Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pharmacol Rep ; 76(1): 51-71, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38194217

RESUMEN

BACKGROUND: Maternal elevated glucocorticoid levels during pregnancy can affect the developing fetus, permanently altering the structure and function of its brain throughout life. Excessive action of these hormones is known to contribute to psychiatric disorders, including depression. MATERIALS: The study was performed in a rat model of depression based on prenatal administration of dexamethasone (DEX) in late pregnancy (0.1 mg/kg, days 14-21). We evaluated the effects of prenatal DEX treatment on the cognition and bioenergetic signaling pathways in the brain of adult male rats, in the frontal cortex and hippocampus, and in response to stress in adulthood, using behavioral and biochemical test batteries. RESULTS: We revealed cognitive deficits in rats prenatally treated with DEX. At the molecular level, a decrease in the orexin A and orexin B levels and downregulation of the AMPK-SIRT1-PGC1α transduction pathway in the frontal cortex of these animals were observed. In the hippocampus, a decreased expression of orexin B was found and changes in the MR/GR ratio were demonstrated. Furthermore, an increase in HDAC5 level triggered by the prenatal DEX treatment in both brain structures and a decrease in MeCP2 level in the hippocampus were reported. CONCLUSIONS: Our study demonstrated that prenatal DEX treatment is associated with cognitive dysfunction and alterations in various proteins leading to metabolic changes in the frontal cortex, while in the hippocampus adaptation mechanisms were activated. The presented results imply that different pathophysiological metabolic processes may be involved in depression development, which may be useful in the search for novel therapies.


Asunto(s)
Trastorno Depresivo , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Ratas , Masculino , Animales , Embarazo , Orexinas/metabolismo , Dexametasona/farmacología , Depresión/metabolismo , Encéfalo/metabolismo , Glucocorticoides/metabolismo , Hipocampo , Modelos Animales , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores de Glucocorticoides/metabolismo
2.
Int J Mol Sci ; 24(2)2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36674678

RESUMEN

Since depression produces a long-term negative impact on quality of life, understanding the pathophysiological changes implicated in this disorder is urgent. There is growing evidence that demonstrates a key role for dysfunctional energy metabolism in driving the onset of depression; thus, bioenergetic alterations should be extensively studied. Brain metabolism is known to be a glucocorticoid-sensitive process, but the long-lasting consequences in adulthood following high levels of glucocorticoids at the early stages of life are unclear. We examined a possible association between brain energetic changes induced by synthetic glucocorticoid-dexamethasone treatment in the prenatal period and depressive-like behavior. The results show a reduction in the oxidative phosphorylation process, Krebs cycle impairment, and a weakening of the connection between the Krebs cycle and glycolysis in the frontal cortex of animals receiving dexamethasone, which leads to ATP reduction. These changes appear to be mainly due to decreased expression of pyruvate dehydrogenase, impairment of lactate transport to neurons, and pyruvate to the mitochondria. Acute stress in adulthood only slightly modified the observed alterations in the frontal cortex, while in the case of the hippocampus, prenatal exposure to dexamethasone made this structure more sensitive to future adverse factors.


Asunto(s)
Glucocorticoides , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Embarazo , Humanos , Glucocorticoides/metabolismo , Dexametasona/efectos adversos , Dexametasona/metabolismo , Depresión/metabolismo , Calidad de Vida , Encéfalo/metabolismo , Homeostasis , Piruvatos/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo
4.
Pharmacol Rep ; 74(5): 859-870, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35951260

RESUMEN

Metabolic disturbances in the brain are assumed to be early changes involved in the pathogenesis of depression, and these alterations may be intensified by a deficiency of thyroid hormones. In contrast to glucose metabolism, the link between altered brain lipids and the pathogenesis of depression is poorly understood, therefore in the present study, we determine transcription factors and enzymes regulating cholesterol and fatty acid biosynthesis in the brain structures in an animal model of depression, hypothyroidism and the coexistence of these diseases.In used model of depression, a decrease in the active form of the transcription factor SREBP-2 in the hippocampus was demonstrated, thus suggesting a reduction in cholesterol biosynthesis. In turn, in the hypothyroidism model, the reduction of cholesterol biosynthesis in the frontal cortex was demonstrated by both the reduction of mature SREBP-2 and the concentration of enzymes involved in cholesterol biosynthesis. The lower expression of LDL receptors in the frontal cortex indicates the restriction of cholesterol uptake into the cells in the model of coexistence of depression and hypothyroidism. Moreover, the identified changes in the levels of SNAP-25, GLP-1R and GLP-2R pointed to disturbances in synaptic plasticity and neuroprotection mechanisms in the examined brain structures.In conclusion, a reduction in cholesterol synthesis in the hippocampus in the model of depression may be the reason for the reduction of synaptic plasticity, whereas a lower level of LDL-R occurring in the frontal cortex in rats from the model of depression and hypothyroidism coexistence could be the reason of anxiogenic and depression-like behaviors.


