened using rat MOR-C to investigate the regulator of opioids dependence in the present study. MAIN METHODS: The brain cDNA library from chronic morphine-dependent rats was constructed using the SMART (Switching Mechanism At 5' end of RNA Transcript) technique. Bacterial two-hybrid system was used to screening the rat MOR-C interacting proteins from the cDNA library. RT-qPCR and immunoblotting were used to determine the variation of MOR-C interacting proteins in rat brain after chronic morphine treatment. Column overlay assays, immunocytochemistry and coimmunoprecipitation were used to demonstrate the interaction of MOR-C and p75NTR-associated cell death executor (NADE). KEY FINDINGS: 21 positive proteins, including 19 known proteins were screened to interact with rat MOR-C. Expression of several of these proteins was altered in specific rat brain regions after chronic morphine treatment. Among these proteins, NADE was confirmed to interact with rat MOR-C by in vitro protein-protein binding and coimmunoprecipitation in Chinese hamster ovary (CHO) cells and rat brain with or without chronic morphine treatment. SIGNIFICANCE: Understanding the rat MOR-C interacting proteins and the proteins variation under chronic morphine treatment may be critical for determining the pathophysiological basis of opioid tolerance and addiction.


Assuntos
Encéfalo/metabolismo , Biblioteca Gênica , Dependência de Morfina/metabolismo , Receptores Opioides mu/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Animais , Encéfalo/efeitos dos fármacos , Células CHO , Cricetinae , Cricetulus , Masculino , Morfina/administração & dosagem , Morfina/metabolismo , Ligação Proteica/fisiologia , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas
18.
Ren Fail ; 37(8): 1344-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26275115

RESUMO

CONTEXT: Podocyte injury is related to increasing proteinuria and contributes to the progression of kidney disease. Calcineurin binding protein 1 (Cabin1) is a repressor of myocyte enhancer factor 2 (MEF2) and calcineurin-mediated transcription in the immune system. Moreover, Cabin1 interacts with p53 and negatively regulates p53 in tumor cells. However, its function in kidney is unknown. OBJECTIVE: To explore the exact localization of Cabin1 in glomeruli, as well as the relationship between Cabin1 and podocyte injury. METHODS: Sprague-Dawley rats were sacrificed to observe the localization and protein expression of Cabin1 in the kidney. Cabin1 localization and protein expression were detected by immunofluorescence staining and western blot, respectively. Mouse podocytes were cultivated at 33 °C to propagate, then cells were transferred to an incubator at 37 °C to allow differentiation. Differentiated podocytes were stimulated by angiotensin II (AngII) or AngII plus tacrolimus. Cells were harvested to detect the localization and protein expression of Cabin1. Cytoplasmic and nuclear protein were separated by protein extraction kit. RESULTS: Cabin1 mainly localized in the nuclei of glomerular innate cells, it colocalized with WT-1 in podocytes nuclei. Western bolt showed Cabin1 protein remarkably expressed in renal cortex. AngII-induced Cabin1 nuclear protein significantly increased, accompanied by cytoskeleton disruption in cultured mouse podocytes. CONCLUSION: Cabin1 localizes in glomerular podocytes. AngII induces nuclear translocation of Cabin1 in cultured podocytes.


Assuntos
Angiotensina II/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Calcineurina/metabolismo , Glomérulos Renais/patologia , Podócitos/patologia , Animais , Células Cultivadas , Masculino , Camundongos , Proteinúria/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Risco
19.
Eur J Pharmacol ; 737: 22-8, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24830319

RESUMO

Opioid analgesics are widely believed to cause spasm of the bile duct sphincter and so impede bile flow. Thienorphine is a partial opioid agonist that is a good candidate for the treatment of opioid dependence; however, to date, no studies have reported the effects of thienorphine on the function of the biliary tract. This study examined the in vivo effects of thienorphine on the guinea pig isolated sphincter of Oddi, choledochus and gall bladder and on bile flow. The area under the curve (AUC) of isolated sphincter of Oddi was not influenced by thienorphine or buprenorphine, whereas morphine increased the AUC of the isolated sphincter of Oddi in a concentration-dependent manner. Thienorphine and buprenorphine concentration-dependently decreased the AUC of isolated choledochus, while morphine increased the AUC of isolated choledochus. Thienorphine had no effect on the contractile amplitude or basal tension of isolated gall bladder muscle strips. In contrast, buprenorphine and morphine increased the contractile basal tension of isolated gall bladder muscle strips in a concentration-dependent manner. Thienorphine (0.01-1.0mg/kg) had no significant inhibitory effect on bile flow. However, morphine (1.0-10mg/kg) and buprenorphine (1.0mg/kg) significantly inhibited bile flow. The maximum inhibition of bile flow by buprenorphine was 63.9±12.9% and by morphine was 74.1±11.3%. In summary, thienorphine has little influence on the guinea pig isolated sphincter of Oddi, choledochus and gall bladder or on bile flow, which may result in a lack of adverse biliary colic effects.


Assuntos
Buprenorfina/análogos & derivados , Ducto Colédoco/efeitos dos fármacos , Vesícula Biliar/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Esfíncter da Ampola Hepatopancreática/efeitos dos fármacos , Animais , Bile/metabolismo , Buprenorfina/farmacologia , Ducto Colédoco/fisiologia , Feminino , Vesícula Biliar/fisiologia , Cobaias , Masculino , Esfíncter da Ampola Hepatopancreática/fisiologia
20.
J. physiol. biochem ; 70(1): 185-191, mar. 2014.
Artigo em Inglês | IBECS | ID: ibc-121617

RESUMO

Mas-related G protein-coupled receptor D (MrgD) is expressed almost exclusively in nociceptive primary sensory neurons and the neurons located in stratum granulosum of skin. More and more evidence suggest that MrgD plays an important role in pain sensation and/or transduction. Recent studies have demonstrated that the receptor is also involved in itch sensation in both mouse and human. In the present study, we identified a robust inward current in MrgD-expressing Xenopusoocytes by using â-alanine, a putative ligand of MrgD. The currents were sensitive to inhibitor of Ca2+-activated chloride channels (CaCCs) and intracellular Ca2+ chelator, suggesting they were produced by endogenous CaCCs. Furthermore, it was demonstrated that upon the application of phospholipase C (PLC) inhibitor, or antisense oligonucleotides of inositol trisphosphate receptor (IP3R), the â-alanine-induced currents were dramatically depressed. However, protein kinase C inhibitor did not display any visible effect on CaCC currents. In summary, our data suggest that the activation of MrgD promotes the open of endogenous CaCCs via Gq-PLC-IP3-Ca2+ pathway. The current findings reveal the functional coupling between MrgD and CaCCs in Xenopus oocytes and also provide a facile model to assay the activity of MrgD


Assuntos
Animais , Xenopus , Dor/fisiopatologia , Bloqueadores dos Canais de Cálcio/farmacocinética , Substâncias Protetoras/farmacocinética , /métodos , Canais de Cloreto/antagonistas & inibidores , Receptores de Superfície Celular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...