Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
BMB Rep ; 55(6): 293-298, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35651327

RESUMEN

Antipsychotics have been widely accepted as a treatment of choice for psychiatric illnesses such as schizophrenia. While atypical antipsychotics such as aripiprazole are not associated with obesity and diabetes, olanzapine is still widely used based on the anticipation that it is more effective in treating severe schizophrenia than aripiprazole, despite its metabolic side effects. To address metabolic problems, metformin is widely prescribed. Hypothalamic proopiomelanocortin (POMC) neurons have been identified as the main regulator of metabolism and energy expenditure. Although the relation between POMC neurons and metabolic disorders is well established, little is known about the effects of olanzapine and metformin on hypothalamic POMC neurons. In the present study, we investigated the effect of olanzapine and metformin on the hypothalamic POMC neurons in female mice. Olanzapine administration for 5 days significantly decreased Pomc mRNA expression, POMC neuron numbers, POMC projections, and induced leptin resistance before the onset of obesity. It was also observed that coadministration of metformin with olanzapine not only increased POMC neuron numbers and projections but also improved the leptin response of POMC neurons in the olanzapine-treated female mice. These findings suggest that olanzapine-induced hypothalamic POMC neuron abnormality and leptin resistance, which can be ameliorated by metformin administration, are the possible causes of subsequent hyperphagia. [BMB Reports 2022; 55(6): 293-298].


Asunto(s)
Antipsicóticos , Metformina , Animales , Antipsicóticos/metabolismo , Antipsicóticos/farmacología , Aripiprazol/metabolismo , Aripiprazol/farmacología , Femenino , Hipotálamo/metabolismo , Leptina/metabolismo , Metformina/metabolismo , Metformina/farmacología , Ratones , Neuronas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Olanzapina/metabolismo , Olanzapina/farmacología , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proopiomelanocortina/farmacología
2.
Cell Rep ; 29(6): 1524-1538.e6, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693893

RESUMEN

Exercise engages signaling networks to control the release of circulating factors beneficial to health. However, the nature of these networks remains undefined. Using high-throughput phosphoproteomics, we quantify 20,249 phosphorylation sites in skeletal muscle-like myotube cells and monitor their responses to a panel of cell stressors targeting aspects of exercise signaling in vivo. Integrating these in-depth phosphoproteomes with the phosphoproteome of acute aerobic exercise in human skeletal muscle suggests that co-administration of ß-adrenergic and calcium agonists would activate complementary signaling relevant to this exercise context. The phosphoproteome of cells treated with this combination reveals a surprising divergence in signaling from the individual treatments. Remarkably, only the combination treatment promotes multisite phosphorylation of SERBP1, a regulator of Serpine1 mRNA stability, a pro-fibrotic secreted protein. Secretome analysis reveals that the combined treatments decrease secretion of SERPINE1 and other deleterious factors. This study provides a framework for dissecting phosphorylation-based signaling relevant to acute exercise.


Asunto(s)
Ejercicio Físico/fisiología , Músculo Esquelético/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Proteoma/metabolismo , Transducción de Señal/fisiología , Estrés Fisiológico/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Agonistas Adrenérgicos beta/metabolismo , Animales , Aripiprazol/metabolismo , Aripiprazol/farmacología , Calcio/agonistas , Calcio/metabolismo , Interacciones Farmacológicas , Humanos , Isoproterenol/metabolismo , Isoproterenol/farmacología , Espectrometría de Masas , Ratones , Fosfoproteínas/química , Fosforilación , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Sistemas de Translocación de Proteínas/genética , Sistemas de Translocación de Proteínas/metabolismo , Proteoma/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ratas , Estrés Fisiológico/fisiología , Tapsigargina/metabolismo , Tapsigargina/farmacología
3.
Psychoneuroendocrinology ; 109: 104400, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31404896

