Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Psychoneuroendocrinology ; 163: 106987, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38340539

RESUMEN

Olanzapine is a second-generation antipsychotic that disrupts metabolism and is associated with an increased risk of type 2 diabetes. The hypothalamus is a key region in the control of whole-body metabolic homeostasis. The objective of the current study was to determine how acute peripheral olanzapine administration affects transcription and serine/threonine kinase activity in the hypothalamus. Hypothalamus samples from rats were collected following the pancreatic euglycemic clamp, thereby allowing us to study endpoints under steady state conditions for plasma glucose and insulin. Olanzapine stimulated pathways associated with inflammation, but diminished pathways associated with the capacity to combat endoplasmic reticulum stress and G protein-coupled receptor activity. These pathways represent potential targets to reduce the incidence of type 2 diabetes in patients taking antipsychotics.


Asunto(s)
Antipsicóticos , Diabetes Mellitus Tipo 2 , Humanos , Ratas , Animales , Olanzapina/farmacología , Olanzapina/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Benzodiazepinas/farmacología , Benzodiazepinas/metabolismo , Antipsicóticos/farmacología , Antipsicóticos/metabolismo , Hipotálamo/metabolismo , Perfilación de la Expresión Génica
2.
Mol Psychiatry ; 27(11): 4741-4753, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36241692

RESUMEN

Hypothalamic detection of elevated circulating glucose triggers suppression of endogenous glucose production (EGP) to maintain glucose homeostasis. Antipsychotics alleviate symptoms associated with schizophrenia but also increase the risk for impaired glucose metabolism. In the current study, we examined whether two acutely administered antipsychotics from different drug classes, haloperidol (first generation antipsychotic) and olanzapine (second generation antipsychotic), affect the ability of intracerebroventricular (ICV) glucose infusion approximating postprandial levels to suppress EGP. The experimental protocol consisted of a pancreatic euglycemic clamp, followed by kinomic and RNA-seq analyses of hypothalamic samples to determine changes in serine/threonine kinase activity and gene expression, respectively. Both antipsychotics inhibited ICV glucose-mediated increases in glucose infusion rate during the clamp, a measure of whole-body glucose metabolism. Similarly, olanzapine and haloperidol blocked central glucose-induced suppression of EGP. ICV glucose stimulated the vascular endothelial growth factor (VEGF) pathway, phosphatidylinositol 3-kinase (PI3K) pathway, and kinases capable of activating KATP channels in the hypothalamus. These effects were inhibited by both antipsychotics. In conclusion, olanzapine and haloperidol impair central glucose sensing. Although results of hypothalamic analyses in our study do not prove causality, they are novel and provide the basis for a multitude of future studies.


Asunto(s)
Antipsicóticos , Antipsicóticos/farmacología , Glucosa/metabolismo , Fosfatidilinositol 3-Quinasas , Factor A de Crecimiento Endotelial Vascular , Olanzapina/farmacología , Olanzapina/metabolismo , Benzodiazepinas/farmacología
3.
Schizophr Res ; 228: 112-117, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33434724

RESUMEN

Antipsychotic use is associated with an increased risk of type 2 diabetes. Recent work suggests antipsychotics can induce insulin resistance immediately and independently of weight gain, and that this may occur via the central nervous system (CNS). We have previously shown that the highly effective and widely prescribed antipsychotic, olanzapine inhibits CNS insulin-mediated suppression of hepatic glucose production, but the mechanisms remain unknown. The ATP-sensitive potassium (KATP) channel is a key metabolic sensor downstream of hypothalamic insulin signalling, involved in the maintenance of glucose homeostasis. Thus, the possibility arises that olanzapine inhibits central KATP channel activation to disrupt glucose metabolism. We replicate that intracerebroventricular (ICV) administration of the KATP channel activator, diazoxide, suppresses hepatic glucose production and additionally demonstrate stimulation of peripheral glucose utilization. We report that olanzapine inhibits the effects of central KATP channel activation resulting in perturbation of whole body insulin sensitivity, specifically via inhibition of glucose utilization, while leaving central KATP channel-mediated suppression of glucose production intact. Perturbation of KATP channel action in the CNS could represent a novel mechanism of antipsychotic-induced diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Adenosina Trifosfato , Glucosa , Humanos , Insulina , Olanzapina
4.
Neuropharmacology ; 163: 107633, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31077731