Asunto(s)
Hipotiroidismo , Metabolismo de los Lípidos , Animales , Ratas , Depresión/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Hipotiroidismo/metabolismo , Hormonas Tiroideas , Colesterol/metabolismo , Encéfalo/metabolismo , Ácidos Grasos , Glucosa/metabolismo , Modelos Animales , Receptores de LDL/metabolismo
5.
Cells ; 10(11)2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34831160

RESUMEN

The developing and adult brain is a target organ for the vast majority of hormones produced by the body, which are able to cross the blood-brain barrier and bind to their specific receptors on neurons and glial cells. Hormones ensure proper communication between the brain and the body by activating adaptive mechanisms necessary to withstand and react to changes in internal and external conditions by regulating neuronal and synaptic plasticity, neurogenesis and metabolic activity of the brain. The influence of hormones on energy metabolism and mitochondrial function in the brain has gained much attention since mitochondrial dysfunctions are observed in many different pathological conditions of the central nervous system. Moreover, excess or deficiency of hormones is associated with cell damage and loss of function in mitochondria. This review aims to expound on the impact of hormones (GLP-1, insulin, thyroid hormones, glucocorticoids) on metabolic processes in the brain with special emphasis on oxidative phosphorylation dysregulation, which may contribute to the formation of pathological changes. Since the brain concentrations of sex hormones and neurosteroids decrease with age as well as in neurodegenerative diseases, in parallel with the occurrence of mitochondrial dysfunction and the weakening of cognitive functions, their beneficial effects on oxidative phosphorylation and expression of antioxidant enzymes are also discussed.


Asunto(s)
Encefalopatías/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Hormonas/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
6.
Cells ; 10(6)2021 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-34198731

RESUMEN

The clinical effectiveness of supportive therapy with thyroid hormones in drug-resistant depression is well-known; however, the mechanisms of action of these hormones in the adult brain have not been fully elucidated to date. We determined the effects of venlafaxine and/or L-thyroxine on metabolic parameters and markers involved in the regulation of synaptic plasticity and cell damage in an animal model of coexisting depression and hypothyroidism, namely, Wistar Kyoto rats treated with propylthiouracil. In this model, in relation to the depression model itself, the glycolysis process in the brain was weakened, and a reduction in pyruvate dehydrogenase in the frontal cortex was normalized only by the combined treatment with L-thyroxine and venlafaxine, whereas changes in pyruvate and lactate levels were affected by all applied therapies. None of the drugs improved the decrease in the expression of mitochondrial respiratory chain enzymes. No intensification of glucocorticoid action was shown, while an unfavorable change caused by the lack of thyroid hormones was an increase in the caspase-1 level, which was not reversed by venlafaxine alone. The results indicated that the combined administration of drugs was more effective in normalizing glycolysis and the transition to the Krebs cycle than the use of venlafaxine or L-thyroxine alone.


Asunto(s)
Depresión , Hipotiroidismo , Plasticidad Neuronal/efectos de los fármacos , Tiroxina/farmacología , Clorhidrato de Venlafaxina/farmacología , Animales , Depresión/complicaciones , Depresión/tratamiento farmacológico , Depresión/metabolismo , Depresión/fisiopatología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Lóbulo Frontal/metabolismo , Lóbulo Frontal/fisiopatología , Humanos , Hipotiroidismo/complicaciones , Hipotiroidismo/tratamiento farmacológico , Hipotiroidismo/metabolismo , Hipotiroidismo/fisiopatología , Masculino , Ratas , Ratas Endogámicas WKY
7.
Pharmacol Rep ; 73(4): 1020-1032, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34003475

RESUMEN

Depression is a highly prevalent mood disorder and one of the major health concerns in modern society. Moreover, it is characterized by a high prevalence of coexistence with many other diseases including metabolic disorders such as type 2 diabetes mellitus (T2DM) and obesity. Currently used antidepressant drugs, which mostly target brain monoaminergic neurotransmission, have limited clinical efficacy. Although the etiology of depression has not been fully elucidated, current scientific data emphasize the role of neurotrophic factors deficiencies, disturbed homeostasis between the nervous system and the immune and endocrine systems, as well as disturbances in brain energy metabolism and dysfunctions in the gut-brain axis as important factors in the pathogenesis of this neuropsychiatric disorder. Therefore, therapeutic options that could work in a way other than classic antidepressants are being sought to increase the effectiveness of the treatment. Interestingly, glucagon-like peptide-1 receptor agonists (GLP-1RAs), used in the treatment of T2DM and obesity, are known to show pro-cognitive and neuroprotective properties, and exert modulatory effects on immune, endocrine and metabolic processes in the central nervous system. This review article discusses the potential antidepressant effects of GLP-1RAs, especially in the context of their action on the processes related to neuroprotection, inflammation, stress response, energy metabolism, gut-brain crosstalk and the stability of the gut microbiota.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Animales , Depresión/metabolismo , Metabolismo Energético/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Neuroprotección/efectos de los fármacos
8.
Mitochondrion ; 58: 169-178, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33766747

RESUMEN

Depression is an affective disease with a complex clinical picture that is characterized by mood and emotional disturbances. It is known that several factors contribute to the risk of developing depression. The concept that mitochondrial dysfunction is one of the causes of depression is supported by a wide range of studies on cell cultures, animal models, and clinical research. An understanding the relationship between mitochondrial processes and central nervous system abnormalities that occur in the course of depression can guide the development of novel mitochondrial targeted therapeutic strategies as well as the usage of currently available antidepressants in a new context. This brief review aims to summarize recent findings on mitochondria dysfunction in depression, provide insight into therapeutic strategies targeting mitochondrial pathways, allude to future promising therapies, and discuss factors that can be used to improve treatment outcomes. The main focus is on new aspects (the effects of nutraceuticals and physical activity on brain metabolism), which can be combined with the available treatment options [monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), selective serotonin reuptake inhibitors (SSRIs) and atypical drugs] to enhance their therapeutic effects.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Animales , Humanos
9.
Int J Mol Sci ; 22(4)2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562494

RESUMEN

The role that thyroid hormone deficiency plays in depression and synaptic plasticity in adults has only begun to be elucidated. This paper analyzes the possible link between depression and hypothyroidism in cognitive function alterations, using Wistar-Kyoto (WKY-an animal model of depression) rats and control Wistar rats under standard and thyroid hormone deficiency conditions (propylthiouracil administration-PTU). A weakening of memory processes in the WKY rats is shown behaviorally, and in the reduction of long-term potentiation (LTP) in the dentate gyrus (DG) and CA1 hippocampal regions. PTU administration decreased LTP and increased basal excitatory transmission in the DG in Wistar rats. A decrease in short-term synaptic plasticity is shown by the paired-pulse ratio measurement, occurring during hypothyroidism in DG and CA1 in WKY rats. Differences between the strains may result from decreases in the p-CaMKII, p-AKT, and the level of acetylcholine, while in the case of the co-occurrence of depression and hypothyroidism, an increase in the p-ERK1-MAP seemed to be important. Obtained results show that thyroid hormones are less involved in the inhibition of glutamate release and/or excitability of the postsynaptic neurons in WKY rats, which may indicate a lower sensitivity of the hippocampus to the action of thyroid hormones in depression.


Asunto(s)
Disfunción Cognitiva/etiología , Depresión/etiología , Hipocampo/fisiopatología , Hipotiroidismo/complicaciones , Animales , Región CA1 Hipocampal/fisiopatología , Disfunción Cognitiva/fisiopatología , Giro Dentado/fisiopatología , Depresión/fisiopatología , Depresión/psicología , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Humanos , Hipotiroidismo/fisiopatología , Hipotiroidismo/psicología , Potenciación a Largo Plazo/fisiología , Masculino , Memoria/fisiología , Plasticidad Neuronal/fisiología , Propiltiouracilo/toxicidad , Ratas , Ratas Endogámicas WKY , Ratas Wistar , Hormonas Tiroideas/deficiencia , Hormonas Tiroideas/fisiología
10.
Front Neurosci ; 14: 586939, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343282

RESUMEN

Although hypothyroidism appears to be an important factor in the pathogenesis of depression, the impact of thyroid hormones on the bioenergetics of the adult brain is still poorly known. Since metabolic changes are reported to be a key player in the manifestation of depressive disorder, we investigated whether there are differences in selected metabolic markers in the frontal cortex and hippocampus of Wistar Kyoto rats (WKY; an animal model of depression) compared to those of control Wistar rats and whether the induction of hypothyroidism by propylthiouracil (PTU) elicits similar effects in these animals or intensifies some parameters in the WKY rats. In our study, we used WKY rats as a model of depression since this strain exhibits lower levels of monoamines in the brain than control rats and exhibits behavioral and hormonal alterations resembling those of depression, including increased reactivity to stress. The findings indicate a decrease in glycolysis intensity in both brain structures in the WKY rats as well as in both strains under hypothyroidism conditions. Furthermore, hypothyroidism disrupted the connection between glycolysis and the Krebs cycle in the frontal cortex and hippocampus in the depression model used in this study. Decreased thyroid hormone action was also shown to attenuate oxidative phosphorylation, and this change was greater in the WKY rats. Our results suggest that both the depression and hypothyroidism models are characterized by similar impairments in brain energy metabolism and mitochondrial function and, additionally, that the co-occurrence of hypothyroidism and depression may exacerbate some of the metabolic changes observed in depression.

11.
Neurotox Res ; 37(2): 406-424, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31782099

RESUMEN

Current data suggest an important role of brain metabolic disturbances in the pathogenesis of depression and obesity, diseases that frequently co-occur. Our aim was to determine whether there are changes in markers characterizing glucose metabolism in prenatal stress (PS; animal model of depression), in rats fed a high-fat diet (HFD), and especially in the model of depression and obesity co-occurrence. The changes in glucose-6-phosphate, glycogen, glucose transporters (GLUT1, GLUT4), glucagon-like peptide-1 receptor (GLP-1R), and mitochondrial complexes levels in the frontal cortex and/or hippocampus were observed. In the case of the coexistence of depression and obesity, the most important changes were (1) the decrease in the membrane form of GLUT4, which may suggest weaker insulin action in the frontal cortex, and (2) the diminished GLP-1R, which could cause neurodegenerative changes in the hippocampus. However, presented results suggested that HFD weakened the PS effect of uncoupling oxidative phosphorylation in the frontal cortex.


Asunto(s)
Encéfalo/metabolismo , Depresión/metabolismo , Obesidad/metabolismo , Fenotipo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Depresión/psicología , Dieta Alta en Grasa/efectos adversos , Femenino , Glucosa/metabolismo , Masculino , Obesidad/psicología , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Ratas , Ratas Sprague-Dawley
12.
Neurotox Res ; 36(2): 357-375, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31197747

RESUMEN

Gestational diabetes is a disorder associated with abnormal chronic inflammation that poses a risk to the developing fetus. We investigated the effects of experimentally induced diabetes (streptozotocin model) in Wistar female rats on the inflammatory status of the hippocampi of their offspring. Additionally, the impact of antidiabetic drugs (metformin and glyburide) on inflammatory processes was evaluated. Organotypic hippocampal cultures (OHCs) were prepared from the brains of the 7-day-old rat offspring of control and diabetic mother rats. On the 7th day in vitro, the cultures were pretreated with metformin (3 µM) or glyburide (1 µM) and then stimulated for 24 h with lipopolysaccharide (LPS, 1 µg/ml). The OHCs obtained from the offspring of diabetic mothers were characterized by the increased mortality of cells and an enhanced susceptibility to damage caused by LPS. Although we showed that LPS stimulated the secretion of pro-inflammatory cytokines (IL-1ß, IL-6, TNF-α) in the control and diabetic cultures, the levels of IL-1ß and IL-6 in the OHC medium obtained from the offspring of diabetic mothers were more pronounced. In the diabetic cultures, enhanced levels of TLR-4 and the overactivation of the NLRP3 inflammasome were demonstrated. Metformin and glyburide pretreatment normalized the LPS-induced IL-1ß secretion in the control and diabetic cultures. Furthermore, glyburide diminished both: LPS-induced IL-6 and TNF-α secretion in the control and diabetic cultures and increased NF-κB p65 subunit phosphorylation. Glyburide also diminished the levels of the NLRP3 subunit and caspase-1, but only in the diabetic cultures. The results showed that maternal diabetes affected inflammatory processes in the offspring brain and increased hippocampal sensitivity to the LPS-induced inflammatory response. The use of antidiabetic agents, especially glyburide, had a beneficial impact on the changes caused by maternal diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Gestacional/metabolismo , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Diabetes Mellitus Experimental/patología , Diabetes Gestacional/inducido químicamente , Diabetes Gestacional/patología , Femenino , Hipocampo/patología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Técnicas de Cultivo de Órganos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar
13.
Int J Mol Sci ; 19(7)2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29976873

RESUMEN

An increasing body of evidence postulates that microglia are the main mediators of inflammation-related disorders, including depression. Since activated microglia produce a wide range of pro- and anti-inflammatory factors, the modulation of M1/M2 microglial polarization by antidepressants may be crucial in the treatment of depression. The current paper aimed to investigate the impact of tianeptine on the microglia's viability/death parameters, and on M1/M2 microglial activation in response to lipopolysaccharide (LPS) stimulation. Furthermore, the molecular mechanisms via which tianeptine affected the LPS-evoked changes were investigated. The results revealed that tianeptine had partially protective effects on the changes in microglia viability/death evoked by LPS. Tianeptine attenuated microglia activation by decreasing the expression of cluster of differentiation 40 (CD40), and major histocompatibility complex class II (MHC II) markers, as well as the release of pro-inflammatory factors: interleukin (IL)-1ß, IL-18, IL-6, tumor necrosis factor alpha (TNF-α), and chemokine CC motif ligand 2 (CCL2), and the production of nitric oxide and reactive oxygen species. In contrast, we did not observe an impact of tianeptine on M2 microglia measured by IL-4, IL-10, TGF-ß, and insulin-like growth factor 1 (IGF-1) expression. Moreover, we demonstrated an inhibitory effect of tianeptine on the LPS-induced activation of the nucleotide-binding oligomerization domain-like (NOD-like) receptor pyrin-containing 3 inflammasome (NLRP3) inflammasome subunits, NLRP3 and caspase-1, as well as the ability of tianeptine to reduce Toll-like receptor 4 (TLR4) levels, as well as the phosphorylation of extracellular signal-related kinases 1 and 2 (ERK1/2) and of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Collectively, we demonstrated that tianeptine has protective properties and inhibits M1 polarization, thus attenuating the production of inflammatory mediators. Moreover, we found that M1 microglia suppression may be related to the NLRP3 inflammasome and TLR4 signaling. These findings suggest that a better understanding of the multifaceted mechanisms of tianeptine action on microglia may increase the effectiveness of therapy, where inflammation is a central hallmark.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Inflamasomas/metabolismo , Microglía/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Tiazepinas/farmacología , Animales , Muerte Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Lipopolisacáridos/farmacología , Óxido Nítrico/metabolismo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 4/metabolismo
14.
J Neuroendocrinol ; 2018 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-29524264

RESUMEN

Obesity is a disease that often co-occurs with depression, and some evidence indicates that chronic stress in the perinatal period, in association with overactive glucocorticoids, can cause permanent changes that increase the risk of the development of both depression and obesity later in life. However, the mechanism responsible for the overly potent action of glucocorticoids in both depression and obesity is not known. The aim of the present study was to determine the expression of glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs) and the factors that affect GR function (FKBP51, Bag-1 and HSP70) in a prenatal stress animal model of depression, a model of obesity and a model of both depression and obesity. Prenatal stress but not high-fat diet (HFD) was found to decrease the GR concentration in the frontal cortex. The level of the Bag-1M (46 kDa) isoform was also decreased in this structure but only in prenatal-stressed animals that did not show depression-like behaviour in the Porsolt test and were fed the standard diet (STD). In the model of depression employed here, decreases in MR expression and GR co-chaperone (FKBP51) levels in the hippocampus were also observed, and HFD intensified the prenatal stress-induced changes in MR expression. The obtained results indicated that prenatal stress affected the expression of GRs, MRs and their co-chaperones in the brain, but its effects were different in the frontal cortex and hippocampus. The decrease in MR density in the hippocampus and increased plasma insulin level seemed to be the most significant changes observed in the model of the co-occurrence of depression and obesity, which could limit the neuroprotective effects associated with the activation of MR and be a marker of peripheral insulin resistance, respectively. This article is protected by copyright. All rights reserved.

15.
Pharmacol Rep ; 70(2): 322-330, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29477041

RESUMEN

BACKGROUND: Alteration in the brain mitochondrial functions have been suggested to participate, as a relevant factor, in the development of mental disorders. Therefore, the brain mitochondria may be a crucial therapeutic target in the course of depression. METHODS: Our goal was to find out the impact of two antidepressant drugs with various mechanisms of action - imipramine and fluoxetine, on the frontal cortex mitochondria-enriched fraction in an animal model of depression based on the prenatal stress procedure. RESULTS: Our results confirmed that the prenatal stress caused depressive-like disturbances in the adult offspring rats, which were normalized by the chronic imipramine and fluoxetine administration. For the first time, using 2D-LC-MS/MS, we demonstrated nine differentially expressed proteins after the imipramine administration. Of these proteins, the up-regulation of the 2',3'-cyclic-nucleotide 3'-phosphodiesterase enzyme and down-regulation of the Hypoxanthine-guanine phosphoribosyltransferase (HPRT), Ras-related proteins (Rap-1A and Rap-1B) and Transgelin-3 (NP25) were the most striking. In contrast, after the chronic fluoxetine treatment, we observed differential expression in five proteins, including the enhanced expression of component of pyruvate dehydrogenase complex and diminished of Glutathione S-transferase P (Gstp-1), as well as Maleylacetoacetate isomerase. CONCLUSIONS: These results overcome the interesting data that brain mitochondria in the frontal cortex may constitute the target for pharmacotherapy. The multifaceted profile of both antidepressant drugs action makes difficult to elucidate the exact mechanism of imipramine and fluoxetine action in the brain mitochondria. Further study of mitochondrial dysfunction in psychiatric disorders will be base to know the possible biological consequences of our observations.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Trastorno Depresivo/tratamiento farmacológico , Lóbulo Frontal/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteoma/efectos de los fármacos , Proteoma/metabolismo , Animales , Depresión/metabolismo , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Femenino , Fluoxetina/farmacología , Lóbulo Frontal/metabolismo , Imipramina/farmacología , Masculino , Mitocondrias/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteómica/métodos , Ratas , Ratas Sprague-Dawley
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 80(Pt C): 279-290, 2018 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-28433460

RESUMEN

The effect of antidepressant drugs on tumor progress is very poorly recognized. The aim of the present study was to examine the effect of individual reactivity to stress and 24-day desipramine (DES) administration on the metastatic colonization of adenocarcinoma MADB 106 cells in the lungs of Wistar rats. Wistar rats were subjected to stress procedure according to the chronic mild stress (CMS) model of depression for two weeks and stress highly-sensitive (SHS) and stress non-reactive (SNR) rats were selected. SHS rats were more prone to cancer metastasis than SNR ones and chronic DES treatment further increased the number of lung metastases by 59% and 50% in comparison to vehicle-treated appropriate control rats. The increase in lung metastases was connected with DES-induced skew macrophage activity towards M2 functional phenotype in SHS and SNR rats. Moreover, during 24h after DES injection in healthy rats, the decreased number of TCD8+ and B cells in SHS and SNR rats as well as NK cell cytotoxic activity in SNR rats could be attributed to the lowered capacity to defend against cancer metastasis observed in chronic DES treated and tumor injected rats.


Asunto(s)
Adenocarcinoma/complicaciones , Adenocarcinoma/secundario , Desipramina/farmacología , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/secundario , Estrés Psicológico/complicaciones , Animales , Antidepresivos/farmacología , Línea Celular Tumoral , Subgrupos Linfocitarios/efectos de los fármacos , Masculino , Ratas , Ratas Endogámicas
17.
Front Pharmacol ; 8: 779, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163165

RESUMEN

An increasing number of studies indicate that the chemokine system may be the third major communication system of the brain. Therefore, the role of the chemokine system in the development of brain disorders, including depression, has been recently proposed. However, little is known about the impact of the administration of various antidepressant drugs on the brain chemokine - chemokine receptor axis. In the present study, we used an animal model of depression based on the prenatal stress procedure. We determined whether chronic treatment with tianeptine, venlafaxine, or fluoxetine influenced the evoked by prenatal stress procedure changes in the mRNA and protein levels of the homeostatic chemokines, CXCL12 (SDF-1α), CX3CL1 (fractalkine) and their receptors, in the hippocampus and frontal cortex. Moreover, the impact of mentioned antidepressants on the TGF-ß, a molecular pathway related to fractalkine receptor (CX3CR1), was explored. We found that prenatal stress caused anxiety and depressive-like disturbances in adult offspring rats, which were normalized by chronic antidepressant treatment. Furthermore, we showed the stress-evoked CXCL12 upregulation while CXCR4 downregulation in hippocampus and frontal cortex. CXCR7 expression was enhanced in frontal cortex but not hippocampus. Furthermore, the levels of CX3CL1 and CX3CR1 were diminished by prenatal stress in the both examined brain areas. The mentioned changes were normalized with various potency by chronic administration of tested antidepressants. All drugs in hippocampus, while tianeptine and venlafaxine in frontal cortex normalized the CXCL12 level in prenatally stressed offspring. Moreover, in hippocampus only fluoxetine enhanced CXCR4 level, while fluoxetine and tianeptine diminished CXCR7 level in frontal cortex. Additionally, the diminished by prenatal stress levels of CX3CL1 and CX3CR1 in the both examined brain areas were normalized by chronic tianeptine and partially fluoxetine administration. Tianeptine modulate also brain TGF-ß signaling in the prenatal stress-induced animal model of depression. Our results provide new evidence that not only prenatal stress-induced behavioral disturbances but also changes of CXCL12 and their receptor and at less extend in CX3CL1-CX3CR1 expression may be normalized by chronic antidepressant drug treatment. In particular, the effect on the CXCL12 and their CXCR4 and CXCR7 receptors requires additional studies to elucidate the possible biological consequences.

18.
Int Immunopharmacol ; 48: 35-42, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28460354

RESUMEN

Some antidepressants show a significantly lower efficacy in elderly patients, particularly in women. Previous studies have shown that antidepressants administered to young animals reduced depression-like symptoms induced by lipopolysaccharide (LPS). The aim of this study was to find out whether the antidepressant and anti-inflammatory properties of fluoxetine (FLU) can be observed also in old female C57BL/6J mice. A depression-like state was evoked by the administration of LPS (100µg/kg for 4 consecutive days) which was followed by reduction of sucrose preference (anhedonia) and enhancement of immobility-time in the forced swim test (FST). Animals, which received FLU (10mg/kg, 11days) exhibited a decreased LPS-induced expression of some inflammatory cytokines in the hippocampus and spleen but this effect was not accompanied by beneficial changes in animals' behavior. Despite the lack of antidepressant-properties of FLU in this model, our studies have proven significant profound anti-inflammatory properties of chronic FLU treatment which may suggest its suitability for fending off inflammatory processes in the elderly.


Asunto(s)
Antiinflamatorios/farmacología , Antidepresivos/farmacología , Citocinas/inmunología , Depresión/inmunología , Fluoxetina/farmacología , Envejecimiento/inmunología , Animales , Antidepresivos/uso terapéutico , Conducta Animal/efectos de los fármacos , Citocinas/genética , Depresión/tratamiento farmacológico , Femenino , Fluoxetina/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Lipopolisacáridos , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología
19.
Artículo en Inglés | MEDLINE | ID: mdl-28526399

RESUMEN

Several lines of evidence indicate that adverse experience in early life may be a triggering factor for disturbances in the brain mitochondrial proteins and lead to the development of depression in adulthood. On the other hand, little is known about the impact of chronic administration of various antidepressant drugs on the brain mitochondria, as a target for the pharmacotherapy of depression. The purpose of our study was to compare the impact of chronic treatment with two antidepressant drugs with different mechanisms of action, a tricyclic antidepressant (TCA), imipramine, and an antidepressant of the selective serotonin reuptake inhibitor (SSRI) class, fluoxetine, on the mitochondria-enriched subproteome profile in the hippocampus of 3-month-old male rats following a prenatal stress procedure (an animal model of depression). We clearly confirmed that chronic imipramine and fluoxetine administration not only normalized depression-like disturbances evoked by the prenatal stress procedure but also modulated the mitochondria-enriched subproteome profile in the hippocampus of adult offspring rats. In line with this, two-dimensional electrophoresis coupled with mass spectrometry showed a statistically significant down-regulation of 14-3-3 and cytochrome bc1 proteins and an up-regulation of COP9 signalosome expression after chronic imipramine treatment in the hippocampus of prenatally stressed offspring. Fluoxetine administration strongly up-regulated the expression of cathepsin D, one of the key proteins involved in the prevention of the development of neurodegenerative processes. Furthermore, this antidepressant treatment enhanced expression of proteins engaged in the improvement of learning and memory processes (STMN1, Dnm-1) as well as in mitochondrial biogenesis and defense against oxidative stress (DJ-1). These findings provide new evidence that chronic administration of antidepressants exerts a varied impact on the mitochondria-enriched subproteome in the hippocampus of adult rats following a prenatal stress procedure. In particular, the effect of fluoxetine requires additional experiments to elucidate the possible beneficial biological consequences underlying the effects mediated by this antidepressant.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Fluoxetina/farmacología , Hipocampo/citología , Imipramina/farmacología , Mitocondrias/efectos de los fármacos , Proteoma/efectos de los fármacos , Proteínas 14-3-3/metabolismo , Animales , Antidepresivos/uso terapéutico , Complejo del Señalosoma COP9/metabolismo , Catepsina D/metabolismo , Depresión/metabolismo , Depresión/patología , Modelos Animales de Enfermedad , Dinaminas/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Femenino , Fluoxetina/uso terapéutico , Imipramina/uso terapéutico , Pérdida de Tono Postural/efectos de los fármacos , Masculino , Mitocondrias/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Proteína Desglicasa DJ-1/metabolismo , Proteómica , Ratas , Estatmina/metabolismo , Estrés Psicológico
20.
Int J Mol Sci ; 18(2)2017 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-28218653

RESUMEN

The role of different genotypes of apolipoprotein E (apoE) in the etiology of Alzheimer's disease is widely recognized. It has been shown that altered functioning of apoE may promote 4-hydroxynonenal modification of mitochondrial proteins, which may result in mitochondrial dysfunction, aggravation of oxidative stress, and neurodegeneration. Mitochondrial aldehyde dehydrogenase (ALDH2) is an enzyme considered to perform protective function in mitochondria by the detoxification of the end products of lipid peroxidation, such as 4-hydroxynonenal and other reactive aldehydes. The goal of our study was to apply a differential proteomics approach in concert with molecular and morphological techniques to elucidate the changes in the frontal cortex and hippocampus of apolipoprotein E knockout (apoE-/-) mice upon treatment with Alda-1-a small molecular weight activator of ALDH2. Despite the lack of significant morphological changes in the brain of apoE-/- mice as compared to age-matched wild type animals, the proteomic and molecular approach revealed many changes in the expression of genes and proteins, indicating the impairment of energy metabolism, neuroplasticity, and neurogenesis in brains of apoE-/- mice. Importantly, prolonged treatment of apoE-/- mice with Alda-1 led to the beneficial changes in the expression of genes and proteins related to neuroplasticity and mitochondrial function. The pattern of alterations implies mitoprotective action of Alda-1, however, the accurate functional consequences of the revealed changes require further research.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/metabolismo , Apolipoproteínas E/deficiencia , Benzamidas/farmacología , Benzodioxoles/farmacología , Lóbulo Frontal/metabolismo , Hipocampo/metabolismo , Proteómica/métodos , Aldehídos/sangre , Animales , Apolipoproteínas E/metabolismo , Apoptosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Inflamación/patología , Marcaje Isotópico , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Biogénesis de Organelos , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...