RESUMEN

INTRODUCTION: Second-generation antipsychotics cause serious metabolic side effects, but the mechanisms behind these effects remain largely unknown. However, emerging evidence supports that antipsychotics may act upon the hypothalamus, the primary brain region understood to regulate energy homeostasis. We have recently reported that the antipsychotics olanzapine, clozapine, and aripiprazole can directly act on hypothalamic rat neurons (rHypoE-19) to impair insulin, energy sensing, and modulate inflammatory pathways. In the current paper, we sought to replicate these findings to a mouse neuronal model. METHODS: The mouse hypothalamic neuronal cell line, mHypoE-46, was treated with olanzapine, clozapine, or aripiprazole. Western blots were used to measure the energy sensing protein AMPK, components of the insulin signalling pathway (AKT, GSK3ß), and components of the MAPK pathway (ERK1/2, JNK, p38), the latter linked to inflammation. RT-qPCR was used to measure mRNA expression of the inflammatory mediators IL-6, IL-10, and BDNF, well as putative receptors in the mHypoE-46 (current) and the rHypoE-19 (previously studied) cell lines. RESULTS: In the mHypoE-46 neurons, olanzapine and aripiprazole increased AMPK phosphorylation, while clozapine and aripiprazole inhibited insulin-induced phosphorylation of AKT. Clozapine increased JNK and aripiprazole decreased ERK1/2 phosphorylation. Olanzapine also decreased IL-6 mRNA expression, while olanzapine and clozapine increased IL-10 mRNA expression. The rHypoE-19 neurons expressed the H1, 5 H T2A, and M3 receptors, while the mHypoE-46 neurons expressed the 5 H T2A, D2, and M3 receptors. Neither cell line expressed the 5 H T2C receptor. CONCLUSION: Similar to observed effects of these agents in rat neurons, induction of AMPK by aripiprazole and olanzapine suggests impaired energy sensing, while suppression of insulin-induced pAKT by clozapine and aripiprazole suggests impaired insulin signalling, seen across both rodent derived hypothalamic cell lines. Conversely, olanzapine-induced suppression of pro-inflammatory IL-6, alongside olanzapine and clozapine-induced IL-10, demonstrate anti-inflammatory effects, which do not corroborate with our prior observations in the rat neuronal line. The different findings between cell lines could be explained by differential expression of neurotransmitters receptors and/or reflect genetic heterogeneity across the rat and mouse lines. However, overall, our findings support direct effects of antipsychotics to impact insulin, energy sensing, and inflammatory pathways in hypothalamic rodent neurons.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Hipotálamo/efectos de los fármacos , Animales , Antipsicóticos/uso terapéutico , Aripiprazol/farmacología , Línea Celular , Clozapina/farmacología , Metabolismo Energético/efectos de los fármacos , Hipotálamo/metabolismo , Inflamación/metabolismo , Insulina/metabolismo , Ratones , Neuronas/metabolismo , Olanzapina/farmacología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
Psychoneuroendocrinology ; 104: 42-48, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30802709