RESUMEN

Insulin signaling in the central nervous system is at the intersection of brain and body interactions, and represents a fundamental link between metabolic and cognitive disorders. Abnormalities in brain insulin action could underlie the development of comorbid schizophrenia and type 2 diabetes. Among its functions, central nervous system insulin is involved in regulation of striatal dopamine levels, peripheral glucose homeostasis, and feeding regulation. In this review, we discuss the role and importance of central nervous system insulin in schizophrenia and diabetes pathogenesis from a historical and mechanistic perspective. We describe central nervous system insulin sites and pathways of action, with special emphasis on glucose metabolism, cognitive functioning, inflammation, and food preferences. Finally, we suggest possible mechanisms that may explain the actions of central nervous system insulin in relation to schizophrenia and diabetes, focusing on glutamate and dopamine signaling, intracellular signal transduction pathways, and brain energetics. Understanding the interplay between central nervous system insulin and schizophrenia is essential to disentangling this comorbid relationship and may provide novel treatment approaches for both neuropsychiatric and metabolic dysfunction. This article is part of the issue entitled 'Special Issue on Antipsychotics'.


Asunto(s)
Encéfalo/fisiopatología , Sistema Nervioso Central/fisiopatología , Insulina/fisiología , Esquizofrenia/fisiopatología , Animales , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Trastornos del Conocimiento/fisiopatología , Dopamina/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Obesidad/metabolismo , Receptor de Insulina/metabolismo , Esquizofrenia/metabolismo , Transducción de Señal/fisiología
5.
Neuropharmacology ; 168: 107655, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31152767

RESUMEN

Insulin action in the central nervous system is a major regulator of energy balance and cognitive processes. The development of central insulin resistance is associated with alterations in dopaminergic reward systems and homeostatic signals affecting food intake, glucose metabolism, body weight and cognitive performance. Emerging evidence has highlighted a role for antipsychotics (APs) to modulate central insulin-mediated pathways. Although APs remain the cornerstone treatment for schizophrenia they are associated with severe metabolic complications and fail to address premorbid cognitive deficits, which characterize the disorder of schizophrenia. In this review, we first explore how the hypothesized association between schizophrenia and CNS insulin dysregulation aligns with the use of APs. We then investigate the proposed relationship between CNS insulin action and AP-mediated effects on metabolic homeostasis, and different domains of psychopathology, including cognition. We briefly discuss a potential role of CNS insulin signaling to explain the hypothesized, but somewhat controversial association between therapeutic efficacy and metabolic side effects of APs. Finally, we propose how this knowledge might inform novel treatment strategies to target difficult to treat domains of schizophrenia. This article is part of the issue entitled 'Special Issue on Antipsychotics'.


Asunto(s)
Antipsicóticos/uso terapéutico , Encéfalo/inmunología , Resistencia a la Insulina/fisiología , Insulina/inmunología , Esquizofrenia/tratamiento farmacológico , Animales , Antipsicóticos/efectos adversos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Cognición/fisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Glucosa/inmunología , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Humanos , Esquizofrenia/inmunología , Esquizofrenia/metabolismo
6.
Psychiatry Res ; 284: 112672, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31780184

RESUMEN

The Schizophrenia International Research Society (SIRS) recently held its first North American congress, which took place in Orlando, Florida from 10-14 April 2019. The overall theme of this year's congress was United in Progress - with the aim of cultivating a collaborative effort towards advancing the field of schizophrenia research. Student travel awardees provided reports of the oral sessions and concurrent symposia that took place during the congress. A collection of these reports is summarized and presented below and highlights the main themes and topics that emerged during the congress. In summary, the congress covered a broad range of topics relevant to the field of psychiatry today.


Asunto(s)
Esquizofrenia , Congresos como Asunto , Florida , Humanos , Sociedades Médicas
7.
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
8.
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
9.
Am J Physiol Endocrinol Metab ; 316(1): E1-E15, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29969315

