Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Drug Des Devel Ther ; 16: 4179-4204, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36514526

RESUMEN

Aim: Depression is a chronic recurrent neuropsychiatric disorder associated with inflammation. This study explored the pharmacological activities of Aerva javanica leaves crude extract (Aj.Cr) on lipopolysaccharide (LPS)-induced depressive-like behavior in experimental mice. Methods: Aj.Cr was evaluated for its phenolic and flavonoid contents, bioactive potential, amino acid profiling and enzyme inhibition assays using different analytical techniques followed by in-silico molecular docking was performed. In addition, three ligands identified in HPLC analysis and standard galantamine were docked to acetyl cholinesterase (AchE) enzyme to assess the ligand interaction along with their binding affinities. In in-vivo analysis, mice were given normal saline (10 mL/kg), imipramine (10 mg/kg) and Aj.Cr (100, 300, and 500 mg/kg) orally for 14-consecutive days. On the 14th day, respective treatment was given 30-minutes before intra-peritoneal administration of (0.83 mg/kg) LPS. Open field, forced swim and tail suspension tests were performed 24-hours after LPS injection, followed by a sucrose preference test 48-hours later. Serum corticosterone levels, as well as levels of nitric oxide (NO), malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), tumor necrosis factor-alpha (TNF-), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), brain-derived neurotrophic factor (BDNF) and catecholamines were determined in brain tissues. Results: In-vitro results revealed that crude extract of Aj.Cr possesses anti-depressant agents with solid antioxidant potential. In-vivo analysis showed that LPS significantly increased depressive-like behavior followed by alteration in serum and tissue biomarkers as compared to normal control (p < 0.001). While imipramine and Aj.Cr (100, 300, and 500 mg/kg) treated groups significantly (p<0.05) improved the depressive-like behavior and biomarkers when compared to the LPS group. Conclusion: The mitigation of LPS-induced depressive-like behavior by Aj.Cr may be linked to the modulation of oxidative stress, neuro-inflammation and catecholamines due to the presence of potent bioactive compounds exerting anti-depressant effects.


Asunto(s)
Amaranthaceae , Lipopolisacáridos , Animales , Ratones , Antidepresivos/metabolismo , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Catecolaminas/metabolismo , Catecolaminas/farmacología , Mezclas Complejas/farmacología , Depresión/inducido químicamente , Depresión/tratamiento farmacológico , Glutatión/metabolismo , Imipramina/metabolismo , Imipramina/farmacología , Inflamación/tratamiento farmacológico , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Metanol/farmacología , Simulación del Acoplamiento Molecular , Factor de Necrosis Tumoral alfa/metabolismo
2.
Cancer Cell ; 40(10): 1111-1127.e9, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36113478

RESUMEN

Glioblastoma (GBM) is poorly responsive to therapy and invariably lethal. One conceivable strategy to circumvent this intractability is to co-target distinctive mechanistic components of the disease, aiming to concomitantly disrupt multiple capabilities required for tumor progression and therapeutic resistance. We assessed this concept by combining vascular endothelial growth factor (VEGF) pathway inhibitors that remodel the tumor vasculature with the tricyclic antidepressant imipramine, which enhances autophagy in GBM cancer cells and unexpectedly reprograms immunosuppressive tumor-associated macrophages via inhibition of histamine receptor signaling to become immunostimulatory. While neither drug is efficacious as monotherapy, the combination of imipramine with VEGF pathway inhibitors orchestrates the infiltration and activation of CD8 and CD4 T cells, producing significant therapeutic benefit in several GBM mouse models. Inclusion up front of immune-checkpoint blockade with anti-programmed death-ligand 1 (PD-L1) in eventually relapsing tumors markedly extends survival benefit. The results illustrate the potential of mechanism-guided therapeutic co-targeting of disparate biological vulnerabilities in the tumor microenvironment.


Asunto(s)
Glioblastoma , Animales , Antidepresivos Tricíclicos/metabolismo , Antidepresivos Tricíclicos/uso terapéutico , Autofagia , Antígeno B7-H1/metabolismo , Glioblastoma/patología , Imipramina/metabolismo , Imipramina/uso terapéutico , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Macrófagos/metabolismo , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptor de Muerte Celular Programada 1 , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Pharm Res ; 39(2): 223-237, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35112227

RESUMEN

PURPOSE: The present study aimed to elucidate the transport properties of imipramine and paroxetine, which are the antidepressants, across the blood-brain barrier (BBB) in rats. METHODS: In vivo influx and efflux transport of imipramine and paroxetine across the BBB were tested using integration plot analysis and a combination of brain efflux index and brain slice uptake studies, respectively. Conditionally immortalized rat brain capillary endothelial cells, TR-BBB13 cells, were utilized to characterize imipramine and paroxetine transport at the BBB in vitro. RESULTS: The in vivo influx clearance of [3H]imipramine and [3H]paroxetine in rats was determined to be 0.322 mL/(min·g brain) and 0.313 mL/(min·g brain), respectively. The efflux clearance of [3H]imipramine and [3H]paroxetine was 0.380 mL/(min·g brain) and 0.126 mL/(min·g brain), respectively. These results suggest that the net flux of paroxetine, but not imipramine, at the BBB in vivo was dominated by transport to the brain from the circulating blood. The uptake of imipramine and paroxetine by TR-BBB13 cells exhibited time- and temperature-dependence and one-saturable kinetics with a Km of 37.6 µM and 89.2 µM, respectively. In vitro uptake analyses of extracellular ion dependency and the effect of substrates/inhibitors for organic cation transporters and transport systems revealed minor contributions to known transporters and transport systems and the difference in transport properties in the BBB between imipramine and paroxetine. CONCLUSIONS: Our study showed the comprehensive outcomes of imipramine and paroxetine transport at the BBB, implying that molecular mechanism(s) distinct from previously reported transporters and transport systems are involved in the transport.


Asunto(s)
Antidepresivos de Segunda Generación/metabolismo , Antidepresivos Tricíclicos/metabolismo , Barrera Hematoencefálica/metabolismo , Imipramina/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Paroxetina/metabolismo , Animales , Antidepresivos de Segunda Generación/administración & dosificación , Antidepresivos Tricíclicos/administración & dosificación , Transporte Biológico , Línea Celular , Imipramina/administración & dosificación , Inyecciones Intravenosas , Cinética , Masculino , Modelos Biológicos , Paroxetina/administración & dosificación , Permeabilidad , Ratas Wistar
4.
Behav Brain Res ; 364: 274-280, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30738101

RESUMEN

Depression is associated with dysregulation of methyl group metabolism such as low S-adenosylmethionine (SAM). We previously reported that Flinders Sensitive Line (FSL) rats, an animal model of depression, had lower concentrations of liver SAM than the control rats, Flinders Resistant Line (FRL) rats. The present study investigated if SAM supplementation may correct liver SAM and behavioral abnormalities in this model. Moreover, we compared one-carbon (C1) metabolites, neurotransmitters, and gastrointestinal (GI) transit in SAM-treated versus imipramine (IMI)-treated animals. FSL rats received vehicle, IMI, SAM, or IMI + SAM (n = 9-10 per group) once daily through oral gavage for 4 weeks; FRL rats received vehicle. Behavior was assessed using standard tests for locomotion, cognition, and depressive-like behavior. Monoamine neurotransmitters and C1 metabolites were measured using UHPLC-ECD and UPLC-MS/MS, respectively. Compared to FRL rats, FSLs had lower liver SAM, higher plasma serotonin, lower hippocampal dopamine and serotonin turnover, and faster GI transit. Behaviorally, FSL rats showed impaired cognitive performance as well as increased depressive-like behavior compared to FRLs. Coadministration of IMI and SAM seemed to have adverse effects on spatial memory. SAM or IMI administration did not reverse C1 metabolites, neurotransmitters, or GI transit in FSLs. Despite low liver SAM in FSL rats, orally administered SAM did not show antidepressant effects in this specific animal model of depression.


Asunto(s)
Depresión/metabolismo , Imipramina/farmacología , S-Adenosilmetionina/farmacología , Animales , Antidepresivos/farmacología , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Depresión/tratamiento farmacológico , Depresión/fisiopatología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Hipocampo/metabolismo , Imipramina/metabolismo , Masculino , Ratas , Ratas Endogámicas , S-Adenosilmetionina/metabolismo , Serotonina/metabolismo , Memoria Espacial/efectos de los fármacos
5.
Behav Brain Res ; 336: 99-110, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28866130

RESUMEN

Depression is commonly associated with hypothalamic-pituitary adrenal (HPA) axis dysfunction that primarily manifests as aberrant glucocorticoid secretion. Glucocorticoids act on Type I mineralocorticoid (MR) and Type II glucocorticoid receptors (GR) to modulate mood and endocrine responses. Successful antidepressant treatment normalizes HPA axis function, in part due to modulatory effects on MR and GR in cortico-limbic structures. Although women are twice as likely to suffer from depression, little is known about how antidepressants modulate brain, endocrine, and behavioral stress responses in females. Here, we assessed the impact of CORT118335 (GR modulator/MR antagonist) and imipramine (tricyclic antidepressant) on neuroendocrine and behavioral responses to restraint or forced swim stress (FST) in female rats (n=10-12/group). Increased immobility CORT118335 in the FST is purported to reflect passive coping or depression-like behavior. CORT118335 dampened adrenocorticotropic hormone (ACTH) and corticosterone responses to the FST, but did not affect immobility. Imipramine suppressed ACTH, but had minimal effects on corticosterone responses to FST. Despite these marginal effects, imipramine decreased immobility, suggesting antidepressant efficacy. In an effort to link brain-endocrine responses with behavior, c-Fos was assessed in HPA axis and mood modulatory regions in response to the FST. CORT118335 upregulated c-Fos expression in the paraventricular nucleus of the hypothalamus. Imipramine decreased c-Fos in the basolateral amygdala and hippocampus (CA1 and CA3), but increased c-Fos in the central amygdala. These data suggest the antidepressant-like (e.g., active coping) properties of imipramine may be due to widespread effects on cortico-limbic circuits that regulate emotional and cognitive processes.


Asunto(s)
Imipramina/farmacología , Estrés Fisiológico/efectos de los fármacos , Timina/análogos & derivados , Hormona Adrenocorticotrópica/farmacología , Animales , Antidepresivos/farmacología , Antidepresivos Tricíclicos/farmacología , Conducta Animal/fisiología , Encéfalo/metabolismo , Corticosterona/metabolismo , Depresión/fisiopatología , Trastorno Depresivo/fisiopatología , Femenino , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Imipramina/metabolismo , Antagonistas de Receptores de Mineralocorticoides/metabolismo , Antagonistas de Receptores de Mineralocorticoides/farmacología , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología , Timina/metabolismo , Timina/farmacología
7.
Biopharm Drug Dispos ; 35(6): 313-20, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24752421

RESUMEN

The microminipig, a small minipig, was bred as a novel experimental animal for nonclinical pharmacology/toxicology studies by Fuji Micra Inc. (Shizuoka, Japan). Species differences in drug metabolism between humans and the microminipig need to be elucidated in more detail in order to discuss the results of nonclinical studies. Glucuronidation catalysed by UDP-glucuronosyltransferase (UGT) is an important pathway in the metabolism of a wide variety of compounds. The aim of the present study was to identify the characteristics of hepatic UGT activity in the microminipig and compare them with those in humans and other experimental animals. This study examined in vitro UGT activities using liver microsomes from microminipigs (8 months old and 1 day old), humans, mice, rats, dogs, monkeys and minipigs. The glucuronides of estradiol, imipramine, serotonin, propofol, 3'-azido-3'-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Estradiol-3-glucuronidation activity was higher in the microminipig than in humans and the other animals. High levels of estradiol-3-glucuronidation were observed in the microsomes of 1-day-old microminipigs. Imipramine-N-glucuronidation, a distinctive conjugation by human UGT1A4, was catalysed by microminipig liver microsomes, but not by dog liver microsomes. Although AZT-glucuronidation activity was low in the microminipig compared with humans, morphine-3-glucuronidation activity in the microminipig was higher than that in humans. The UGT activities in the microminipig were similar to those in the minipig. The results of the present study have provided useful information for selecting an appropriate animal model for nonclinical studies.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Microsomas Hepáticos/metabolismo , Porcinos Enanos , Adulto , Anciano , Animales , Cromatografía Liquida , Perros , Estradiol/metabolismo , Femenino , Glucurónidos/metabolismo , Humanos , Imipramina/metabolismo , Macaca fascicularis , Masculino , Ratones Endogámicos ICR , Persona de Mediana Edad , Morfina/metabolismo , Propofol/metabolismo , Ratas Sprague-Dawley , Serotonina/metabolismo , Especificidad de la Especie , Porcinos , Espectrometría de Masas en Tándem , Adulto Joven , Zidovudina/metabolismo
8.
J Pharmacol Exp Ther ; 344(1): 113-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23010362

RESUMEN

The interaction of the selective norepinephrine reuptake inhibitor (-)-reboxetine with the human α4ß2 nicotinic acetylcholine receptor (nAChR) in different conformational states was studied by several functional and structural approaches. Patch-clamp and Ca(2+)-influx results indicate that (-)-reboxetine does not activate hα4ß2 nAChRs via interaction with the orthosteric sites, but inhibits agonist-induced hα4ß2 activation by a noncompetitive mechanism. Consistently, the results from the electrophysiology-based functional approach suggest that (-)-reboxetine may act via open channel block; therefore, it is capable of producing a use-dependent type of inhibition of the hα4ß2 nAChR function. We tested whether (-)-reboxetine binds to the luminal [(3)H]imipramine site. The results indicate that, although (-)-reboxetine binds with low affinity to this site, it discriminates between the resting and desensitized hα4ß2 nAChR ion channels. Patch-clamp results also indicate that (-)-reboxetine progressively inhibits the hα4ß2 nAChR with two-fold higher potency at the end of one-second application of agonist, compared with the peak current. The molecular docking studies show that (-)-reboxetine blocks the ion channel at the level of the imipramine locus, between M2 rings 6' and 14'. In addition, we found a (-)-reboxetine conformer that docks in the helix bundle of the α4 subunit, near the middle region. According to molecular dynamics simulations, (-)-reboxetine binding is stable for both sites, albeit less stable than imipramine. The interaction of these drugs with the helix bundle might alter allostericaly the functionality of the channel. In conclusion, the clinical action of (-)-reboxetine may be produced (at least partially) by its inhibitory action on hα4ß2 nAChRs.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Morfolinas/farmacología , Receptores Nicotínicos/metabolismo , Inhibidores de Captación Adrenérgica/química , Alcaloides/metabolismo , Animales , Azocinas/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/antagonistas & inhibidores , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Fenómenos Electrofisiológicos , Células Epiteliales/efectos de los fármacos , Células HEK293 , Humanos , Imipramina/metabolismo , Modelos Moleculares , Conformación Molecular , Morfolinas/química , Agonistas Nicotínicos/farmacología , Técnicas de Placa-Clamp , Piridinas/antagonistas & inhibidores , Piridinas/farmacología , Quinolizinas/metabolismo , Ensayo de Unión Radioligante , Reboxetina , Receptores Nicotínicos/química , Receptores Nicotínicos/efectos de los fármacos , Torpedo
9.
Protein Expr Purif ; 76(2): 211-20, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21129485

RESUMEN

The serotonin transporter (SERT), a member of the solute carrier 6 family, is responsible for reuptake of the monoamine neurotransmitter serotonin (5-hydroxytryptamine) from the synaptic cleft on the neural cells, and a vital target for several antidepressants. To investigate biophysical studies of this pharmacologically relevant transporter, we developed a mammalian expression system with tetracycline-inducible HEK293 cells using synthetic human SERT genes produced by PCR-based self-assembly method. Codon-optimization of this de novo constructed genes and construction of stable cell lines improved expression 3.5-fold and single-step immunoaffinity purification with FLAG-epitope tag yielded around one milligram functional SERT per liter culture medium assessed by [(3)H] imipramine ligand binding. Some characterizations including electrospray ionization MS/MS analysis, subcellular localization and cellular-uptake assay demonstrated that expressed human SERT was properly expressed, folded and fully functional. The long cytosolic N-terminal of SERT was predicted as containing 'intrinsically disordered region (IDR)' (∼85 residues) by DISOPRED2 program. We engineered this salient region by step-wise truncation and ligand binding assay determined that dissociation constant for a series of de novo designed truncation constructs was close to the one for full-length wild type SERT. Our expression platform using synthetic codon-optimized gene and mammalian stable cell lines is feasible to produce milligram-scale functional membrane transporter for further biophysical and biochemical studies.


Asunto(s)
Proteínas Recombinantes de Fusión/biosíntesis , Proteínas de Transporte de Serotonina en la Membrana Plasmática/biosíntesis , Tetraciclina/farmacología , Secuencia de Aminoácidos , Western Blotting , Cromatografía de Afinidad , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Glicosilación , Células HEK293 , Humanos , Imipramina/análisis , Imipramina/metabolismo , Espacio Intracelular/metabolismo , Microscopía Fluorescente , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Oligopéptidos , Péptidos/genética , Péptidos/metabolismo , Conformación Proteica , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Espectrometría de Masas en Tándem , Tritio/análisis
10.
Drug Metab Dispos ; 38(5): 863-70, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20133892

RESUMEN

The role of human UDP glucuronosyltransferase (UGT) 2B10 in the N-glucuronidation of a number of tricyclic antidepressants was investigated and compared with that of UGT1A4 in both the Sf9 expressed system and human liver microsomes. The apparent K(m) (S(50)) values for the formation of quaternary N-glucuronides of amitriptyline, imipramine, clomipramine, and trimipramine were 2.60, 16.8, 14.4, and 11.2 microM in UGT2B10 and 448, 262, 112, and 258 microM in UGT1A4, respectively. The kinetics of amitriptyline and imipramine glucuronidation in human liver microsomes exhibited a biphasic character, where the high- and low-affinity components were in good agreement with our results in expressed UGT2B10 and UGT1A4, respectively. The kinetics of clomipramine and trimipramine glucuronidation in human liver microsomes were sigmoidal in nature, and the S(50) values were similar to those found for expressed UGT1A4. The in vitro clearances (CL(int) or CL(max)) were comparable between UGT2B10 and UGT1A4 for glucuronidation of imipramine, clomipramine, and trimipramine, whereas CL(int) of amitriptyline glucuronidation by UGT2B10 was more than 10-fold higher than that by UGT1A4. Nicotine was found to selectively inhibit UGT2B10 but not UGT1A4 activity. At a low tricyclic antidepressant concentration, nicotine inhibited their glucuronidation by 33 to 50% in human liver microsomes. Our results suggest that human UGT2B10 is a high-affinity enzyme for tricyclic antidepressant glucuronidation and is likely to be a major UGT isoform responsible for the glucuronidation of these drugs at therapeutic concentrations in vivo.


Asunto(s)
Amitriptilina/metabolismo , Antidepresivos Tricíclicos/metabolismo , Clomipramina/metabolismo , Ácido Glucurónico/metabolismo , Glucuronosiltransferasa/metabolismo , Imipramina/metabolismo , Trimipramina/metabolismo , Biocatálisis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucurónidos/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Glucuronosiltransferasa/genética , Humanos , Cinética , Microsomas Hepáticos/enzimología , Nicotina/farmacología , Proteínas Recombinantes/metabolismo , Sapogeninas/farmacología
11.
PLoS One ; 4(9): e7197, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19787068

RESUMEN

BACKGROUND: Dietary lipids or pharmacologic modulation of lipid metabolism are potential therapeutic strategies in inflammatory bowel disease (IBD). Therefore, we analysed alterations of bioactive lipids in experimental models of colitis and examined the functional consequence of the second messenger ceramide in inflammatory pathways leading to tissue destruction. METHODOLOGY/PRINCIPAL FINDINGS: Chronic colitis was induced by dextran-sulphate-sodium (DSS) or transfer of CD4(+)CD62L(+) cells into RAG1(-/-)-mice. Lipid content of isolated murine intestinal epithelial cells (IEC) was analysed by tandem mass spectrometry. Concentrations of MMP-1 in supernatants of Caco-2-IEC and human intestinal fibroblasts from patients with ulcerative colitis were determined by ELISA. Imipramine was used for pharmacologic inhibition of acid sphingomyelinase (ASM). Ceramide increased by 71% in chronic DSS-induced colitis and by 159% in the transfer model of colitis. Lysophosphatidylcholine (LPC) decreased by 22% in both models. No changes were detected for phosphatidylcholine. Generation of ceramide by exogenous SMase increased MMP-1-protein production of Caco-2-IEC up to 7-fold. Inhibition of ASM completely abolished the induction of MMP-1 by TNF or IL-1beta in Caco-2-IEC and human intestinal fibroblasts. CONCLUSIONS/SIGNIFICANCE: Mucosal inflammation leads to accumulation of ceramide and decrease of LPC in the intestinal epithelium. One aspect of ceramide generation is an increase of MMP-1. Induction of MMP-1 by TNF or IL-1beta is completely blocked by inhibition of ASM with imipramine. Therefore, inhibition of ASM may offer a treatment strategy to reduce MMP-1 expression and tissue destruction in inflammatory conditions.


Asunto(s)
Ceramidas/metabolismo , Colitis/patología , Regulación Enzimológica de la Expresión Génica , Imipramina/metabolismo , Interleucina-1beta/metabolismo , Lípidos/química , Metaloproteinasa 1 de la Matriz/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células CACO-2 , Humanos , Selectina L/biosíntesis , Selectina L/metabolismo , Metabolismo de los Lípidos , Lisofosfatidilcolinas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Esfingomielina Fosfodiesterasa/metabolismo
12.
Biochemistry ; 48(21): 4506-18, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19334677

RESUMEN

To characterize the binding sites and the mechanisms of inhibition of bupropion on muscle-type nicotinic acetylcholine receptors (AChRs), structural and functional approaches were used. The results established that bupropion (a) inhibits epibatidine-induced Ca(2+) influx in embryonic muscle AChRs, (b) inhibits adult muscle AChR macroscopic currents in the resting/activatable state with approximately 100-fold higher potency compared to that in the open state, (c) increases the desensitization rate of adult muscle AChRs from the open state and impairs channel opening from the resting state, (d) inhibits binding of [(3)H]TCP and [(3)H]imipramine to the desensitized/carbamylcholine-bound Torpedo AChR with higher affinity compared to the resting/alpha-bungarotoxin-bound AChR, (e) binds to the Torpedo AChR in either state mainly by an entropy-driven process, and (f) interacts with a binding domain located between the serine (position 6') and valine (position 13') rings, by a network of van der Waals, hydrogen bond, and polar interactions. Collectively, our data indicate that bupropion first binds to the resting AChR, decreasing the probability of ion channel opening. The remnant fraction of open ion channels is subsequently decreased by accelerating the desensitization process. Bupropion interacts with a luminal binding domain shared with PCP that is located between the serine and valine rings, and this interaction is mediated mainly by an entropy-driven process.


Asunto(s)
Bupropión/metabolismo , Músculos/metabolismo , Antagonistas Nicotínicos/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Animales , Unión Competitiva , Transporte Biológico/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Bupropión/farmacología , Calcio/metabolismo , Línea Celular , Descubrimiento de Drogas , Conductividad Eléctrica , Humanos , Imipramina/metabolismo , Proteínas Inmovilizadas/antagonistas & inhibidores , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Activación del Canal Iónico , Cinética , Ratones , Modelos Moleculares , Antagonistas Nicotínicos/farmacología , Piperidinas/química , Piperidinas/metabolismo , Unión Proteica , Conformación Proteica , Piridinas/farmacología , Termodinámica , Torpedo
13.
EMBO J ; 28(8): 1043-54, 2009 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-19300439

RESUMEN

We have earlier shown that microglia, the immune cells of the CNS, release microparticles from cell plasma membrane after ATP stimulation. These vesicles contain and release IL-1beta, a crucial cytokine in CNS inflammatory events. In this study, we show that microparticles are also released by astrocytes and we get insights into the mechanism of their shedding. We show that, on activation of the ATP receptor P2X7, microparticle shedding is associated with rapid activation of acid sphingomyelinase, which moves to plasma membrane outer leaflet. ATP-induced shedding and IL-1beta release are markedly reduced by the inhibition of acid sphingomyelinase, and completely blocked in glial cultures from acid sphingomyelinase knockout mice. We also show that p38 MAPK cascade is relevant for the whole process, as specific kinase inhibitors strongly reduce acid sphingomyelinase activation, microparticle shedding and IL-1beta release. Our results represent the first demonstration that activation of acid sphingomyelinase is necessary and sufficient for microparticle release from glial cells and define key molecular effectors of microparticle formation and IL-1beta release, thus, opening new strategies for the treatment of neuroinflammatory diseases.


Asunto(s)
Micropartículas Derivadas de Células/enzimología , Neuroglía/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Inhibidores de Captación Adrenérgica/metabolismo , Marcadores de Afinidad/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Micropartículas Derivadas de Células/ultraestructura , Células Cultivadas , Activación Enzimática , Imipramina/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Neuroglía/citología , Tamaño de la Partícula , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/metabolismo
14.
J Mass Spectrom ; 43(9): 1181-90, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18300330

RESUMEN

A control sample background-subtraction algorithm was developed for thorough subtraction of background and matrix-related signals in high-resolution, accurate mass liquid chromatography/mass spectrometry (LC/MS) data to reveal ions of interest in an analyte sample. This algorithm checked all ions in the control scans within a specified time window around the analyte scan for potential subtraction of ions found in that analyte scan. Applying this method, chromatographic fluctuations between runs were dealt with and background and matrix-related signals in the sample could be thoroughly subtracted. The effectiveness of this algorithm was demonstrated using four test compounds, clozapine, diclofenac, imipramine, and tacrine, to reveal glutathione (GSH)-trapped reactive metabolites after incubation with human liver microsomes supplemented with GSH (30 microM compound, 45-min incubation). Using this algorithm with a+/-1.0 min control scan time window, a+/-5 ppm mass error tolerance, and appropriate control samples, the GSH-trapped metabolites were revealed as the major peaks in the processed LC/MS profiles. Such profiles allowed for comprehensive and reliable identification of these metabolites without the need for any presumptions regarding their behavior or properties with respect to mass spectrometric detection. The algorithm was shown to provide superior results when compared to several commercially available background-subtraction algorithms. Many of the metabolites detected were doubly charged species which would be difficult to detect with traditional GSH adduct screening techniques, and thus, some of the adducts have not previously been reported in the literature.


Asunto(s)
Algoritmos , Glutatión/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Cromatografía Líquida de Alta Presión , Clozapina/metabolismo , Diclofenaco/metabolismo , Glutatión/análogos & derivados , Humanos , Imipramina/metabolismo , Microsomas Hepáticos/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier/estadística & datos numéricos , Tacrina/metabolismo
15.
Drug Metab Dispos ; 35(10): 1781-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17620344

RESUMEN

Protein-protein interactions between human UDP-glucuronosyltransferase (UGT) 1A1, UGT1A4, and UGT1A6 were investigated using double expression systems in HEK293 cells (UGT1A1/UGT1A4, UGT1A1/UGT1A6, and UGT1A4/UGT1A6). The substrates specific for UGT1A1 (estradiol and bilirubin), UGT1A4 (imipramine and trifluoperazine), and UGT1A6 (serotonin and diclofenac) were used to determine the effects of the coexpression of the other UGT1A isoforms on the enzymatic activity. The coexpression of UGT1A4 and UGT1A6 decreased the S(50) and V(max) values of UGT1A1-catalyzed estradiol 3-O-glucuronide formation and increased the V(max) value of UGT1A1-catalyzed bilirubin O-glucuronide formation. The coexpression of UGT1A1 decreased the V(max) value of UGT1A4-catalyzed imipramine N-glucuronide formation but had no effect on UGT1A4-catalyzed trifluoperazine N-glucuronide formation. The coexpression of UGT1A6 had no effect on UGT1A4-catalyzed imipramine N-glucuronide formation but increased the K(m) and V(max) of UGT1A4-catalyzed trifluoperazine N-glucuronide formation. The coexpression of both UGT1A1 and UGT1A4 increased the V(max) values of UGT1A6-catalyzed serotonin and diclofenac O-glucuronide formation. Thus, the effects of the coexpression of other UGT1A isoforms on the kinetics of specific activities were different depending on the UGT1A isoforms and substrates. Native polyacrylamide gel electrophoresis analysis of the double expression systems showed multiple bands at approximately 110 kDa, indicating the existence of heterodimers as well as homodimers of UGTs. In conclusion, we found that human UGT1A1, UGT1A4, and UGT1A6 interact with each other, possibly by heterodimerization, and that their effects on the enzymatic activities are complex depending on the isoforms and substrates.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Bilirrubina/metabolismo , Catálisis , Línea Celular , Diclofenaco/metabolismo , Dimerización , Estradiol/metabolismo , Glucurónidos/metabolismo , Humanos , Imipramina/metabolismo , Isoenzimas/metabolismo , Serotonina/metabolismo , Trifluoperazina/metabolismo
16.
Drug Metab Dispos ; 35(5): 747-57, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17293379

RESUMEN

We established stable HEK293 cell lines expressing double isoforms, UGT1A1 and UGT1A9, UGT1A4 and UGT1A9, or UGT1A6 and UGT1A9, as well as stable cell lines expressing each single isoform. To analyze the protein-protein interaction between the UGT1As, we investigated the thermal stability and resistance to detergent. UGT1A9 uniquely demonstrated thermal stability, which was enhanced in the presence of UDP-glucuronic acid (>90% of control), and resistance to detergent. Interestingly, UGT1A1, UGT1A4, and UGT1A6 acquired thermal stability and resistance to detergent by the coexpression of UGT1A9. An immunoprecipitation assay revealed that UGT1A6 and UGT1A9 interact in the double expression system. Using the single expression systems, it was confirmed that estradiol 3-O-glucuronide, imipramine N-glucuronide, serotonin O-glucuronide, and propofol O-glucuronide formations are specific for UGT1A1, UGT1A4, UGT1A6, and UGT1A9, respectively. By kinetic analyses, we found that the coexpressed UGT1A9 significantly affected the kinetics of estradiol 3-O-glucuronide formation (decreased Vmax), imipramine N-glucuronide formation (increased Km and Vmax), and serotonin O-glucuronide formation (decreased Vmax) catalyzed by UGT1A1, UGT1A4, and UGT1A6, respectively. On the other hand, the coexpressed UGT1A1 increased Km and decreased the Vmax of the propofol O-glucuronide formation catalyzed by UGT1A9. The coexpressed UGT1A4 and UGT1A6 also increased the Vmax of the propofol O-glucuronide formation by UGT1A9. This is the first study showing that human UGT1A isoforms interact with other isoforms to change the enzymatic characteristics.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glucuronosiltransferasa/metabolismo , Catálisis/efectos de los fármacos , Línea Celular , Detergentes/farmacología , Estabilidad de Enzimas/efectos de los fármacos , Estradiol/metabolismo , Glucurónidos/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Glucuronosiltransferasa/genética , Humanos , Imipramina/análogos & derivados , Imipramina/metabolismo , Immunoblotting , Inmunoprecipitación , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Octoxinol/farmacología , Propofol/metabolismo , Serotonina/metabolismo , Temperatura , Transfección , UDP Glucuronosiltransferasa 1A9
17.
J Pharmacol Exp Ther ; 321(1): 237-48, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17237257

RESUMEN

Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide that plays a role in the modulation of food intake and mood. In rodents, the actions of MCH are mediated via the MCHR1 receptor. The goal of this study was to investigate the effects of acute (1 h) and chronic (28 days) p.o. dosing of a novel MCHR1 antagonist, N-[3-(1-{[4-(3,4-difluorophenoxy)-phenyl]methyl}(4-piperidyl))-4-methylphenyl]-2-methylpropanamide (SNAP 94847), in three mouse models predictive of antidepressant/anxiolytic-like activity: novelty suppressed feeding (NSF) in 129S6/SvEvTac mice and light/dark paradigm (L/D) and forced swim test (FST) in BALB/cJ mice. A significant increase in the time spent in the light compartment of the L/D box was observed in response to acute and chronic treatment with SNAP 94847. An anxiolytic/antidepressant-like effect was found in the NSF test after acute and chronic treatment, whereas no effect was observed in the FST. Because neurogenesis in the dentate gyrus has been shown to be a requirement for the effects of antidepressants in the NSF test, we investigated whether neurogenesis was required for the effect of SNAP 94847. We showed that chronic treatment with SNAP 94847 stimulated proliferation of progenitors in the dentate gyrus. The efficacy of SNAP 94847 in the NSF test, however, was unaltered in mice in which neurogenesis was suppressed by X-irradiation. These results indicate that SNAP 94847 has a unique anxiolytic-like profile after both acute and chronic administration and that its mechanism of action is distinct from that of selective serotonin reuptake inhibitors and tricyclic antidepressants.


Asunto(s)
Ansiolíticos , Antidepresivos , Ansiedad/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Piperidinas/farmacología , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Antidepresivos Tricíclicos/metabolismo , Antimetabolitos , Ansiedad/psicología , Bromodesoxiuridina , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citalopram/metabolismo , Evaluación Preclínica de Medicamentos , Conducta Alimentaria/efectos de los fármacos , Hipocampo/citología , Imipramina/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo , Rayos X
18.
Biochim Biophys Acta ; 1760(11): 1704-12, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17008009

RESUMEN

Imatinib is a selective tyrosine kinase inhibitor, successfully used for the treatment of chronic myelogenous leukaemia. Its strong plasma protein binding referred to alpha1-acid glycoprotein (AGP) component was found to inhibit the pharmacological activity. AGP shows genetic polymorphism and the two main genetic variants have different drug binding properties. The binding characteristics of imatinib to AGP genetic variants and the possibility of its binding interactions were investigated by various methods. The results proved that binding of imatinib to the two main genetic variants is very different, the high affinity binding belongs dominantly to the F1-S variant. This interaction is accompanied with specific spectral changes (induced circular dichroism, UV change, intrinsic fluorescence quenching), suggesting that the bound ligand has chiral conformation that would largely overlap with other ligands inside the protein cavity. Binding parameters of Ka=1.7(+/-0.2)x10(6)M(-1) and n=0.94 could be determined for the binding on the F1-S variant at 37 degrees . Imatinib binding on the A variant is weaker and less specific. The binding affinity of imatinib to human serum albumin (nKa approximately 3 x 10(4)M(-1)) is low. Pharmacologically relevant binding interactions with other drugs can be expected on the F1-S variant of AGP.


Asunto(s)
Orosomucoide/efectos de los fármacos , Orosomucoide/genética , Piperazinas/química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Naranja de Acridina/química , Naranja de Acridina/metabolismo , Benzamidas , Sitios de Unión , Dicroismo Circular , Humanos , Mesilato de Imatinib , Imipramina/química , Imipramina/metabolismo , Naftalenosulfonatos/química , Naftalenosulfonatos/metabolismo , Orosomucoide/química , Piperazinas/metabolismo , Polimorfismo Genético , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Pirimidinas/metabolismo , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta , Relación Estructura-Actividad
19.
Toxicol Appl Pharmacol ; 205(3): 237-46, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15922009

RESUMEN

The drug-toxicant interaction between the antidepressant imipramine (IMI) and three organophosphorothionate pesticides (OPTs), to which humans may be chronically and simultaneously exposed, has been investigated in vitro. Concentrations of IMI (2-400 microM) and OPTs (< or =10 microM) representative of actual human exposure have been tested with recombinant human CYPs and human liver microsomes (HLM). The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. The OPTs significantly inhibited (up to >80%) IMI bioactivation catalyzed by the recombinant CYPs tested, except CYP2D6, and by HLM; the inhibition was dose-dependent and started at low pesticide concentrations (0.25-2.5 microM). The OPTs, having lower K(m) values, efficiently competed with IMI for the enzyme active site, as in the case of CYP2C19. However, with CYP1A2 and CYP3A4, a time- and NADPH-dependent mechanism-based inactivation also occurred, consistently with irreversible inhibition due to the release of the sulfur atom, binding to the active CYP during OPT desulfuration. At low IMI and OPT concentrations, lower IC50 values have been obtained with recombinant CYP1A2 (0.7-1.1 microM) or with HLM rich in 1A2-related activity (2-10.8 microM). The K(i) values (2-14 microM), independent on substrate concentrations, were quite low and similar for the three pesticides. Exposure to OPTs during IMI therapeutic treatments may lead to decreased DES formation, resulting in high plasma levels of the parent drug, eventual impairment of its pharmacological action and possible onset of adverse drug reactions (ADRs).


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Imipramina/antagonistas & inhibidores , Imipramina/metabolismo , Hígado/enzimología , Compuestos Organotiofosforados/efectos adversos , Biotransformación/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Interacciones Farmacológicas , Humanos , Imipramina/uso terapéutico , Hígado/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Compuestos Organotiofosforados/química , Oxidorreductasas N-Desmetilantes , Plaguicidas/efectos adversos , Plaguicidas/química
20.
SAR QSAR Environ Res ; 16(1-2): 79-91, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15844444

RESUMEN

In the present study, we investigated structure-permeability relationships for the blood-brain barrier (BBB) of 16 imipramine and phenothiazine derivatives. The compounds belong to structurally related chemical classes of catamphiphiles, representatives of which have previously been investigated for membrane activity and ability to overcome multidrug resistance (MDR) in tumour cells. These studies show that phenothiazines and structurally related drugs (imipramines, thioxanthenes, acridines) interact with membrane phospholipids, and additionally inhibit the MDR transport P-glycoprotein. This study aimed to identify common 3D structural characteristics of these compounds related to their mechanism of transport across the BBB. For this purpose Genetic Algorithm Similarity Programme (GASP), Comparative Molecular Field Analysis (CoMFA) and Comparative Molecular Similarity Index Analysis (CoMSIA) were applied. The results demonstrate the importance of the spatial distribution of molecular hydrophobicity for the BBB penetration of the investigated compounds. It suggests that the compounds should follow a specific profile of two hydrophobic and one hydrophilic centres in a particular space configuration, for optimal BBB penetration.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Imipramina/metabolismo , Fenotiazinas/metabolismo , Relación Estructura-Actividad Cuantitativa , Interacciones Hidrofóbicas e Hidrofílicas , Imipramina/análogos & derivados , Modelos Biológicos , Conformación Molecular , Fenotiazinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA