Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Bioorg Chem ; 128: 105905, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35710525

RESUMEN

We identified, via high-throughput screening using a FLIPR® calcium assay, compound 1, which incorporated a dihydroquinolinyl-2-oxoethylsulfanyl-(1H,5H)-pyrimidinedione core and activated the µ-opioid receptor (MOR) in the presence of naloxone or naltrexone. A structure-activity relationship study of the analogs of 1 led to the design of compound 21, which activated MOR in the presence of naloxone with an EC50 of 3.3 ± 0.2 µM. MOR activation by the compound 21-antagonist pair was antagonist-dependent. Compound 21 did not affect the potency of the orthosteric agonist, morphine, toward MOR, indicating that it affected the function of MOR antagonists rather than that of the agonists. Computer modeling of the compound 21-MOR-naloxone complex revealed major interactions between compound 21 and MOR, including hydrogen bonding with Ser196, π-π stacking with Tyr149, and sulfur-aromatic interaction with Trp192. This study may pave the way for developing agents capable of safe and effective MOR modulation.


Asunto(s)
Naloxona , Naltrexona , Analgésicos Opioides , Imidazoles , Naloxona/farmacología , Naltrexona/farmacología , Receptores Opioides , Sulfonamidas , Tiofenos
2.
Biochim Biophys Acta Proteins Proteom ; 1865(3): 336-343, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28012872

RESUMEN

Naloxone is an alkaloid antagonist that acts as an antidote to opioids through the mu-opioid receptor (MOR), a G protein-coupled receptor. However, its binding site on the MOR remains unknown. To investigate the binding interfaces necessary for naloxone and MOR, available structural information was combined with a cell-based photocrosslinking approach. Computer prediction revealed that four binding sites on MOR were required for naloxone binding. In addition, in the photocrosslinking approach, an amber stop codon was used to replace the sense codon of the MOR at 266 selected individual positions, in order to introduce the photoreactive amino acid p-benzoyl-l-phenylalanine (BzF) into MOR to evaluate the results of the computer analysis. The BzF-incorporated MOR mutant genes were expressed in CHO cells, in which MOR retained the ability to interact with its ligands, such as morphine, and exhibited MOR-dependent activation of ERK signaling following morphine stimulation. Notably, after treatment with tritium-labeled naloxone and exposure to UV light, we observed naloxone crosslinking with BzF replacement at hydrophobic residues and some polar/uncharged residues in the computer-predicted sites 1 and 3, indicating that these two sites in the MOR interact with naloxone. In conclusion, these results indicate that MOR has two naloxone binding sites and that the hydrophobic and polar/uncharged residues within these sites are important for naloxone binding.


Asunto(s)
Reactivos de Enlaces Cruzados/metabolismo , Naloxona/metabolismo , Receptores Opioides mu/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Cricetulus , Ligandos , Sistema de Señalización de MAP Quinasas/fisiología , Transducción de Señal/fisiología
3.
Anesthesiology ; 126(5): 952-966, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28212204

RESUMEN

BACKGROUND: The authors investigated the pharmacology and signaling pathways of the opioid receptors modulated by compound 1, 1-(2,4-dibromophenyl)-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-one. METHODS: In vitro studies of compound 1 were assessed by using a radioligand-binding assay (n = 3), a cyclic adenosine monophosphate assay (n = 3), a ß-arrestin assay (n = 3), an internalization assay (n = 3), and an immunohistochemistry (n = 8). In vivo studies of compound 1 were characterized using a tail-flick test (n = 5 to 6), tail-clip test (n = 7), von Frey hair test (n = 5), and charcoal meal test (n = 5). RESULTS: Compound 1 elicited robust effects in µ-opioid (mean ± SD; binding affinity: 15 ± 2 nM; cyclic adenosine monophosphate assay: 24 ± 6 nM), δ-opioid (82 ± 7 nM; 1.9 ± 0.1 µM), and κ-opioid (76 ± 9 nM; 1.4 ± 0.5 µM) receptor-expressing cells. Compound 1 acts as a full agonist of ß-arrestin-2 recruitment in µ-opioid (1.1 ± 0.3 µM) and δ-opioid (9.7 ± 1.9 µM) receptor-expressing cells. Compound 1 caused less gastrointestinal dysfunction (charcoal meal test: morphine: 82 ± 5%; compound 1: 42 ± 5%) as well as better antinociception in mechanical pain hypersensitivity (tail-clip test: morphine: 10 ± 3 s; compound 1: 19 ± 1 s) and in cancer-induced pain (von Frey hair test: morphine: 0.1 ± 0.1 g; compound 1: 0.3 ± 0.1 g) than morphine at equi-antinociceptive doses. CONCLUSIONS: Compound 1 produced antinociception with less gastrointestinal dysfunction than morphine.