RESUMEN

Since the serendipitous discovery of the first antipsychotic (AP) drug in the 1950s, APs remain the cornerstone of treatment for schizophrenia. A shift over the past two decades away from first-generation, conventional APs to so-called "atypical" (or 2nd/3rd generation) APs parallels acknowledgment of serious metabolic side-effects associated in particular with these newer agents. As will be reviewed, AP drugs and type 2 diabetes are now inextricably linked, contributing to the three- to fivefold increased risk of type 2 diabetes observed in schizophrenia. However, this association is not straightforward. Biological and lifestyle-related illness factors contribute to the association between type 2 diabetes and metabolic disease independently of AP treatment. In addition, APs have a well-established weight gain propensity which could also account for elevated risk of insulin resistance and type 2 diabetes. However, compelling preclinical and clinical evidence now suggests that these drugs can rapidly and directly influence pathways of glucose metabolism independently of weight gain and even in absence of psychiatric illness. Mechanisms of these direct effects remain poorly elucidated but may involve central and peripheral antagonism of neurotransmitters implicated not only in the therapeutic effects of APs but also in glucose homeostasis, possibly via effects on the autonomic nervous system. The clinical relevance of studying "direct" effects of these drugs on glucose metabolism is underscored by the widespread use of these medications, both on and off label, for a growing number of mental illnesses, extending safety concerns well beyond schizophrenia.


Asunto(s)
Antipsicóticos/uso terapéutico , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Esquizofrenia/tratamiento farmacológico , Animales , Sistema Nervioso Autónomo/metabolismo , Clozapina/uso terapéutico , Diabetes Mellitus Tipo 2/epidemiología , Técnica de Clampeo de la Glucosa , Humanos , Resistencia a la Insulina , Receptores de Dopamina D2/metabolismo , Receptores de Serotonina 5-HT2/metabolismo , Aumento de Peso
10.
Front Psychiatry ; 9: 622, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30568606

RESUMEN

Cognitive impairment is a core symptom domain of schizophrenia. The effect of antipsychotics, the cornerstone of treatment in schizophrenia, on this domain is not fully clear. There is some evidence suggesting that antipsychotics may partially improve cognitive function, and that this improvement may vary depending on the specific cognitive domain. However, this research is confounded by various factors, such as age, duration/stage of illness, medication adherence, and extrapyramidal side effects that complicate the relationship between antipsychotics and cognitive improvement. Furthermore, antipsychotics-particularly the second generation, or "atypical" antipsychotics-can induce serious metabolic side effects, such as obesity, dyslipidemia and type 2 diabetes, illnesses which themselves have been linked to impairments in cognition. Thus, the inter-relationships between cognition and metabolic side effects are complex, and this review aims to examine them in the context of schizophrenia and antipsychotic treatment. The review also speculates on potential mechanisms underlying cognitive functioning and metabolic risk in schizophrenia. We conclude that the available literature examining the inter-section of antipsychotics, cognition, and metabolic effects in schizophrenia is sparse, but suggests a relationship between metabolic comorbidity and worse cognitive function in patients with schizophrenia. Further research is required to determine if there is a causal connection between the well-recognized metabolic adverse effects of antipsychotics and cognitive deficits over the course of the illness of schizophrenia, as well as, to determine underlying mechanisms. In addition, findings from this review highlight the importance of monitoring metabolic disturbances in parallel with cognition, as well as, the importance of interventions to minimize metabolic abnormalities for both physical and cognitive health.

11.
Psychoneuroendocrinology ; 98: 127-130, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30142549

RESUMEN

The use of antipsychotics is associated with severe disruptions in whole body glucose and lipid metabolism which may in part occur through the central nervous system and impaired insulin action at the brain. Here we investigated whether olanzapine treatment might also affect the ability of central insulin treatment to regulate food intake and fuel preference in the light and dark cycle. Male Sprague-Dawley rats were treated with olanzapine (or vehicle solution; 3 mg/kg, subcutaneous) and a simultaneous acute intracerebral ventricular (ICV) infusion of insulin (or vehicle; 3 µL at 10mU; ICV) at the beginning of the 12-h light and dark cycles. Olanzapine treatment reduced RER in the dark and light phases (most consistently in the 4-hours post-treatment), while ICV insulin reduced average RER predominantly in the dark phase, but also at the end of the light cycle. The RER lowering effect of ICV-insulin during the light cycle was absent in the group co-administered olanzapine. The reduction in RER during the dark phase was mirrored by decreased food intake with ICV insulin, but not olanzapine treated rats. The reduction in food intake by ICV-insulin was abolished in rats co-administered olanzapine suggesting rapid induction of central insulin resistance. A combination of ICV-insulin and olanzapine similarly reduced RER in the dark phase, independent of changes in food intake. Olanzapine treatment, alone or in combination with ICV-insulin, significantly reduced VCO2 at regular intervals in the dark phase (specifically 3 h post-treatment), while VO2 was not significantly altered by either treatment. Finally, heat production was increased by olanzapine treatment in the light phase, though this effect was not consistent. The findings confirm that acute olanzapine treatment directly reduces RER and suggest that treatment with this drug may also override central insulin-mediated reductions in food intake at the hypothalamus (while still independently favoring fatty acid oxidation). Acute central insulin similarly reduces RER, but in contrast to olanzapine, this may represent a physiologically appropriate response to reduction in food intake.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Olanzapina/farmacología , Animales , Antipsicóticos/farmacología , Espiración/efectos de los fármacos , Glucosa/metabolismo , Hipotálamo/efectos de los fármacos , Insulina , Resistencia a la Insulina/fisiología , Masculino , Olanzapina/metabolismo , Ratas , Ratas Sprague-Dawley
12.
J Psychiatry Neurosci ; 42(6): 424-431, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29083297