RESUMEN

INTRODUCTION: Second generation antipsychotic (AP)s remain the gold-standard treatment for schizophrenia and are widely used on- and off-label for other psychiatric illnesses. However, these agents cause serious metabolic side-effects. The hypothalamus is the primary brain region responsible for whole body energy regulation, and disruptions in energy sensing (e.g. insulin signaling) and inflammation in this brain region have been implicated in the development of insulin resistance and obesity. To elucidate mechanisms by which APs may be causing metabolic dysregulation, we explored whether these agents can directly impact energy sensing and inflammation in hypothalamic neurons. METHODS: The rat hypothalamic neuronal cell line, rHypoE-19, was treated with olanzapine (0.25-100 uM), clozapine (2.5-100 uM) or aripiprazole (5-20 uM). Western blots measured the energy sensing protein AMPK, components of the insulin signaling pathway (AKT, GSK3ß), and components of the MAPK pathway (ERK1/2, JNK, p38). Quantitative real-time PCR was performed to determine changes in the mRNA expression of interleukin (IL)-6, IL-10 and brain derived neurotrophic factor (BDNF). RESULTS: Olanzapine (100 uM) and clozapine (100, 20 uM) significantly increased pERK1/2 and pJNK protein expression, while aripiprazole (20 uM) only increased pJNK. Clozapine (100 uM) and aripiprazole (5 and 20 uM) significantly increased AMPK phosphorylation (an orexigenic energy sensor), and inhibited insulin-induced phosphorylation of AKT. Olanzapine (100 uM) treatment caused a significant increase in IL-6 while aripiprazole (20 uM) significantly decreased IL-10. Olanzapine (100 uM) and aripiprazole (20 uM) increased BDNF expression. CONCLUSIONS: We demonstrate that antipsychotics can directly regulate insulin, energy sensing, and inflammatory pathways in hypothalamic neurons. Increased MAPK activation by all antipsychotics, alongside olanzapine-associated increases in IL-6, and aripiprazole-associated decreases in IL-10, suggests induction of pro-inflammatory pathways. Clozapine and aripiprazole inhibition of insulin-stimulated pAKT and increases in AMPK phosphorylation (an orexigenic energy sensor) suggests impaired insulin action and energy sensing. Conversely, olanzapine and aripiprazole increased BDNF, which would be expected to be metabolically beneficial. Overall, our findings suggest differential effects of antipsychotics on hypothalamic neuroinflammation and energy sensing.


Asunto(s)
Antipsicóticos/farmacología , Metabolismo Energético/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Antipsicóticos/metabolismo , Aripiprazol/metabolismo , Aripiprazol/farmacología , Línea Celular , Clozapina/metabolismo , Clozapina/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inflamación/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Olanzapina/metabolismo , Olanzapina/farmacología , Fosforilación/efectos de los fármacos , Ratas , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Int J Mol Sci ; 19(11)2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30463253

RESUMEN

To explore pathophysiology of schizophrenia, this study analyzed the regulation mechanisms that are associated with cystine/glutamate antiporter (Sxc), group-II (II-mGluR), and group-III (III-mGluR) metabotropic glutamate-receptors in thalamo-cortical glutamatergic transmission of MK801-induced model using dual-probe microdialysis. L-glutamate release in medial pre-frontal cortex (mPFC) was increased by systemic- and local mediodorsal thalamic nucleus (MDTN) administrations of MK801, but was unaffected by local administration into mPFC. Perfusion into mPFC of activators of Sxc, II-mGluR, and III-mGluR, and into the MDTN of activators of Sxc, II-mGluR, and GABAA receptor inhibited MK801-evoked L-glutamate release in mPFC. Perfusion of aripiprazole (APZ) into MDTN and mPFC also inhibited systemic MK801-evoked L-glutamate release in mPFC. Inhibition of II-mGluR in mPFC and MDTN blocked inhibitory effects of Sxc-activator and APZ on MK801-evoked L-glutamate release; however, their inhibitory effects were blocked by the inhibition of III-mGluR in mPFC but not in MDTN. These results indicate that reduced activation of the glutamate/NMDA receptor (NMDAR) in MDTN enhanced L-glutamate release in mPFC possibly through GABAergic disinhibition in MDTN. Furthermore, MDTN-mPFC glutamatergic transmission receives inhibitory regulation of Sxc/II-mGluR/III-mGluR functional complex in mPFC and Sxc/II-mGluR complex in MDTN. Established antipsychotic, APZ inhibits MK801-evoked L-glutamate release through the activation of Sxc/mGluRs functional complexes in both MDTN and mPFC.


Asunto(s)
Antiportadores/metabolismo , Aripiprazol/farmacología , Maleato de Dizocilpina/farmacología , Ácido Glutámico/metabolismo , N-Metilaspartato/antagonistas & inhibidores , Corteza Prefrontal/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Tálamo/fisiopatología , Acetilcisteína/farmacología , Animales , Aripiprazol/administración & dosificación , Maleato de Dizocilpina/administración & dosificación , Masculino , Modelos Biológicos , Perfusión , Corteza Prefrontal/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Tálamo/efectos de los fármacos
6.
BMC Complement Altern Med ; 18(1): 106, 2018 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-29566693

RESUMEN

BACKGROUND: Dependence on antipsycotic drugs like aripriprazole (ARI) is increasing at alarming rate, hence, this study was undertaken to support the hypothesis that supplementation of Citrus paradisi (Grapefruit) juice having high concentration of polyphenols might potentiate and synergize the therapeutic effect of ARI, by increasing its bioavailability and inherent antioxidant potential. These benefits together might decrease the daily dosage of the ARI and thus alleviate the possible side effects of drug. METHODS: In this study the antioxidant and anti-inflammatory potential of ARI alone and in combination with GFJ was evaluated for hydrogen peroxide (H2O2) induced oxidative stress in mice. Seventy mice (4 weeks old), were randomly divided into seven groups. Group I: Control; Group II: H2O2 treated; Group III; ARI treated; Group IV GFJ treated; Group V: GFJ and H2O2 treated; Group VI; ARI and H2O2 treated; Group VII; ARI, GFJ and H2O2 treated. Serum levels of alanine aminotransferase (ALT), blood urea nitrogen (BUN), creatinine kinase (CK), creatinine and total protein were measured. Furthermore, pro-inflammatory cytokines Interleukin (IL)-1α, IL-2, IL-10 and tumor necrosis factor-α (TNF-α) concentrations were also measured. RESULTS: The mice group that was treated with ARI, GFJ or combination of the two showed significant improvement in the H2O2 altered parameters with the combination group showing more significant improvement than the ARI and GFJ alone groups indicating a synergistic and potentiating effect of the antioxidant and anti-inflammatory potential of GFJ on ARI. CONCLUSION: Supplementing GFJ to ARI might increase an anti-oxidative potential of ARI due to inherent antioxidant and anti-inflammatory activity of GFJ and thus could alleviate the possible dosage dependent side effects of ARI.


Asunto(s)
Antiinflamatorios , Antioxidantes , Aripiprazol , Citrus paradisi/química , Jugos de Frutas y Vegetales , Peróxido de Hidrógeno/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Aripiprazol/química , Aripiprazol/farmacología , Citocinas/análisis , Citocinas/metabolismo , Sinergismo Farmacológico , Jugos de Frutas y Vegetales/análisis , Masculino , Ratones
8.
J Psychopharmacol ; 31(12): 1605-1614, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29069975

RESUMEN

Pre-clinical and clinical evidence suggests that the antidepressant efficacy of the selective serotonin reuptake inhibitor escitalopram can be enhanced by the dopamine and serotonin partial agonist aripiprazole. Given the range of possible neurochemical interactions between these drugs, the current study investigated whether aripiprazole alters the hedonic and psychomotor effects of escitalopram. Male Sprague Dawley rats ( n=116) received 10 mg/kg/day escitalopram (subcutaneous), 2 mg/kg/day aripiprazole (subcutaneous), or combined aripiprazole + escitalopram, and were tested for consumption of incentive nutritional stimuli (high-fructose corn syrup and chow), stereotypy and locomotor activity. At the conclusion of behavioral testing, mRNAs of two genes involved in reward processes were quantified: hypothalamic pro-opiomelanocortin and hippocampal brain-derived neurotrophic factor. Escitalopram produced a selective, but temporary, decrease in high fructose corn syrup consumption that was not altered by aripiprazole co-administration. Escitalopram had no significant effect on locomotion, but aripiprazole co-administration produced a persistent increase in stereotypy. Both brain-derived neurotrophic factor and pro-opiomelanocortin mRNA levels were lower in the aripiprazole + escitalopram group relative to the escitalopram group. Taken together, these results suggest that aripiprazole may enhance the antidepressant efficacy of escitalopram through improvement of psychomotor functions.


Asunto(s)
Aripiprazol/farmacología , Citalopram/farmacología , Desempeño Psicomotor/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Sinergismo Farmacológico , Ingestión de Alimentos/efectos de los fármacos , Hipocampo/metabolismo , Hipotálamo/metabolismo , Locomoción/efectos de los fármacos , Masculino , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratas , Conducta Estereotipada/efectos de los fármacos
9.
Neurorehabil Neural Repair ; 31(1): 25-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27225976

RESUMEN

BACKGROUND: Antipsychotic drugs (APDs) are used to manage traumatic brain injury (TBI)-induced behavioral disturbances, such as agitation and aggression. However, APDs exhibiting D2 receptor antagonism impede cognitive recovery after experimental TBI. Hence, empirical evaluation of APDs with different mechanistic actions is warranted. Aripiprazole (ARIP) is a D2 and 5-hydroxytryptamine1A (5-HT1A) receptor agonist; pharmacotherapies with these properties enhance cognition after TBI. OBJECTIVE: To test the hypothesis that ARIP would increase behavioral performance and decrease histopathology after TBI. METHODS: Adult male rats were subjected to either a controlled cortical impact (CCI) or sham injury and then randomly assigned to ARIP (0.1 or 1.0 mg/kg) or VEH (1.0 mL/kg, saline vehicle) groups. Treatments began 24 hours after surgery and were administered once daily for 19 days. Motor (beam-balance/beam-walk) and cognitive (Morris water maze) performance was assessed on postoperative days 1 to 5 and 14 to 19, respectively, followed by quantification of hippocampal CA1,3 neuron survival and cortical lesion volume. RESULTS: Beam-balance was significantly improved in the CCI + ARIP (1.0 mg/kg) group versus CCI + ARIP (0.1 mg/kg) and CCI + VEH (P < .05). Spatial learning and memory retention were significantly improved in the CCI + ARIP (0.1 mg/kg) group versus the CCI + ARIP (1.0 mg/kg) and CCI + VEH groups (P < .05). Both doses of ARIP reduced lesion size and CA3 cell loss versus VEH (P < .05). Importantly, neither dose of ARIP impeded functional recovery as previously reported with other APDs. CONCLUSION: These findings support the hypothesis and endorse ARIP as a safer APD for alleviating behavioral disturbances after TBI.


Asunto(s)
Antipsicóticos/farmacología , Aripiprazol/farmacología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/psicología , Animales , Antipsicóticos/efectos adversos , Aripiprazol/efectos adversos , Lesiones Traumáticas del Encéfalo/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
11.
Asian J Psychiatr ; 22: 74-5, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27520898

RESUMEN

The 5,10-methylenetetrahydrofolate reductase (MTHFR) gene plays a central role in folate metabolism. Many studies have demonstrated an association between MTHFR C677 T variant with depression, schizophrenia and bipolar disorder as one of them being comorbid to other. This has justified the use of folate supplement in psychiatric disorders mainly depression but still not in various other comorbid complex psychiatric disorders. Here we have tried to show how the l-methylfolate in conjunction with the conventional psychotropic drugs can be useful in a state of such complex psychiatric phenomenon and comorbid diagnosis with genetic polymorphism of MTHFR C677 T mutation.


Asunto(s)
Antidepresivos/farmacología , Antipsicóticos/farmacología , Trastorno Bipolar/tratamiento farmacológico , Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Trastorno Obsesivo Compulsivo/tratamiento farmacológico , Tetrahidrofolatos/farmacología , Complejo Vitamínico B/farmacología , Adulto , Antidepresivos/administración & dosificación , Antipsicóticos/administración & dosificación , Aripiprazol/administración & dosificación , Aripiprazol/farmacología , Quimioterapia Combinada , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Humanos , Compuestos de Litio/administración & dosificación , Compuestos de Litio/farmacología , Masculino , Polimorfismo Genético , Tetrahidrofolatos/administración & dosificación , Complejo Vitamínico B/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA