Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Molecules ; 26(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34833919

RESUMEN

BACKGROUND: Recent studies suggest that lipids, including free fatty acids (FFAs), are necessary for proper µ opioid receptor (MOR) binding and that activation of opioid receptors (ORs) improves intestinal inflammation. The objective of the study was to investigate a possible interaction between the ORs and FFA receptors (FFARs) ligands in the colitis. METHODS: The potential synergistic effect of ORs and FFARs ligands was evaluated using mouse model of acute colitis induced by dextran sulfate sodium (DSS, 4%). Compounds were injected intraperitoneally (i.p.) once or twice daily at the doses of 0.01 or 0.02 mg/kg body weight (BW) (DAMGO-an MOR agonist), 0.3 mg/kg BW (DPDPE-a δ OR (DOR) agonist) and 1 mg/kg BW (naloxone-a non-selective OR antagonist, GLPG 0974-a FFAR2 antagonist, GSK 137647-a FFAR4 agonist and AH 7614-a FFAR4 antagonist) for 4 days. RESULTS: Myeloperoxidase (MPO) activity was significantly decreased after DAMGO (0.02 mg/kg BW) and GSK 137647 (1 mg/kg BW) administration and co-administration as compared to DSS group. CONCLUSIONS: Treatment with ligands of ORs and FFARs may affect the immune cells in the inflammation; however, no significant influence on the severity of colitis and no synergistic effect were observed.


Asunto(s)
Colitis/tratamiento farmacológico , Colitis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/metabolismo , Compuestos de Anilina/administración & dosificación , Animales , Butiratos/administración & dosificación , Colitis/inmunología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina D-Penicilamina (2,5)/administración & dosificación , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos BALB C , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Peroxidasa/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Opioides/agonistas , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Sulfonamidas/administración & dosificación , Tiofenos/administración & dosificación , Xantenos/administración & dosificación
2.
Mol Pharm ; 18(3): 1470-1479, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33586444

RESUMEN

To enhance the water solubility, oral bioavailability, and tumor targeting of gambogenic acid (GNA), polydopamine nanoparticles (PDA NPs) were prepared to encapsulate and stabilize GNA surface modified by folic acid (FA) and then coated with sodium alginate (GNA@PDA-FA SA NPs) to achieve an antitumor effect by oral administration. GNA@PDA-FA SA NPs exhibited in vitro pH-sensitive release behavior. In vitro cell studies manifested that GNA@PDA-FA NPs had higher cytotoxicity to 4T1 cells compared with raw GNA (IC50 = 2.58 µM vs 7.57 µM). After being modified with FA, GNA@PDA-FA NPs were taken up easily by 4T1 cells. In vivo studies demonstrated that the area under the curve (AUC0→∞) of the plasma drug concentration-time of GNA@PDA-FA SA NPs was 2.97-fold higher than that of raw GNA, along with improving drug distribution in the liver, lung, and kidney tissues. In vivo anti-tumor experiments, GNA@PDA-FA SA NPs significantly inhibited the growth of breast tumors in the 4T1 xenograft breast cancer model via oral administration without obvious toxicity on major organs. Our studies indicated that the GNA@PDA-FA SA NPs modified with FA and coated with SA were a promising drug delivery system for targeting tumor therapy via oral administration.


Asunto(s)
Indoles/química , Nanopartículas/química , Polímeros/química , Xantenos/administración & dosificación , Administración Oral , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Portadores de Fármacos/química , Ácido Fólico/administración & dosificación , Humanos , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Ratas , Ratas Sprague-Dawley , Solubilidad/efectos de los fármacos
3.
Kaohsiung J Med Sci ; 36(5): 344-353, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32293112

RESUMEN

The aim of present study was to develop folic acid (FA)-modified nonionic surfactant vesicles (NISVs, niosomes) as carrier systems for targeted delivery of gambogenic acid (GNA). The FA-GNA-NISVs exhibited a mean particle size of 180.77 ± 2.41 nm with a narrow poly dispersion index of 0.147 ± 0.08 determined by dynamic light scattering. Transmission electron microscopy also revealed that the FA-GNA-NISVs were spherical with double-layer structure. Entrapment efficiency (EE%) and zeta potential of the optimal FA-GNA-NISVs were 87.84 ± 1.06% and -37.33 ± 0.33 mV, respectively. Differential scanning calorimetry demonstrated that the GNA was in a molecular or amorphous state inside the FA-NISVs in vitro release profiles suggested that FA-GNA-NISVs could release GNA at a sustained manner, and less than 60% of GNA was released from the FA-NISVs within 12 hours of dialysis. in vivo pharmacokinetic results illustrated that FA-GNA-NISVs had considerably higher Cmax , area under curve (AUC0 - t ) and accumulation in lung. The cell proliferation study shown that the FA-GNA-NISVs significantly enhanced the in vitro cytotoxicity against A549 cells. Flow cytometry and fluorescence microscopy further demonstrated that the FA-GNA-NISVs increased apoptosis compared with nonmodified GNA-NISVs and free GNA. Moreover, FA-GNA-NISVs induced A549 cell apoptosis in a dose-dependent manner. In addition, cellular uptake assays showed a higher uptake of FA-GNA-NISVs than GNA-NISVs as well as free GNA. Taken together, it could be concluded that FA-GNA-NISVs were proposed as a novel targeting carriers for efficient delivering of GNA to cancers cells.


Asunto(s)
Ácido Fólico/química , Tensoactivos/química , Xantenos/farmacología , Células A549 , Animales , Apoptosis/efectos de los fármacos , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Humanos , Liposomas , Tamaño de la Partícula , Ratas Sprague-Dawley , Electricidad Estática , Distribución Tisular/efectos de los fármacos , Xantenos/administración & dosificación , Xantenos/química , Xantenos/farmacocinética
4.
J Microencapsul ; 36(6): 566-575, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31411510

RESUMEN

Objective: To improve the water solubility and enhance the oral bioavailability of gambogenic acid (GNA). Methods: GNA-phospholipid complex (GNA-PLC) micelles were successfully prepared by anti-solvent method. Results: The encapsulation efficiency of GNA-PLC micelles can reach 99.33 % (w/w). The average particle size of the GNA-PLC micelles was 291.23 nm which was approximate agreed with the transmission electron microscopy (TEM). In vitro release profile showed the GNA-PLC and GNA-PLC micelles have significant sustained-release of GNA compared with crude GNA. Pharmacokinetic parameters indicated that the area under concentration-time curve (AUC0→t) of GNA in cases of GNA-PLC and GNA-PLC micelles are 2.04- and 3.92-fold higher than crude GNA, respectively. Conclusions: The better water solubility and higher bioavailability of GNA in GNA-PLC micelles with significant sustained-release of GNA endow the nanoparticle with great potential in GNA delivery system.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Preparaciones de Acción Retardada/química , Micelas , Fosfolípidos/química , Xantenos/administración & dosificación , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Disponibilidad Biológica , Liberación de Fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacocinética , Femenino , Garcinia/química , Células Hep G2 , Humanos , Masculino , Ratas Sprague-Dawley , Solubilidad , Xantenos/química , Xantenos/farmacocinética
5.
J Biol Chem ; 294(27): 10649-10662, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31142616

RESUMEN

Proteases sustain hyperexcitability and pain by cleaving protease-activated receptor-2 (PAR2) on nociceptors through distinct mechanisms. Whereas trypsin induces PAR2 coupling to Gαq, Gαs, and ß-arrestins, cathepsin-S (CS) and neutrophil elastase (NE) cleave PAR2 at distinct sites and activate it by biased mechanisms that induce coupling to Gαs, but not to Gαq or ß-arrestins. Because proteases activate PAR2 by irreversible cleavage, and activated PAR2 is degraded in lysosomes, sustained extracellular protease-mediated signaling requires mobilization of intact PAR2 from the Golgi apparatus or de novo synthesis of new receptors by incompletely understood mechanisms. We found here that trypsin, CS, and NE stimulate PAR2-dependent activation of protein kinase D (PKD) in the Golgi of HEK293 cells, in which PKD regulates protein trafficking. The proteases stimulated translocation of the PKD activator Gßγ to the Golgi, coinciding with PAR2 mobilization from the Golgi. Proteases also induced translocation of a photoconverted PAR2-Kaede fusion protein from the Golgi to the plasma membrane of KNRK cells. After incubation of HEK293 cells and dorsal root ganglia neurons with CS, NE, or trypsin, PAR2 responsiveness initially declined, consistent with PAR2 cleavage and desensitization, and then gradually recovered. Inhibitors of PKD, Gßγ, and protein translation inhibited recovery of PAR2 responsiveness. PKD and Gßγ inhibitors also attenuated protease-evoked mechanical allodynia in mice. We conclude that proteases that activate PAR2 by canonical and biased mechanisms stimulate PKD in the Golgi; PAR2 mobilization and de novo synthesis repopulate the cell surface with intact receptors and sustain nociceptive signaling by extracellular proteases.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteína Quinasa C/metabolismo , Receptor PAR-2/metabolismo , Animales , Catepsinas/metabolismo , Membrana Celular/metabolismo , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/patología , Hiperalgesia/prevención & control , Elastasa de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa C/antagonistas & inhibidores , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Receptor PAR-2/agonistas , Transducción de Señal/efectos de los fármacos , Xantenos/administración & dosificación , Xantenos/farmacología
6.
Int J Pharm ; 562: 218-227, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30902707

RESUMEN

PURPOSE: To develop a three-dimensional visualization method for evaluating the distribution of pulmonary drug delivery systems and compare four tissue-clearing techniques (ClearT2, CUBIC, ScaleS, and SeeDB2) using intrapulmonary liposomes as drug carriers. METHODS: Rhodamine B-labeled liposomes were administered intrapulmonarily to mice using a MicroSprayer, and then fluorescent-labeled tomato lectin was administered intravenously to visualize the general lung structure. Tissue-clearing treatment of the mouse lungs was performed using the standard protocols of the ClearT2, CUBIC, ScaleS, and SeeDB2 techniques. Lung clearing was clarified using laser-scanning confocal microscopy, and three-dimensional images were reconstructed. RESULTS: Fluorescent-labeled tomato lectin was preserved using ClearT2 and SeeDB2 but not using CUBIC and ScaleS. In addition, the liposomes were stable in ClearT2 reagent, but they were mostly degraded in other reagents by surface-active agents. ClearT2 treatment enabled the three-dimensional visualization of intrapulmonary rhodamine B-labeled liposomes at the alveolar scale. CONCLUSIONS: These results suggest that the ClearT2 tissue-clearing technique was appropriate for the three-dimensional visualization of intrapulmonary liposomes at the alveolar scale. This study provides important information for selecting and optimizing suitable optical tissue-clearing techniques in lungs for evaluating the distribution of pulmonary drug delivery systems.


Asunto(s)
Imagenología Tridimensional/métodos , Liposomas/administración & dosificación , Alveolos Pulmonares/metabolismo , Animales , Dextranos/administración & dosificación , Fluoresceína-5-Isotiocianato/administración & dosificación , Fluoresceína-5-Isotiocianato/análogos & derivados , Colorantes Fluorescentes/administración & dosificación , Masculino , Ratones Endogámicos ICR , Lectinas de Plantas/administración & dosificación , Rodaminas/administración & dosificación , Distribución Tisular , Xantenos/administración & dosificación
7.
Chin J Nat Med ; 16(9): 641-643, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30269840

RESUMEN

Garcinia, a kind of dry resin secreted by Garcinia hanburyi Hook. F. G., is a traditional Chinese medicine with various biological functions such as detoxification, anti-inflammatory, and anthelmintic activities. Recent studies suggest that garcinia has potential anticancer activity. Increasing evidences indicate that the main active monomer gambogic acid isolated from garcinia can inhibit the growth of various cancer cells. Neogambogic acid is an isolated compound with a similar chemical structure as gambogic acid. Preliminary studies show that the neogambogic acid can selectively inhibit the growth of various cancer cells, and has a broader antitumor activity and lower toxicity than gambogic acid. In this review, we summarize the advances made in the investigation of the anti-tumor effect of neogambogic acid in recent years.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Garcinia/química , Neoplasias/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Xantenos/administración & dosificación , Animales , Antineoplásicos Fitogénicos/química , Humanos , Extractos Vegetales/química , Xantenos/química
8.
Colloids Surf B Biointerfaces ; 172: 26-36, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30125771

RESUMEN

Gambogenic acid (GNA), which possesses diverse anti-tumor activities both in vitro and in vivo, is regarded as a potential anticancer compound. However, the excessive irritation to the blood vessel, short elimination half-life and poor aqueous solubility restricted its clinical application. In this study, Gambogenic acid-loaded PEGylated liposomes (GNA-PEG-LPs) were developed to reduce toxicity, prolong the half-life and enhance anticancer efficacy both in vitro and in vivo. The average particle size of GNA-PEG-LPs was 90.13 ± 0.16 nm and their polydispersity index (PDI) was 0.092 ± 0.003. The encapsulation efficiency, drug loading and zeta potential of GNA-PEG-LPs were 88.13 ± 1.31%, 3.72 ± 0.04%, -22.10 ± 0.20 mV, respectively. Compared to free GNA, GNA-PEG-LPs showed enhanced cytotoxicity and apoptosis induction effect against A549, SGC-7901 and HepG2 cells. Mechanistically, western blot analysis revealed that up-regulation of Bax, down-regulation of Bcl-2 and activation of caspase-3 contributed to apoptosis. In addition, the blood vessel irritation test showed that the vascular irritation of GNA could be reduced by liposomal encapsulation. The hemolysis assay revealed that good hemocompatibility of the liposomes. Furthermore, pharmacokinetic study showed that the t1/2 and the AUC of GNA-PEG-LPs were higher than GNA solution approximately 2.84-fold and 2.97-fold, respectively. In vivo antitumor efficacy demonstrated that GNA-PEG-LPs significantly inhibited the tumor growth in LLC tumor-bearing C57BL/6 mouse model. Results of Immunohistochemistry indicated that GNA-PEG-LPs significantly suppressed the expression of Bcl-2 and increased the expression of Bax and caspase-3 compared with free GNA. Collectively, PEGylated liposomes could be a potential nanocarrier to prolong half-life, reduce toxicity and enhance anticancer efficacy both in vitro and in vivo.


Asunto(s)
Sistemas de Liberación de Medicamentos , Polietilenglicoles/química , Xantenos/administración & dosificación , Xantenos/farmacología , Administración Intravenosa , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular , Línea Celular Tumoral , Liposomas/ultraestructura , Ratones Endogámicos C57BL , Conejos , Ratas Sprague-Dawley , Coloración y Etiquetado , Xantenos/química , Xantenos/farmacocinética
9.
J Control Release ; 286: 145-153, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30009893

RESUMEN

Focused ultrasound combined with microbubble-mediated intranasal delivery (FUSIN) is a new brain drug delivery technique. FUSIN utilizes the nasal route for direct nose-to-brain drug administration, thereby bypassing the blood-brain barrier (BBB) and minimizing systemic exposure. It also uses FUS-induced microbubble cavitation to enhance transport of intranasally (IN) administered agents to the FUS-targeted brain location. Previous studies have provided proof-of-concept data showing the feasibility of FUSIN to deliver dextran and the brain-derived neurotrophic factor to the caudate putamen of mouse brains. The objective of this study was to evaluate the biodistribution of IN administered gold nanoclusters (AuNCs) and assess the feasibility and short-term safety of FUSIN for the delivery of AuNCs to the brainstem. Three experiments were performed. First, the whole-body biodistribution of IN administered 64Cu-alloyed AuNCs (64Cu-AuNCs) was assessed using in vivo positron emission tomography/computed tomography (PET/CT) and verified with ex vivo gamma counting. Control mice were intravenously (IV) injected with the 64Cu-AuNCs. Second, 64Cu-AuNCs and Texas red-labeled AuNCs (TR-AuNCs) were used separately to evaluate FUSIN delivery outcome in the brain. 64Cu-AuNCs or TR-AuNCs were administered to mice through the nasal route, followed by FUS sonication at the brainstem in the presence of systemically injected microbubbles. The spatial distribution of 64Cu-AuNCs and TR-AuNCs were examined by autoradiography and fluorescence microscopy of ex vivo brain slices, respectively. Third, histological analysis was performed to evaluate any potential histological damage to the nose and brain after FUSIN treatment. The experimental results revealed that IN administration induced significantly lower 64Cu-AuNCs accumulation in the blood, lungs, liver, spleen, kidney, and heart compared with IV injection. FUSIN enhanced the delivery of 64Cu-AuNCs and TR-AuNCs at the FUS-targeted brain region compared with IN delivery alone. No histological-level tissue damage was detected in the nose, trigeminal nerve, and brain. These results suggest that FUSIN is a promising technique for noninvasive, spatially targeted, and safe delivery of nanoparticles to the brain with minimal systemic exposure.


Asunto(s)
Encéfalo/metabolismo , Medios de Contraste/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Oro/administración & dosificación , Microburbujas , Administración Intranasal , Animales , Barrera Hematoencefálica/metabolismo , Medios de Contraste/farmacocinética , Femenino , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/farmacocinética , Oro/farmacocinética , Ratones Endogámicos C57BL , Tomografía Computarizada por Tomografía de Emisión de Positrones , Sonicación/métodos , Distribución Tisular , Xantenos/administración & dosificación , Xantenos/farmacocinética
10.
Biomacromolecules ; 19(6): 2071-2081, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29630839

RESUMEN

We report on the preparation of photo- and reduction-responsive diblock copolymers through reversible addition-fragmentation chain transfer (RAFT) polymerization of a coumarin-based disulfide-containing monomer (i.e., CSSMA) using a poly(ethylene oxide) (PEO)-based macroRAFT agent. The resulting amphiphilic PEO- b-PCSSMA copolymers self-assembled into polymersomes with hydrophilic PEO shielding coronas and hydrophobic bilayer membranes. Upon irradiating the polymersomes with visible light (e.g., 430 nm), the coumarin moieties within the bilayer membranes were cleaved with the generation of primary amine groups, which spontaneously underwent inter/intrachain amidation reactions with the ester moieties, thereby tracelessly cross-linking and permeating the bilayer membranes. Notably, this process only gave rise to the release of small molecule payloads (e.g., doxorubicin hydrochloride, DOX) while large molecule encapsulants (e.g., Texas red-labeled dextran, TR-dextran) were retained within the cross-linked polymersomes due to the preservation of the integrity of the vesicular nanostructures. However, cross-linked polymersomes undergo further structural disintegration upon incubation with glutathione (GSH) due to the scission of disulfide linkages, resulting in the release of macromolecular payloads. Thus, dual-stimuli responsive polymersomes with tracelessly cross-linkable characteristics enable sequential release of payloads with spatiotemporal precision, which could be of promising applications in synergistic loading and programmed release of therapeutics.


Asunto(s)
Portadores de Fármacos/química , Sustancias Macromoleculares/química , Polímeros/química , Cumarinas/química , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Liberación de Fármacos , Glutatión/química , Luz , Sustancias Macromoleculares/síntesis química , Procesos Fotoquímicos , Polietilenglicoles/química , Polimerizacion , Polímeros/síntesis química , Rayos Ultravioleta , Xantenos/administración & dosificación , Xantenos/química , Xantenos/farmacocinética
11.
Front Immunol ; 9: 494, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593735

RESUMEN

Current investigations underline the important roles of C-C motif ligands in the development of neuropathic pain; however, their participation in diabetic neuropathy is still undefined. Therefore, the goal of our study was to evaluate the participation of macrophage inflammatory protein-1 (MIP-1) family members (CCL3, CCL4, CCL9) in a streptozotocin (STZ)-induced mouse model of diabetic neuropathic pain. Single intrathecal administration of each MIP-1 member (10, 100, or 500 ng/5 µl) in naïve mice evoked hypersensitivity to mechanical (von Frey test) and thermal (cold plate test) stimuli. Concomitantly, protein analysis has shown that, 7 days following STZ injection, the levels of CCL3 and CCL9 (but not CCL4) are increased in the lumbar spinal cord. Performed additionally, immunofluorescence staining undoubtedly revealed that CCL3, CCL9, and their receptors (CCR1 and CCR5) are expressed predominantly by neurons. In vitro studies provided evidence that the observed expression of CCL3 and CCL9 may be partially of glial origin; however, this observation was only partially possible to confirm by immunohistochemical study. Single intrathecal administration of CCL3 or CCL9 neutralizing antibody (2 and 4 µg/5 µl) delayed neuropathic pain symptoms as measured at day 7 following STZ administration. Single intrathecal injection of a CCR1 antagonist (J113863; 15 and 20 µg/5 µl) also attenuated pain-related behavior as evaluated at day 7 after STZ. Both neutralizing antibodies, as well as the CCR1 antagonist, enhanced the effectiveness of morphine in STZ-induced diabetic neuropathy. These findings highlight the important roles of CCL3 and CCL9 in the pathology of diabetic neuropathic pain and suggest that they play pivotal roles in opioid analgesia.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Proteínas Inflamatorias de Macrófagos/metabolismo , Macrófagos/inmunología , Derivados de la Morfina/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuronas/fisiología , Animales , Células Cultivadas , Quimiocina CCL3/metabolismo , Quimiocina CCL4/metabolismo , Quimiocinas CC/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Receptores CCR1/antagonistas & inhibidores , Xantenos/administración & dosificación
12.
BMC Res Notes ; 10(1): 541, 2017 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084601

RESUMEN

OBJECTIVE: We previously reported that the olfactory receptor OR51E2, overexpressed in LNCaP prostate cancer cells, promotes cell invasiveness upon stimulation of its agonist ß-ionone, and this phenomenon increases metastatic spread. Furthermore, we showed that the induced cell invasiveness involves a PI3 kinase dependent signalling pathway. We report here the results of a new investigation to address whether gallein, a small inhibitor of G protein ßγ subunit interaction with PI3 kinase, can inhibit ß-ionone effects both in vitro and in vivo. RESULTS: We demonstrate that gallein can inhibit the ß-ionone-induced cell invasiveness in vitro, as well as the spread of metastases in vivo. LNCaP cell invasiveness, assessed using spheroid cultures in collagen gels in vitro, was increased by ß-ionone and the effect was reversed by co-administration of gallein. LNCaP tumour cells, subcutaneously inoculated to immunodeficient mice, generated more metastases in vivo when ß-ionone was applied through the skin. Furthermore, the intraperitoneal injection of gallein inhibited this increased metastasis spread. Our results thus support the role of OR51E2 in the ß-ionone observed effects, and suggest that gallein could be a potential new agent in personalized medicine of the tumours expressing OR51E2.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Norisoprenoides/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Odorantes/metabolismo , Xantenos/farmacología , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/tratamiento farmacológico , Transducción de Señal , Células Tumorales Cultivadas , Xantenos/administración & dosificación
13.
Neuropharmacology ; 111: 169-179, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27569995

RESUMEN

Clinical studies have shown that the muscarinic receptor antagonist scopolamine induces a potent and rapid antidepressant effect relative to conventional antidepressants. However, potential undesirable effects, including memory impairment, partially limit the use of scopolamine in psychiatry. In the present study, we propose to overcome these limitations and enhance the therapeutic effects of scopolamine via administration in combination with the group II metabotropic glutamate (mGlu) receptor antagonist, LY341495. Joint administration of sub-effective doses of scopolamine (0.03 or 0.1 mg/kg, i.p.) with a sub-effective dose of LY341495 (0.1 mg/kg, i.p.) induced a profound antidepressant effect in the tail suspension test (TST) and in the forced swim test (FST) in mice. This drug combination did not impair memory, as measured using the Morris water maze (MWM), and did not influence the locomotor activity of mice. Furthermore, we found that an AMPA receptor antagonist, NBQX (10 mg/kg), completely reversed the antidepressant-like activity of a mixture of scopolamine and LY341495 in the TST. However, this effect was not influenced by para-chlorophenylalanine (PCPA) pre-treatment, indicating a lack of involvement of serotonergic system activation in the antidepressant-like effects of jointly given scopolamine and LY341495. Therefore, the combined administration of low doses of the antimuscarinic drug scopolamine and the group II mGlu receptor antagonist LY341495 might be a new, effective and safe strategy in the therapy of depression.


Asunto(s)
Aminoácidos/administración & dosificación , Antidepresivos/administración & dosificación , Trastorno Depresivo/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Escopolamina/administración & dosificación , Xantenos/administración & dosificación , Animales , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Quinoxalinas/administración & dosificación , Receptores AMPA/antagonistas & inhibidores , Serotonina/metabolismo
14.
Biofactors ; 42(5): 533-544, 2016 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-27130074

RESUMEN

Mangiferin (MGF), a glucoside of xanthone existing in phytomedicines and food, is increasingly attracting attention on diabetes treatment, while the underlying mechanism leading to its low oral bioavailability is unclear. Norathyriol (NTR), an active metabolite with hypoglycemic activity and its exposure after MGF dosing remains unclear. Hence, a rapid and sensitive LC-MS/MS method was established and validated to determine MGF and NTR and applied in the PK study in rats. Correspondingly, the in vitro experiments on temperature-dependent uptake, and MGF metabolism in hepatocyte and enterobacteria samples were performed. Results revealed that hepatic first-pass effect slightly contributed to the poor bioavailability of MGF, based on the MGF exposure in portal vein plasma was nearly similar to that in systemic plasma, and the MGF accumulation in the liver was limited, so was that of NTR. Correspondingly, the in vitro study revealed the MGF uptake was mainly dependent on poor passive transport, possibly leading to its limited hepatic metabolism and accumulation. Moreover, the NTR exposure remained considerably low (Cmax < 3 ng/mL, AUCNTR /AUCMGF < 3%) in plasma after single MGF dosing, corresponding to its tiny proportion (0.1%) of MGF in MGF-incubated enterobacteria samples. However, given the low generation and elimination rates of NTR, NTR might accumulate in plasma and exert effects after repeated MGF dosing, although requires further study. This work is the first systemic study on PK profiles of MGF and NTR in vitro and in vivo, which is important for the interpretation on the poor bioavailability and pharmacodynamics of MGF. © 2016 BioFactors, 42(5):533-544, 2016.


Asunto(s)
Hipoglucemiantes/farmacocinética , Hígado/metabolismo , Xantenos/farmacocinética , Xantonas/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Células Cultivadas , Evaluación Preclínica de Medicamentos , Enterobacteriaceae/metabolismo , Microbioma Gastrointestinal , Hepatocitos/metabolismo , Hipoglucemiantes/administración & dosificación , Masculino , Cultivo Primario de Células , Ratas Wistar , Xantenos/administración & dosificación , Xantonas/administración & dosificación
15.
Biofactors ; 42(5): 545-555, 2016 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-27151461

RESUMEN

The poor bioavailability of mangiferin (MGF) is a major obstacle on its further development. Aimed to illustrate the underlying mechanism and improve its poor exposure, the compared PK profiles of MGF and norathyriol (NTR) after different MGF preparation were performed: pure MGF, the Rhizoma Anemarrhenae (Zhi-mu) decoction, MGF, and timosaponin B2 (TB-2) combination. Furthermore, the potential contributing factors, including uridine diphosphoglucuronosyltransferase (UGT), cytochrome P450 (CYP450), P-gp, and enterobacterial were investigated by comparing the PK profiles with and without the corresponding inhibitors or in different rat models. After taking MGF, CYP450 and UGT inhibition could decrease MGF and NTR exposure; P-gp inhibition slightly enhanced (48%) MGF exposure, whereas more apparent for the improved NTR exposure (302%); enterobacterial inhibition almost completely stopped the NTR production, but no such effect was observed for MGF. Compared with the limited improvement by the abovementioned inhibition, the MGF and NTR exposure could significantly increase by 11.5- and 5.9-fold in the Zhi-mu decoction compared with the MGF treatment, probably contributed to TB-2 as an absorption enhancer because the MGF and TB-2 combination produced a similar level of improvement on the PK paremeters of MGF and NTR to the herb treatment. Likewise, most of the effects by UGT, CYP450, P-gp, and enterobacteria followed a similar variation tendency between them. Therefore, the poor bioavailability of MGF possibly mainly attributed to its poor membrane permeability, but not transporters or metabolic enzymes, and the compatibility of MGF and TB-2 could probably expand the prospective application of MGF by improving its bioavailability. © 2016 BioFactors, 42(5):545-555, 2016.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Hipoglucemiantes/farmacocinética , Saponinas/farmacología , Esteroides/farmacología , Xantenos/farmacocinética , Xantonas/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Administración Oral , Anemarrhena/química , Animales , Disponibilidad Biológica , Sistema Enzimático del Citocromo P-450/metabolismo , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Enterobacteriaceae/metabolismo , Microbioma Gastrointestinal , Glucuronosiltransferasa/metabolismo , Hipoglucemiantes/administración & dosificación , Inactivación Metabólica , Masculino , Ratas Wistar , Rizoma/química , Xantenos/administración & dosificación , Xantonas/administración & dosificación
16.
Pharm Biol ; 54(9): 1680-6, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26916555

RESUMEN

Context Mangiferin has been reported to possess a potential hypouricaemic effect. However, the pharmacokinetic studies in rats showed that its oral bioavailability was only 1.2%, suggesting that mangiferin metabolites might exert the action. Objective The hypouricaemic effect and the xanthine oxidase inhibition of mangiferin and norathyriol, a mangiferin metabolite, were investigated. Inhibition of norathyriol analogues (compounds 3-9) toward xanthine oxidase was also evaluated. Materials and methods For a dose-dependent study, mangiferin (1.5-6.0 mg/kg) and norathyriol (0.92-3.7 mg/kg) were administered intragastrically to mice twice daily for five times. For a time-course study, mice received mangiferin and norathyriol both at a single dose of 7.1 µmol/kg. In vitro, inhibition of test compounds (2.4-2.4 mM) against xanthine oxidase activity was evaluated by the spectrophotometrical method. The inhibition type was identified from Lineweaver-Burk plots. Results Norathyriol (0.92, 1.85 and 3.7 mg/kg) dose dependently decreased the serum urate levels by 27.0, 33.6 and 37.4%, respectively. The action was more potent than that of mangiferin at the low dose, but was equivalent at the higher doses. Additionally, the hypouricaemic action of them exhibited a time dependence. In vitro, norathyriol markedly inhibited the xanthine oxidase activities, with the IC50 value of 44.6 µM, but mangiferin did not. The kinetic studies showed that norathyriol was an uncompetitive inhibitor by Lineweaver-Burk plots. The structure-activity relationships exhibited that three hydroxyl groups in norathyriol at the C-1, C-3 and C-6 positions were essential for maintaining xanthine oxidase inhibition. Discussion and conclusion Norathyriol was responsible for the hypouricaemic effect of mangiferin via inhibiting xanthine oxidase activity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Supresores de la Gota/farmacología , Hiperuricemia/tratamiento farmacológico , Ácido Úrico/sangre , Xantenos/farmacología , Xantina Oxidasa/antagonistas & inhibidores , Xantonas/farmacología , Administración Oral , Animales , Biomarcadores/sangre , Biotransformación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Supresores de la Gota/administración & dosificación , Supresores de la Gota/metabolismo , Hiperuricemia/sangre , Hiperuricemia/inducido químicamente , Hiperuricemia/enzimología , Cinética , Ratones , Estructura Molecular , Ácido Oxónico , Relación Estructura-Actividad , Xantenos/administración & dosificación , Xantenos/metabolismo , Xantina Oxidasa/metabolismo , Xantonas/administración & dosificación , Xantonas/metabolismo
17.
Sci Rep ; 6: 18868, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26743682

RESUMEN

Hydrogen sulphide (H2S), the third endogenous gaseous signalling molecule, has attracted attention in biochemical research. The selective detection of H2S in living systems is essential for studying its functions. Fluorescence detection methods have become useful tools to explore the physiological roles of H2S because of their real-time and non-destructive characteristics. Herein we report a near-infrared fluorescent probe, NIR-HS, capable of tracking H2S in living organisms. With high sensitivity, good selectivity and low cytotoxicity, NIR-HS was able to recognize both the exogenous and endogenous H2S in living cells. More importantly, it realized the visualization of endogenous H2S generated in cells overexpressing cystathionine ß-synthase (CBS), one of the enzymes responsible for producing endogenous H2S. The probe was also successfully applied to detect both the exogenous and endogenous H2S in living mice. The superior sensing properties of the probe render it a valuable research tool in the H2S-related medical research.


Asunto(s)
Dinitrobencenos/química , Colorantes Fluorescentes/química , Sulfuro de Hidrógeno/análisis , Imagen Molecular/métodos , Sondas Moleculares/química , Imagen Óptica/métodos , Xantenos/química , Animales , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Dinitrobencenos/administración & dosificación , Dinitrobencenos/síntesis química , Diseño de Fármacos , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/síntesis química , Expresión Génica , Humanos , Sulfuro de Hidrógeno/metabolismo , Inyecciones Intraperitoneales , Células MCF-7 , Masculino , Ratones , Sondas Moleculares/administración & dosificación , Sondas Moleculares/síntesis química , Sensibilidad y Especificidad , Xantenos/administración & dosificación , Xantenos/síntesis química
18.
Brain Struct Funct ; 221(1): 21-37, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25233810

RESUMEN

This study investigates the mechanism of action of spadin, a putative fast-acting peptidic antidepressant (AD) and a functional blocker of the K(+) TREK-1 channel, in relation with the medial prefrontal cortex (mPFC)-dorsal raphé (DRN) serotonergic (5-HT) neurons connectivity. Spadin increased 5-HT neuron firing rate by 113%, an augmentation abolished after electrolytic lesion of the mPFC. Among the few receptor subtypes known to modulate TREK-1, the stimulation of 5-HT4 receptors and the blockade of mGluR2/3 ones both activated 5-HT impulse flow, effects also suppressed by mPFC lesion. The combination of spadin with the 5-HT4 agonist RS 67333 paradoxically reduced 5-HT firing, an effect reversed by acutely administering the 5-HT1A agonist flesinoxan. It also had a robust synergetic effect on the expression of Zif268 within the DRN. Together, these results strongly suggest that 5-HT neurons underwent a state of depolarization block, and that the mechanisms underlying the influences exerted by spadin and RS 67333 are additive and independent from each other. In contrast, the mGluR2/3 antagonist LY 341495 occluded the effect of spadin, showing that it likely depends on mPFC TREK-1 channels coupled to mGluR2/3 receptors. These in vivo electrophysiological data were confirmed by in vitro Ca(2+) cell imaging performed in cultured cortical neurons. Altogether, our results indicate that spadin, as a natural compound, constitutes a very good candidate to explore the "glutamatergic path" of fast-acting AD research. In addition, they provide the first evidence of 5-HT depolarization block, showing that the combination of 5-HT activators for strategies of AD augmentation should be performed with extreme caution.


Asunto(s)
Antidepresivos/administración & dosificación , Péptidos/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Receptores de Serotonina 5-HT4/fisiología , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/fisiología , Aminoácidos/administración & dosificación , Compuestos de Anilina/administración & dosificación , Animales , Calcio/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Indoles/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Piperazinas/administración & dosificación , Piperidinas/administración & dosificación , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/genética , Ratas Sprague-Dawley , Agonistas del Receptor de Serotonina 5-HT4/administración & dosificación , Agonistas de Receptores de Serotonina/administración & dosificación , Sulfonamidas/administración & dosificación , Xantenos/administración & dosificación
19.
Drug Deliv ; 23(8): 2772-2779, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26292058

RESUMEN

Nanosuspensions (NS) can enhance the saturation solubility and dissolution velocity of poorly soluble drugs. PEG as a non-ionic surfactant plays an important role in surface modification of nanoparticles for prolonging in vivo circulation. In this study, anti-solvent precipitation method was introduced to prepare gambogenic acid nanosuspensions (GNA-NS) with PVPK30 and PEG2000 as stabilizers to settle the disadvantages of GNA. The obtained nanoparticles were spherical with a mean particle size of 183.7 nm and a zeta potential of -22.8 mV. The entrapment efficiency and drug loading of the resultant formulation were 97.3 and 29.73%. X-ray diffraction analysis confirmed the amorphous phase of GNA in NS. Fourier transform infrared indicated there may be hydrogen bond interaction between the drug and excipients. After lyophilization of GNA-NS, the freeze-dried powder displayed sufficient long-term physical stability at 4 and 25 °C. In comparison to GNA solution, in vitro studies of GNA-NS showed much slower release and higher cytotoxicity in HepG2 cells. What's more, the pharmacokinetic study in rats revealed that the AUC0-∞ and t1/2 of GNA-NS were increased 2.63- and 1.77-fold than that of the reference formulation. Taken together, in vitro/in vivo evaluations showed NS would be an effectively strategy to change the poor aqueous solubility and prolong the half-life for GNA. The GNA-NS with enhanced bioavailability and drug efficacy provided a promising delivery system for the application of GNA.


Asunto(s)
Nanopartículas/química , Suspensiones/química , Xantenos/administración & dosificación , Xantenos/química , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Química Farmacéutica/métodos , Composición de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/métodos , Estabilidad de Medicamentos , Excipientes/química , Femenino , Liofilización/métodos , Semivida , Células Hep G2 , Humanos , Masculino , Nanopartículas/administración & dosificación , Tamaño de la Partícula , Polietilenglicoles , Polivinilos/química , Ratas , Ratas Sprague-Dawley , Solubilidad , Solventes/química , Tensoactivos/química , Suspensiones/administración & dosificación
20.
Neuropsychopharmacology ; 41(4): 1046-56, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26245499

RESUMEN

We have reported the antidepressant effects of both metabotropic glutamate 2/3 (mGlu2/3) receptor antagonists and ketamine in several animal models, and proposed that serotonergic (5-HTergic) transmission is involved in these actions. Given that the projections from the medial prefrontal cortex (mPFC) to the dorsal raphe nucleus (DRN), where the majority of serotonin (5-HT) neurons exist, are reportedly involved in the antidepressant effects, in this study, we investigated using the forced swimming test (FST) of C57BL/6J male mice, the role of 5-HT neurons in the DRN regulated by the mPFC-DRN projections in the antidepressant effects of an mGlu2/3 receptor antagonist, LY341495, and ketamine. Following systemic administration/microinjection into the mPFC, both LY341495 and ketamine were found to exert antidepressant effects in the FST, and the effects were attenuated by depletion of 5-HT by treatment with an inhibitor of 5-HT synthesis, PCPA. The antidepressant effects of LY341495 and ketamine were also blocked by systemic administration/microinjection into the mPFC of an AMPA receptor antagonist, NBQX. Moreover, systemic administration/microinjection into the mPFC of LY341495 and ketamine significantly increased the c-Fos expression in the 5-HT neurons in the DRN, and the effect of systemic administration of these drugs on the neuronal c-Fos expression was attenuated by microinjection of NBQX into the mPFC. Our findings suggest that activation of 5-HT neurons in the DRN regulated by stimulation of the AMPA receptor in the mPFC may be involved in the antidepressant effects of an mGlu2/3 receptor antagonist and ketamine.


Asunto(s)
Antidepresivos/administración & dosificación , Depresión/prevención & control , Núcleo Dorsal del Rafe/fisiología , Ketamina/administración & dosificación , Corteza Prefrontal/fisiología , Receptores AMPA/fisiología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Neuronas Serotoninérgicas/fisiología , Aminoácidos/administración & dosificación , Animales , Núcleo Dorsal del Rafe/efectos de los fármacos , Fenclonina/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Corteza Prefrontal/efectos de los fármacos , Quinoxalinas/administración & dosificación , Receptores AMPA/antagonistas & inhibidores , Neuronas Serotoninérgicas/efectos de los fármacos , Xantenos/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...