RESUMEN

BACKGROUND: Insulin receptors are widely expressed in the brain and may represent a crossroad between metabolic and cognitive disorders. Although antipsychotics, such as olanzapine, are the cornerstone treatment for schizophrenia, they are associated with high rates of type 2 diabetes and lack efficacy for illness-related cognitive deficits. Historically, this risk of diabetes was attributed to the weight gain propensity of antipsychotics, but recent work suggests antipsychotics can have weight-independent diabetogenic effects involving unknown brain-mediated mechanisms. Here, we examined whether antipsychotics disrupt central insulin action, hypothesizing that olanzapine would impair the well-established ability of central insulin to supress hepatic glucose production. METHODS: Pancreatic euglycemic clamps were used to measure glucose kinetics alongside a central infusion of insulin or vehicle into the third ventricle. Male rats were pretreated with olanzapine or vehicle per our established model of acute olanzapine-induced peripheral insulin resistance. Groups included (central-peripheral) vehicle-vehicle (n = 11), insulin-vehicle (n = 10), insulin-olanzapine (n = 10) and vehicle-olanzapine (n = 8). RESULTS: There were no differences in peripheral glucose or insulin levels. Unexpectedly, we showed that central insulin increased glucose uptake, and this effect was not perturbed by olanzapine. We replicated suppression of glucose production by insulin (clamp relative to basal: 77.9% ± 13.1%, all p < 0.05), an effect abolished by olanzapine (insulin-olanzapine: 7.7% ± 14%). LIMITATIONS: This study used only male rats and an acute dose of olanzapine. CONCLUSION: To our knowledge, this is the first study suggesting olanzapine may impair central insulin sensing, elucidating a potential mechanism of antipsychotic-induced diabetes and opening avenues of investigation related to domains of schizophrenia psychopathology.


Asunto(s)
Antipsicóticos/farmacología , Benzodiazepinas/farmacología , Glucosa/metabolismo , Insulina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Catéteres de Permanencia , Infusiones Intraventriculares , Insulina/administración & dosificación , Resistencia a la Insulina/fisiología , Masculino , Olanzapina , Ratas Sprague-Dawley , Absorción Subcutánea
13.
Psychopharmacology (Berl) ; 233(14): 2629-53, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27251130

RESUMEN

RATIONALE: So-called atypical antipsychotics (AAPs) are associated with varying levels of weight gain and associated metabolic disturbances, which in patients with serious mental illness (SMI) have been linked to non-compliance and poor functional outcomes. Mechanisms underlying AAP-induced metabolic abnormalities are only partially understood. Antipsychotic-induced weight gain may occur as a result of increases in food intake and/or changes in feeding. OBJECTIVE: In this review, we examine the available human and preclinical literature addressing AAP-related changes in feeding behavior, to determine whether changes in appetite and perturbations in regulation of food intake could be contributing factors to antipsychotic-induced weight gain. RESULTS: In general, human studies point to disruption by AAPs of feeding behaviors and food consumption. In rodents, increases in cumulative food intake are mainly observed in females; however, changes in feeding microstructure or motivational aspects of food intake appear to occur independent of sex. CONCLUSIONS: The findings from this review indicate that the varying levels of AAP-related weight gain reflect changes in both appetite and feeding behaviors, which differ by type of AAP. However, inconsistencies exist among the studies (both human and rodent) that may reflect considerable differences in study design and methodology. Future studies examining underlying mechanisms of antipsychotic-induced weight gain are recommended in order to develop strategies addressing the serious metabolic side effect of AAPs.


Asunto(s)
Antipsicóticos/farmacología , Conducta Alimentaria/efectos de los fármacos , Aumento de Peso/efectos de los fármacos , Animales , Apetito/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Humanos , Ratones , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...