Asunto(s)
Enfermedades Gastrointestinales/inducido químicamente , Indazoles/farmacología , Morfina , Receptores Opioides/agonistas , Analgésicos Opioides/farmacología , Animales , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL
4.
J Biomed Sci ; 23(1): 81, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27863490

RESUMEN

BACKGROUND: Glioma stem-like cells (GSCs) are proposed to be responsible for high resistance in glioblastoma multiforme (GBM) treatment. In order to find new strategies aimed at reducing GSC stemness and improving GBM patient survival, we investigated the effects and mechanism of a histone deacetylases (HDACs) inhibitor, suberoylanilide hydroxamic acid (SAHA), since HDAC activity has been linked to cancer stem-like cell (CSC) abundance and properties. METHODS: Human GBM cell lines were plated in serum-free suspension cultures allowed for sphere forming and CSC enrichment. Subsequently, upon SAHA treatment, the stemness markers, cell proliferation, and viability of GSCs as well as cellular apoptosis and senescence were examined in order to clarify whether inhibition of GSCs occurs. RESULTS: We demonstrated that SAHA attenuated cell proliferation and diminished the expression stemness-related markers (CD133 and Bmi1) in GSCs. Furthermore, at high concentrations (more than 5 µM), SAHA triggered apoptosis of GSCs accompanied by increases in both activation of caspase 8- and caspase 9-mediated pathways. Interestingly, we found that a lower dose of SAHA (1 µM and 2.5 µM) inhibited GSCs via cell cycle arrest and induced premature senescence through p53 up-regulation and p38 activation. CONCLUSION: SAHA induces apoptosis and functions as a potent modulator of senescence via the p38-p53 pathway in GSCs. Our results provide a perspective on targeting GSCs via SAHA treatment, and suggest that SAHA could be used as a potent agent to overcome drug resistance in GBM patients.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/enzimología , Glioblastoma/genética , Glioblastoma/patología , Glioma/enzimología , Glioma/genética , Glioma/patología , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/genética
5.
Nucleic Acids Res ; 42(21): 13012-25, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361975

RESUMEN

Heterogeneous nuclear ribonucleoprotein K (hnRNP K) binds to the promoter region of mu-opioid receptor (MOR) to regulate its transcriptional activity. How hnRNP K contributes to the analgesic effects of morphine, however, is largely unknown. We provide evidence that morphine increases hnRNP K protein expression via MOR activation in rat primary cortical neurons and HEK-293 cells expressing MORs, without increasing mRNA levels. Using the bicistronic reporter assay, we examined whether morphine-mediated accumulation of hnRNP K resulted from translational control. We identified potential internal ribosome entry site elements located in the 5' untranslated regions of hnRNP K transcripts that were regulated by morphine. This finding suggests that internal translation contributes to the morphine-induced accumulation of hnRNP K protein in regions of the central nervous system correlated with nociceptive and antinociceptive modulatory systems in mice. Finally, we found that down-regulation of hnRNP K mediated by siRNA attenuated morphine-induced hyperpolarization of membrane potential in AtT20 cells. Silencing hnRNP K expression in the spinal cord increased nociceptive sensitivity in wild-type mice, but not in MOR-knockout mice. Thus, our findings identify the role of translational control of hnRNP K in morphine-induced analgesia through activation of MOR.


Asunto(s)
Regiones no Traducidas 5'/efectos de los fármacos , Analgésicos Opioides/farmacología , Ribonucleoproteína Heterogénea-Nuclear Grupo K/biosíntesis , Morfina/farmacología , Neuronas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Receptores Opioides mu/metabolismo , Animales , Secuencia de Bases , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Secuencia Conservada , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Ratones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Nocicepción , Ratas , Ribosomas/metabolismo , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Regulación hacia Arriba
6.
Bioorg Med Chem ; 22(17): 4694-703, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25087049

RESUMEN

The µ-opioid receptor (MOR) is the major opioid receptor targeted by most analgesics in clinical use. However, the use of all known MOR agonists is associated with severe adverse effects. We reported that the 1-phenyl-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-ones are novel opioid receptor agonists. Subsequent structural modification resulted in the potent MOR/KOR (κ-opioid receptor) agonists 19, 20, and 21. Testing the analgesic effect of these in WT B6 mice (tail-flick test) gave ED50 values of 8.4, 10.9, and 26.6mg/kg, respectively. The 1-phenyl-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-one core could be addressed in 1 or 2 synthetic steps with moderate to high percent of yield. In the adenylyl cyclase assay, compound 19 displayed a MOR/KOR agonist profile, with IC50 values of 0.73 and 0.41µM, respectively. Current results suggest that compound 19 is a promising lead to go further development and in vitro/in vivo adverse effects studies.


Asunto(s)
Analgésicos/farmacología , Descubrimiento de Drogas , Indazoles/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Analgésicos/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Indazoles/síntesis química , Indazoles/química , Ratones , Ratones Congénicos , Estructura Molecular , Dolor/tratamiento farmacológico , Dimensión del Dolor , Relación Estructura-Actividad , Cola (estructura animal)/efectos de los fármacos
7.
Cell Chem Biol ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39025070

RESUMEN

Morphinan antagonists, which block opioid effects at mu-opioid receptors, have been studied for their analgesic potential. Previous studies have suggested that these antagonists elicit analgesia with fewer adverse effects in the presence of the mutant mu-opioid receptor (MOR; S196A). However, introducing a mutant receptor for medical applications represents significant challenges. We hypothesize that binding a chemical compound to the MOR may elicit a comparable effect to the S196A mutation. Through high-throughput screening and structure-activity relationship studies, we identified a modulator, 4-(2-(4-fluorophenyl)-4-oxothiazolidin-3-yl)-3-methylbenzoic acid (BPRMU191), which confers agonistic properties to small-molecule morphinan antagonists, which induce G protein-dependent MOR activation. Co-application of BPRMU191 and morphinan antagonists resulted in MOR-dependent analgesia with diminished side effects, including gastrointestinal dysfunction, antinociceptive tolerance, and physical and psychological dependence. Combining BPRMU191 and morphinan antagonists could serve as a potential therapeutic strategy for severe pain with reduced adverse effects and provide an avenue for studying G protein-coupled receptor modulation.

8.
J Biol Chem ; 287(27): 22533-48, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22563078

RESUMEN

The nucleolar 58-kDa microspherule protein (MSP58) protein is a candidate oncogene implicated in modulating cellular proliferation and malignant transformation. In this study, we show that knocking down MSP58 expression caused aneuploidy and led to apoptosis, whereas ectopic expression of MSP58 regulated cell proliferation in a context-dependent manner. Specifically, ectopic expression of MSP58 in normal human IMR90 and Hs68 diploid fibroblasts, the H184B5F5/M10 mammary epithelial cell line, HT1080 fibrosarcoma cells, primary mouse embryonic fibroblasts, and immortalized NIH3T3 fibroblasts resulted in induction of premature senescence, an enlarged and flattened cellular morphology, and increased senescence-associated ß-galactosidase activity. MSP58-driven senescence was strictly dependent on the presence of functional p53 as revealed by the fact that normal cells with p53 knockdown by specific shRNA or cells with a mutated or functionally impaired p53 pathway were effective in bypassing MSP58-induced senescence. At least two senescence mechanisms are induced by MSP58. First, MSP58 activates the DNA damage response and p53/p21 signaling pathways. Second, MSP58, p53, and the SWI/SNF chromatin-remodeling subunit Brahma-related gene 1 (BRG1) form a ternary complex on the p21 promoter and collaborate to activate p21. Additionally, MSP58 protein levels increased in cells undergoing replicative senescence and stress-induced senescence. Notably, the results of analyzing expression levels of MSP58 between tumors and matched normal tissues showed significant changes (both up- and down-regulation) in its expression in various types of tumors. Our findings highlight new aspects of MSP58 in modulating cellular senescence and suggest that MSP58 has both oncogenic and tumor-suppressive properties.


Asunto(s)
Senescencia Celular/fisiología , ADN Helicasas/metabolismo , Proteínas Nucleares/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/fisiología , División Celular/fisiología , Línea Celular Transformada , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Daño del ADN/fisiología , ADN Helicasas/genética , Fibrosarcoma , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/fisiología , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Transducción de Señal/fisiología , Estrés Fisiológico/fisiología , Factores de Transcripción/genética
9.
Nucleic Acids Res ; 39(13): 5412-23, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21441538

RESUMEN

The exact mechanism underlying increases in Sp1 and the physiological consequences thereafter remains unknown. In rat primary cortical neurons, oxygen-glucose deprivation (OGD) causes an increase in H(2)O(2) as well as Sp1 in early ischaemia but apparently does not change mRNA level or Sp1 stability. We hereby identified a longer 5'-UTR in Sp1 mRNA that contains an internal ribosome entry site (IRES) that regulates rapid and efficient translation of existing mRNAs. By using polysomal fragmentation and bicistronic luciferase assays, we found that H(2)O(2) activates IRES-dependent translation. Thus, H(2)O(2) or tempol, a superoxide dismutase-mimetic, increases Sp1 levels in OGD-treated neurons. Further, early-expressed Sp1 binds to Sp1 promoter to cause a late rise in Sp1 in a feed-forward manner. Short hairpin RNA against Sp1 exacerbates OGD-induced apoptosis in primary neurons. While Sp1 levels increase in the cortex in a rat model of stroke, inhibition of Sp1 binding leads to enhanced apoptosis and cortical injury. These results demonstrate that neurons can use H(2)O(2) as a signalling molecule to quickly induce Sp1 translation through an IRES-dependent translation pathway that, in cooperation with a late rise in Sp1 via feed-forward transcriptional activation, protects neurons against ischaemic damage.


Asunto(s)
Regiones no Traducidas 5' , Isquemia Encefálica/metabolismo , Peróxido de Hidrógeno/farmacología , Biosíntesis de Proteínas , Factor de Transcripción Sp1/genética , Animales , Glucosa/fisiología , Humanos , Masculino , Neuronas/metabolismo , Oxígeno/fisiología , Ratas , Ribosomas/metabolismo , Factor de Transcripción Sp1/biosíntesis , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Activación Transcripcional
10.
Eur J Med Chem ; 258: 115608, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37437352

RESUMEN

The compelling demand of a consummate analgesic medication without addiction is rising due to the clinical mistreatment. Additionally, the series of severe untoward effects usually deterred the utilization while coping with serious pain. As a possible turning point, we revealed that compound 14 is a dual agonist of mu opioid receptor (MOR) and nociceptin-orphanin FQ opioid peptide (NOP) receptor in this study. More importantly, compound 14 achieves pain relieving at very small doses, meanwhile, reduces several unwanted side effects such as constipation, reward, tolerance and withdrawal effects. Here, we evaluated the antinociception and side effects of this novel compound from wild type and humanized mice to further develop a safer prescription analgesic drug.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Receptores Opioides mu , Ratones , Animales , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Receptor de Nociceptina , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Analgésicos Opioides/efectos adversos , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Analgésicos/efectos adversos , Nociceptina
11.
Eur J Med Chem ; 243: 114728, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36084534

RESUMEN

Currently, there is a significant unmet need for novel analgesics with fewer side effects. In this study, we carried out structural modification of a hit compound previously identified in an artificial-intelligence (AI) virtual screening and discovered the potent analgesic, benzo[b]thiophene-2-carboxamide analog (compound 25) with new structural scaffold. We investigated the signaling pathways of opioid receptors mediated by compound 25, and found this racemic compound activated mu-opioid receptor through the cyclic adenosine monophosphate (cAMP) and ß-arrestin-2-mediated pathways with strong potency and efficacy, and accompanying nociceptin-orphanin FQ opioid peptide and delta-opioid receptors through the cAMP pathway with weak potencies. Compound 25 elicited potent antinociception in thermal-stimulated pain (ED50 value of 127.1 ± 34.65 µg/kg) and inflammatory-induced allodynia models with less gastrointestinal transit inhibition and antinociceptive tolerance than morphine. Overall, this study revealed a novel analgesic with reduced risks of side effects.


Asunto(s)
Analgésicos Opioides , Tiofenos , Humanos , Tiofenos/farmacología , Tiofenos/uso terapéutico , Analgésicos Opioides/efectos adversos , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Péptidos Opioides , Morfina/farmacología , Analgésicos/farmacología , Analgésicos/uso terapéutico , Analgésicos/química , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico
12.
Neurobiol Learn Mem ; 93(3): 372-82, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19995614

RESUMEN

In many instances, increase in neuronal activity can induce biphasic secretion of a modulator. The initial release of the modulator triggers the induction of synaptic plasticity, whereas the second-phase release reinforces the efficacy of synaptic transmission and growth of dendrites and axons. In this study, we showed that fear conditioning not only induced the first but also a second peak of brain-derived neurotrophic factor (BDNF) expression. Fluorescent immunohistostaining confirmed that BDNF expression increased at 1 and 12 h after conditioning and returned to baseline at 30 h after conditioning. Mature BDNF expression increased in a similar manner. TrkB-IgG or K252a infusion before training impaired fear memory on days 1 and 7 after training. In contrast, TrkB-IgG or K252a infusion 9 h after fear conditioning did not affect memory retention on day 1 after training but impaired fear memory on day 7 after training. Fear conditioning significantly enhanced Zif268 expression in the amygdala at 12 h after training; this enhanced expression was completely inhibited by TrkB-IgG infusion 9 h after training. The level of growth-associated protein 43 (GAP-43), a marker of newly formed synapses, in the amygdala increased 7 days after fear conditioning. Moreover, conditioned rats had higher AMPA/NMDA ratio than unpaired rats. These results suggest that consolidated memory could be continuously modulated by previous molecular changes produced during memory acquisition.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Miedo , Expresión Génica/genética , Memoria/fisiología , Estimulación Acústica , Animales , Carbazoles/metabolismo , Técnica del Anticuerpo Fluorescente , Hipocampo/metabolismo , Alcaloides Indólicos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptor trkB/genética , Receptor trkB/metabolismo , Reflejo de Sobresalto , Transducción de Señal/fisiología , Factores de Tiempo
13.
Cancers (Basel) ; 12(4)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326583

RESUMEN

Patients with glioblastoma are at high risk of local recurrences after initial treatment with standard therapy, and recurrent tumor cells appear to be resistant to first-line drug temozolomide. Thus, finding an effective second-line agent for treating primary and recurrent glioblastomas is critical. Betulinic acid (BA), a natural product of plant origin, can cross the blood-brain barrier. Here, we investigated the antitumor effects of BA on typical glioblastoma cell lines and primary glioblastoma cells from patients, as well as corresponding temozolomide-resistant cells. Our findings verified that BA significantly reduced growth in all examined cells. Furthermore, gene-expression array analysis showed that the unfolded-protein response was significantly affected by BA. Moreover, BA treatment increased activation of the protein kinase RNA-like endoplasmic reticulum kinase (PERK)/C/EBP homologous protein (CHOP) apoptotic pathway, and reduced specificity protein 1 (Sp1) expression. However, Sp1 overexpression reversed the observed cell-growth inhibition and PERK/CHOP signaling activation induced by BA. Because temozolomide-resistant cells exhibited significantly increased Sp1 expression, we concluded that Sp1-mediated PERK/CHOP signaling inhibition protects glioblastoma against cancer therapies; hence, BA treatment targeting this pathway can be considered as an effective therapeutic strategy to overcome such chemoresistance and tumor relapse.

14.
Pain ; 161(6): 1177-1190, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32040076

RESUMEN

Morphine is a strong painkiller acting through mu-opioid receptor (MOR). Full-length 7-transmembrane (TM) variants of MOR share similar amino acid sequences of TM domains in rodents and humans; however, interspecies differences in N- and C-terminal amino acid sequences of MOR splice variants dramatically affect the downstream signaling. Thus, it is essential to develop a mouse model that expresses human MOR splice variants for opioid pharmacological studies. We generated 2 lines of fully humanized MOR mice (hMOR; mMOR mice), line #1 and #2. The novel murine model having human OPRM1 genes and human-specific variants was examined by reverse-transcription polymerase chain reaction and the MinION nanopore sequencing. The differences in the regional distribution of MOR between wild-type and humanized MOR mice brains were detected by RNAscope and radioligand binding assay. hMOR; mMOR mice were characterized in vivo using a tail-flick, charcoal meal, open field, tail suspension, naloxone precipitation tests, and rectal temperature measurement. The data indicated that wild-type and humanized MOR mice exhibited different pharmacology of morphine, including antinociception, tolerance, sedation, and withdrawal syndromes, suggesting the presence of species difference between mouse and human MORs. Therefore, hMOR; mMOR mice could serve as a novel mouse model for pharmacogenetic studies of opioids.


Asunto(s)
Hipotermia , Morfina , Receptores Opioides mu , Secuencia de Aminoácidos , Analgésicos Opioides/farmacología , Animales , Tolerancia a Medicamentos , Humanos , Ratones , Ratones Transgénicos , Morfina/farmacología , Receptores Opioides mu/genética
15.
Sci Rep ; 10(1): 16771, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033310

RESUMEN

Machine learning is a well-known approach for virtual screening. Recently, deep learning, a machine learning algorithm in artificial neural networks, has been applied to the advancement of precision medicine and drug discovery. In this study, we performed comparative studies between deep neural networks (DNN) and other ligand-based virtual screening (LBVS) methods to demonstrate that DNN and random forest (RF) were superior in hit prediction efficiency. By using DNN, several triple-negative breast cancer (TNBC) inhibitors were identified as potent hits from a screening of an in-house database of 165,000 compounds. In broadening the application of this method, we harnessed the predictive properties of trained model in the discovery of G protein-coupled receptor (GPCR) agonist, by which computational structure-based design of molecules could be greatly hindered by lack of structural information. Notably, a potent (~ 500 nM) mu-opioid receptor (MOR) agonist was identified as a hit from a small-size training set of 63 compounds. Our results show that DNN could be an efficient module in hit prediction and provide experimental evidence that machine learning could identify potent hits in silico from a limited training set.


Asunto(s)
Antineoplásicos/uso terapéutico , Aprendizaje Profundo , Receptores Acoplados a Proteínas G/agonistas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Algoritmos , Descubrimiento de Drogas/métodos , Humanos , Redes Neurales de la Computación
16.
Neuro Oncol ; 22(10): 1439-1451, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32328646

RESUMEN

BACKGROUND: Glioblastoma is associated with poor prognosis and high mortality. Although the use of first-line temozolomide can reduce tumor growth, therapy-induced stress drives stem cells out of quiescence, leading to chemoresistance and glioblastoma recurrence. The specificity protein 1 (Sp1) transcription factor is known to protect glioblastoma cells against temozolomide; however, how tumor cells hijack this factor to gain resistance to therapy is not known. METHODS: Sp1 acetylation in temozolomide-resistant cells and stemlike tumorspheres was analyzed by immunoprecipitation and immunoblotting experiments. Effects of the histone deacetylase (HDAC)/Sp1 axis on malignant growth were examined using cell proliferation-related assays and in vivo experiments. Furthermore, integrative analysis of gene expression with chromatin immunoprecipitation sequencing and the recurrent glioblastoma omics data were also used to further determine the target genes of the HDAC/Sp1 axis. RESULTS: We identified Sp1 as a novel substrate of HDAC6, and observed that the HDAC1/2/6/Sp1 pathway promotes self-renewal of malignancy by upregulating B cell-specific Mo-MLV integration site 1 (BMI1) and human telomerase reverse transcriptase (hTERT), as well as by regulating G2/M progression and DNA repair via alteration of the transcription of various genes. Importantly, HDAC1/2/6/Sp1 activation is associated with poor clinical outcome in both glioblastoma and low-grade gliomas. However, treatment with azaindolyl sulfonamide, a potent HDAC6 inhibitor with partial efficacy against HDAC1/2, induced G2/M arrest and senescence in both temozolomide-resistant cells and stemlike tumorspheres. CONCLUSION: Our study uncovers a previously unknown regulatory mechanism in which the HDAC6/Sp1 axis induces cell division and maintains the stem cell population to fuel tumor growth and therapeutic resistance.


Asunto(s)
Glioblastoma , Apoptosis , Línea Celular Tumoral , Resistencia a Antineoplásicos , Puntos de Control de la Fase G2 del Ciclo Celular , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Histona Desacetilasa 1/genética , Humanos , Factor de Transcripción Sp1/genética
17.
Neuropharmacology ; 166: 107678, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31278929

RESUMEN

There is unmet need to design an analgesic with fewer side effects for severe pain management. Although traditional opioids are the most effective painkillers, they are accompanied by severe adverse responses, such as respiratory depression, constipation symptoms, tolerance, withdrawal, and addiction. We indicated BPR1M97 as a dual mu opioid receptor (MOP)/nociceptin-orphanin FQ peptide (NOP) receptor full agonist and investigated the pharmacology of BPR1M97 in multiple animal models. In vitro studies on BPR1M97 were assessed using cyclic-adenosine monophosphate production, ß-arrestin, internalization, and membrane potential assays. In vivo studies were characterized using the tail-flick, tail-clip, lung functional, heart functional, acetone drop, von Frey hair, charcoal meal, glass bead, locomotor activity, conditioned place preference (CPP) and naloxone precipitation tests. BPR1M97 elicited full agonist properties for all cell-based assays tested in MOP-expressing cells. However, it acted as a G protein-biased agonist for NOP. BPR1M97 initiated faster antinociceptive effects at 10 min after subcutaneous injection and elicited better analgesia in cancer-induced pain than morphine. Unlike morphine, BPR1M97 caused less respiratory, cardiovascular, and gastrointestinal dysfunction. In addition, BPR1M97 decreased global activity and induced less withdrawal jumping precipitated by naloxone. Thus, BPR1M97 could serve as a novel small molecule dual receptor agonist for antinociception with fewer side effects than morphine. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Analgésicos/uso terapéutico , Morfina/uso terapéutico , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Células CHO , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/patología , Cricetulus , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Dimensión del Dolor/métodos , Resultado del Tratamiento , Receptor de Nociceptina
18.
J Neurosci ; 28(52): 14259-70, 2008 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-19109507

RESUMEN

Inflammation is involved in some neurodegenerative disorders. NMDA glutamate receptors play an important role in neuronal development. Here, we show that NR1 expression in the cerebral cortex and primary neurons of rats was upregulated after lipopolysaccharide (LPS) treatment. This increase in NR1 expression was considered to be strongly associated with hypoxia-inducible factor-1alpha (HIF-1alpha) activation because the treatment of primary neurons with either echinomycin or small interfering RNA (siRNA) targeting HIF-1alpha could block NR1 expression. HIF-1alpha could be induced by an increase in the translational efficiency of the cells. After this, it was transported into the nucleus where it bound to the NR1 promoter and regulated the induction of NR1 transcriptional activity by LPS. LPS injection into the prefrontal cortex caused neuronal death, and this condition was aggravated by intracerebroventricular injection of echinomycin. Furthermore, knockdown of HIF-1alpha and NR1 by the appropriate siRNAs reduced the neurite outgrowth and viability of the primary neurons. These results suggest that NR1 expression is regulated by HIF-1alpha and plays a protective role in neurons during LPS challenge.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/farmacología , Neuronas/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Equinomicina/farmacología , Lipopolisacáridos/farmacología , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosfopiruvato Hidratasa/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Factores de Tiempo
19.
Sci Rep ; 9(1): 2405, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787373

RESUMEN

Morphine is a unique opioid analgesic that activates the mu-opioid receptor (MOR) without efficiently promoting its endocytosis that may underlie side effects. Our objective was to discover a novel enhancer of ligand-induced MOR endocytosis and determine its effects on analgesia, tolerance and dependence. We used high-throughput screening to identify convallatoxin as an enhancer of ligand-induced MOR endocytosis with high potency and efficacy. Treatment of cells with convallatoxin enhanced morphine-induced MOR endocytosis through an adaptor protein 2 (AP2)/clathrin-dependent mechanism, attenuated morphine-induced phosphorylation of MOR, and diminished desensitization of membrane hyperpolarization. Furthermore, co-treatment with chronic convallatoxin reduced morphine tolerance in animal models of acute thermal pain and chronic inflammatory pain. Acute convallatoxin administration reversed morphine tolerance and dependence in morphine-tolerant mice. These findings suggest convallatoxin are potentially therapeutic for morphine side effects and open a new avenue to study MOR trafficking.


Asunto(s)
Analgésicos/farmacología , Morfina/farmacología , Receptores Opioides mu/genética , Estrofantinas/farmacología , Analgesia/métodos , Analgésicos/química , Animales , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Humanos , Ligandos , Ratones , Receptores Opioides mu/efectos de los fármacos
20.
Eur J Med Chem ; 167: 312-323, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30776693

RESUMEN

Morphine is widely used for the treatment of severe pain. This analgesic effect is mediated principally by the activation of µ-opioid receptors (MOR). However, prolonged activation of MOR also results in tolerance, dependence, addiction, constipation, nausea, sedation, and respiratory depression. To address this problem, we sought alternative ways to activate MOR - either by use of novel ligands, or via a novel activation mechanism. To this end, a series of compounds were screened using a sensitive CHO-K1/MOR/Gα15 cell-based FLIPR® calcium high-throughput screening (HTS) assay, and the bithiazole compound 5a was identified as being able activate MOR in combination with naloxone. Structural modifications of 5a resulted in the discovery of lead compound 5j, which could effectively activate MOR in combination with the MOR antagonist naloxone or naltrexone. In vivo, naloxone in combination with 100 mg/kg of compound 5j elicited antinociception in a mouse tail-flick model with an ED50 of 17.5 ±â€¯4 mg/kg. These results strongly suggest that the mechanism by which the 5j/naloxone combination activates MOR is worthy of further study, as its discovery has the potential to yield an entirely novel class of analgesics.


Asunto(s)
Analgésicos/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/uso terapéutico , Receptores Opioides mu/agonistas , Tiazoles/farmacología , Aminas , Animales , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Muridae , Antagonistas de Narcóticos/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA