Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Neuroinflammation ; 18(1): 290, 2021 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-34895261

RESUMO

BACKGROUND: Major depressive disorder (MDD) represents a major public health concern, particularly due to its steadily rising prevalence and the poor responsiveness to standard antidepressants notably in patients afflicted with chronic inflammatory conditions, such as obesity. This highlights the need to improve current therapeutic strategies, including by targeting inflammation based on its role in the pathophysiology and treatment responsiveness of MDD. Nevertheless, dissecting the relative contribution of inflammation in the development and treatment of MDD remains a major issue, further complicated by the lack of preclinical depression models suitable to experimentally dissociate inflammation-related vs. inflammation-unrelated depression. METHODS: While current models usually focus on one particular MDD risk factor, we compared in male C57BL/6J mice the behavioral, inflammatory and neurobiological impact of chronic exposure to high-fat diet (HFD), a procedure known to induce inflammation-related depressive-like behaviors, and unpredictable chronic mild stress (UCMS), a stress-induced depression model notably renowned for its responsivity to antidepressants. RESULTS: While both paradigms induced neurovegetative, depressive-like and anxiety-like behaviors, inflammation and downstream neurobiological pathways contributing to inflammation-driven depression were specifically activated in HFD mice, as revealed by increased circulating levels of inflammatory factors, as well as brain expression of microglial activation markers and enzymes from the kynurenine and tetrahydrobiopterin (BH4) pathways. In addition, serotoninergic and dopaminergic systems were differentially impacted, depending on the experimental condition. CONCLUSIONS: These data validate an experimental design suitable to deeply study the mechanisms underlying inflammation-driven depression comparatively to non-inflammatory depression. This design could help to better understand the pathophysiology of treatment resistant depression.


Assuntos
Encéfalo/metabolismo , Transtorno Depressivo Maior/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Animais , Transtorno Depressivo Maior/etiologia , Transtorno Depressivo Maior/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Brain Behav Immun ; 80: 179-192, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30872090

RESUMO

The accumulation of adverse events in utero and during childhood differentially increases the vulnerability to psychiatric diseases in men and women. Gut microbiota is highly sensitive to the early environment and has been recently hypothesized to affect brain development. However, the impact of early-life adversity on gut microbiota, notably with regards to sex differences, remains to be explored. We examined the effects of multifactorial early-life adversity on behavior and microbiota composition in C3H/HeN mice of both sexes exposed to a combination of maternal immune activation (lipopolysaccharide injection on embryonic day 17, 120 µg/kg, i.p.), maternal separation (3hr per day from postnatal day (PND)2 to PND14) and maternal unpredictable chronic mild stress. At adulthood, offspring exposed to multi-hit early adversity showed sex-specific behavioral phenotypes with males exhibiting deficits in social behavior and females showing increased anxiety in the elevated plus maze and increased compulsive behavior in the marble burying test. Early adversity also differentially regulated gene expression in the medial prefrontal cortex (mPFC) according to sex. Interestingly, several genes such as Arc, Btg2, Fosb, Egr4 or Klf2 were oppositely regulated by early adversity in males versus females. Finally, 16S-based microbiota profiling revealed sex-dependent gut dysbiosis. In males, abundance of taxa belonging to Lachnospiraceae and Porphyromonadaceae families or other unclassified Firmicutes, but also Bacteroides, Lactobacillus and Alloprevotella genera was regulated by early adversity. In females, the effects of early adversity were limited and mainly restricted to Lactobacillus and Mucispirillum genera. Our work reveals marked sex differences in a multifactorial model of early-life adversity, both on emotional behaviors and gut microbiota, suggesting that sex should systematically be considered in preclinical studies both in neurogastroenterology and psychiatric research.


Assuntos
Microbioma Gastrointestinal/fisiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Animais , Animais Recém-Nascidos , Ansiedade/metabolismo , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Disbiose/metabolismo , Feminino , Masculino , Privação Materna , Camundongos , Camundongos Endogâmicos C3H , Microbiota , Córtex Pré-Frontal/metabolismo , Fatores Sexuais , Comportamento Social
3.
Proc Natl Acad Sci U S A ; 112(7): E738-46, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25646470

RESUMO

Allostatic load (AL) is a measure of overall physiological wear-and-tear over the life course, which could partially be the consequence of early life exposures. AL could allow a better understanding of the potential biological pathways playing a role in the construction of the social gradient in adult health. To explore the biological embedding hypothesis, we examined whether adverse childhood experiences (ACEs) are associated with elevated AL in midlife. We used imputed data on 3,782 women and 3,753 men of the National Child Development Study in Britain followed up seven times. ACEs were measured using prospective data collected at ages 7, 11, and 16. AL was operationalized using data from the biomedical survey collected at age 44 on 14 parameters representing four biological systems. We examined the role of adult health behaviors, body mass index (BMI), and socioeconomic status as potential mediators using a path analysis. ACEs were associated with higher AL for both men and women after adjustment for early life factors and childhood pathologies. The path analysis showed that the association between ACEs and AL was largely explained by early adult factors at age 23 and 33. For men, the total mediated effect was 59% (for two or more ACEs) via health behaviors, education level, and wealth. For women, the mediated effect represented 76% (for two or more ACEs) via smoking, BMI, education level, and wealth. Our results indicate that early psychosocial stress has an indirect lasting impact on physiological wear-and-tear via health behaviors, BMI, and socioeconomic factors in adulthood.


Assuntos
Nível de Saúde , Acontecimentos que Mudam a Vida , Estudos de Coortes , Humanos , Pessoa de Meia-Idade , Reino Unido
4.
J Neurosci ; 35(9): 4092-103, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25740536

RESUMO

In addition to metabolic and cardiovascular disorders, obesity is associated with adverse cognitive and emotional outcomes. Its growing prevalence during adolescence is particularly alarming since recent evidence indicates that obesity can affect hippocampal function during this developmental period. Adolescence is a decisive period for maturation of the amygdala and the hypothalamic-pituitary-adrenal (HPA) stress axis, both required for lifelong cognitive and emotional processing. However, little data are available on the impact of obesity during adolescence on amygdala function. Herein, we therefore evaluate in rats whether juvenile high-fat diet (HFD)-induced obesity alters amygdala-dependent emotional memory and whether it depends on HPA axis deregulation. Exposure to HFD from weaning to adulthood, i.e., covering adolescence, enhances long-term emotional memories as assessed by odor-malaise and tone-shock associations. Juvenile HFD also enhances emotion-induced neuronal activation of the basolateral complex of the amygdala (BLA), which correlates with protracted plasma corticosterone release. HFD exposure restricted to adulthood does not modify all these parameters, indicating adolescence is a vulnerable period to the effects of HFD-induced obesity. Finally, exaggerated emotional memory and BLA synaptic plasticity after juvenile HFD are alleviated by a glucocorticoid receptor antagonist. Altogether, our results demonstrate that juvenile HFD alters HPA axis reactivity leading to an enhancement of amygdala-dependent synaptic and memory processes. Adolescence represents a period of increased susceptibility to the effects of diet-induced obesity on amygdala function.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Emoções , Glucocorticoides/metabolismo , Memória , Plasticidade Neuronal , Obesidade/psicologia , Animais , Ansiedade/psicologia , Aprendizagem da Esquiva , Medo/psicologia , Masculino , Obesidade/fisiopatologia , Ratos , Ratos Wistar , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
5.
Front Nutr ; 10: 1190392, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37565037

RESUMO

Introduction: In humans, adversity in childhood exerts enduring effects on brain and increases the vulnerability to psychiatric diseases. It also leads to a higher risk of eating disorders and obesity. Maternal separation (MS) in mice has been used as a proxy of stress during infancy. We hypothesized that MS in mice affects motivation to obtain palatable food in adulthood and changes gene expression in reward system. Methods: Male and female pups from C57Bl/6J and C3H/HeN mice strains were subjected to a daily MS protocol from postnatal day (PND) 2 to PND14. At adulthood, their motivation for palatable food reward was assessed in operant cages. Results: Compared to control mice, male and female C3H/HeN mice exposed to MS increased their instrumental response for palatable food, especially when the effort required to obtain the reward was high. Importantly, this effect is shown in animals fed ad libitum. Transcriptional analysis revealed 375 genes differentially expressed in the nucleus accumbens of male MS C3H/HeN mice compared to the control group, some of these being associated with the regulation of the reward system (e.g., Gnas, Pnoc). Interestingly, C57Bl/6J mice exposed to MS did not show alterations in their motivation to obtain a palatable reward, nor significant changes in gene expression in the nucleus accumbens. Conclusion: MS produces long-lasting changes in motivation for palatable food in C3H/HeN mice, but has no impact in C57Bl/6J mice. These behavioral alterations are accompanied by drastic changes in gene expression in the nucleus accumbens, a key structure in the regulation of motivational processes.

6.
Front Immunol ; 13: 896179, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35677049

RESUMO

Type-2 diabetes is a complex disorder that is now considered to have an immune component, with functional impairments in many immune cell types. Type-2 diabetes is often accompanied by comorbid obesity, which is associated with low grade inflammation. However,the immune status in Type-2 diabetes independent of obesity remains unclear. Goto-Kakizaki rats are a non-obese Type-2 diabetes model. The limited evidence available suggests that Goto-Kakizaki rats have a pro-inflammatory immune profile in pancreatic islets. Here we present a detailed overview of the adult Goto-Kakizaki rat immune system. Three converging lines of evidence: fewer pro-inflammatory cells, lower levels of circulating pro-inflammatory cytokines, and a clear downregulation of pro-inflammatory signalling in liver, muscle and adipose tissues indicate a limited pro-inflammatory baseline immune profile outside the pancreas. As Type-2 diabetes is frequently associated with obesity and adipocyte-released inflammatory mediators, the pro-inflammatory milieu seems not due to Type-2 diabetes per se; although this overall reduction of immune markers suggests marked immune dysfunction in Goto-Kakizaki rats.


Assuntos
Diabetes Mellitus Tipo 2 , Animais , Biomarcadores , Sistema Imunitário , Obesidade , Ratos , Ratos Wistar
7.
Proc Nutr Soc ; 79(1): 113-132, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31250784

RESUMO

The developmental period constitutes a critical window of sensitivity to stress. Indeed, early-life adversity increases the risk to develop psychiatric diseases, but also gastrointestinal disorders such as the irritable bowel syndrome at adulthood. In the past decade, there has been huge interest in the gut-brain axis, especially as regards stress-related emotional behaviours. Animal models of early-life adversity, in particular, maternal separation (MS) in rodents, demonstrate lasting deleterious effects on both the gut and the brain. Here, we review the effects of MS on both systems with a focus on stress-related behaviours. In addition, we discuss more recent findings showing the impact of gut-directed interventions, including nutrition with pre- and probiotics, illustrating the role played by gut microbiota in mediating the long-term effects of MS. Overall, preclinical studies suggest that nutritional approaches with pro- and prebiotics may constitute safe and efficient strategies to attenuate the effects of early-life stress on the gut-brain axis. Further research is required to understand the complex mechanisms underlying gut-brain interaction dysfunctions after early-life stress as well as to determine the beneficial impact of gut-directed strategies in a context of early-life adversity in human subjects.


Assuntos
Gastroenteropatias/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Privação Materna , Animais , Encéfalo/fisiologia , Modelos Animais de Doenças , Feminino , Absorção Intestinal/fisiologia , Masculino , Camundongos , Prebióticos , Probióticos/metabolismo , Ratos
8.
Alcohol Clin Exp Res ; 33(8): 1346-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19413649

RESUMO

BACKGROUND: Although it is generally believed that chronic ethanol consumption impairs learning and memory, results obtained in experimental animals are not univocal, and there are conditions in which ethanol paradoxically improves cognitive functions. In the present work, we investigated the effects of prenatal stress and of chronic ethanol exposure during adulthood on spatial memory in rats. METHODS: Rats were subjected to a prenatal stress delivered as 3 daily 45-minute sections of restraint stress to the mothers during the last 10 days of pregnancy (PRS rats). After 7 months of ethanol exposure (ethanol 10%, oral intake), memory performances were evaluated in a spatial discrimination test in control and PRS male rats. Then, the oxidative damages and the expression of metabotropic glutamate (mGlu) receptors were assessed in their hippocampus. RESULTS: Chronic ethanol exposure resulted in a reduced performance in a spatial recognition task in control animals. Unexpectedly, however, the same treatment attenuated spatial memory deficits in rats that had been subjected to prenatal stress. This paradigm of ethanol administration did not produce detectable signs of oxidative damage in the hippocampus in either unstressed or PRS rats. Interestingly, ethanol intake resulted in differential effects in the expression of mGlu receptor subtypes implicated in mechanisms of learning and memory. In control rats, ethanol intake reduced mGlu2/3 and mGlu5 receptor levels in the hippocampus; in PRS rats, which exhibited a constitutive reduction in the levels of these mGlu receptor subtypes, ethanol increased the expression of mGlu1a receptors but did not change the expression of mGlu2/3 or mGlu5 receptors. CONCLUSION: Our findings support the idea that stress-related events occurring before birth have long-lasting effects on brain function and behavior, and suggest that the impact of ethanol on cognition is not only dose- and duration-dependent, but also critically influenced by early life experiences.


Assuntos
Etanol/administração & dosagem , Hipocampo/metabolismo , Transtornos da Memória/prevenção & controle , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Estresse Psicológico/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/efeitos dos fármacos , Masculino , Transtornos da Memória/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Comportamento Espacial/efeitos dos fármacos , Comportamento Espacial/fisiologia , Regulação para Cima/efeitos dos fármacos
9.
Brain Res Rev ; 57(2): 571-85, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18164765

RESUMO

Exposure to hostile conditions results in a series of coordinated responses aimed at enhancing the probability of survival. The activation of the hypothalamo-pituitary-adrenocortical (HPA) axis plays a pivotal role in the stress response. While the short-term activation of the HPA axis allows adaptive responses to the challenge, in the long run this can be devastating for the organism. In particular, life events occurring during the perinatal period have strong long-term effects on the behavioral and neuroendocrine response to stressors. In male and female rats exposed to prenatal restraint stress (PRS), these effects include a long-lasting hyperactivation of the HPA response associated with an altered circadian rhythm of corticosterone secretion. Furthermore, male animals exhibit sleep disturbances. In males, these HPA dysfunctions have been reported in infant, young, adult and aged animals, thus suggesting a permanent effect of early stress. Interestingly, after exposure to an intense inescapable footshock, female PRS rats durably exhibit a blunted corticosterone secretion response to stress. In male PRS rats exposed to an alcohol challenge, the HPA axis is similarly hyporesponsive. Rats exposed to PRS also show behavioral disturbances. Both male and female PRS rats show high anxiety levels and depression-like behavior during adulthood, although some studies suggest that female PRS rats present low anxiety levels. With ageing, male and female PRS rats exhibit memory impairments in hippocampus-dependent tasks, while female PRS rats improve their memory performance during adulthood. The gender effect on behavior seems to be related to a reduction in hippocampal plasticity in male PRS rats, and an increase in female PRS rats. Despite the permanent imprinting induced by early stress, the dysfunctions observed after PRS can be reversed by environmental or pharmacological strategies such as environmental enrichment or antidepressive and neurotrophic treatments. Mechanisms underlying the effects of PRS on the offspring remain largely unknown. However, previous studies have demonstrated that maternal glucocorticoids during pregnancy play an important role in the HPA disturbances reported in male offspring. Finally, gestational stress has long-lasting effects on the HPA axis and on behavior in the dams. Alterations in maternal behavior could thus also make a strong contribution to the long-term effects of PRS, through epigenetic mechanisms.


Assuntos
Epigênese Genética/fisiologia , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Animais , Feminino , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Sistema Hipófise-Suprarrenal/fisiopatologia , Gravidez , Ratos , Restrição Física
10.
Psychopharmacology (Berl) ; 236(5): 1583-1596, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31147734

RESUMO

RATIONALE: Intestinal permeability plays an important role in gut-brain axis communication. Recent studies indicate that intestinal permeability increases in neonate pups during maternal separation (MS). OBJECTIVES: The present study aims to determine whether pharmacological inhibition of myosin light chain kinase (MLCK), which regulates tight junction contraction and controls intestinal permeability, in stressed neonates, protects against the long-term effects of MS. METHODS: Male Wistar rats were exposed to MS (3 h per day from post-natal day (PND)2 to PND14) or left undisturbed and received daily intraperitoneal injection of a MLCK inhibitor (ML-7, 5 mg/kg) or vehicle during the same period. At adulthood, emotional behaviors, corticosterone response to stress, and gut microbiota composition were analyzed. RESULTS: ML-7 restored gut barrier function in MS rats specifically during the neonatal period. Remarkably, ML-7 prevented MS-induced sexual reward-seeking impairment and reversed the alteration of corticosterone response to stress at adulthood. The effects of ML-7 were accompanied by the normalization of the abundance of members of Lachnospiraceae, Clostridiales, Desulfovibrio, Bacteroidales, Enterorhabdus, and Bifidobacterium in the feces of MS rats at adulthood. CONCLUSIONS: Altogether, our work suggests that improvement of intestinal barrier defects during development may alleviate some of the long-term effects of early-life stress and provides new insight on brain-gut axis communication in a context of stress.


Assuntos
Azepinas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Privação Materna , Naftalenos/farmacologia , Estresse Psicológico/metabolismo , Animais , Animais Recém-Nascidos , Azepinas/uso terapêutico , Corticosterona/metabolismo , Relação Dose-Resposta a Droga , Feminino , Microbioma Gastrointestinal/fisiologia , Masculino , Quinase de Cadeia Leve de Miosina/farmacologia , Quinase de Cadeia Leve de Miosina/uso terapêutico , Naftalenos/uso terapêutico , Gravidez , Ratos , Ratos Wistar , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/psicologia , Fatores de Tempo
11.
Brain Res ; 1191: 55-62, 2008 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18096141

RESUMO

Prenatal restraint stress (PRS) in rats is associated with hippocampal dysfunctions and several behavioural and endocrine disorders related to this brain area. Recently, we have reported that the PRS modifies the hypothalamic-pituitary-adrenal (HPA) response to an ethanol challenge in adolescent animals. Since hippocampus is particularly sensitive to the deleterious effects of ethanol during adolescence, we investigated in this study the combined effects of PRS and ethanol administration on the oxidative status in the hippocampus of 28-day-old male rats. Thirty minutes after an intraperitoneal (i.p.) injection of ethanol (1.5 g/kg), the activities of several antioxidant enzymes (superoxide dismutase, catalase and glutathione peroxidase) but also non-enzymatic antioxidant (reduced glutathione) were assayed. Thiobarbituric acid reactive substances (TBARS) levels were also measured as a marker of lipid peroxidation. Ethanol enhanced superoxide dismutase activity in control rats but not in PRS rats. At basal level, catalase activity was lower in PRS rats than in control rats, indicating a potentially higher sensitivity to oxidative damages after this early stress. However, the hippocampal TBARS levels were not significantly affected by the ethanol administration, showing that an acute ethanol exposure does not induce oxidative damage in adolescent male rats. In conclusion, our data suggest that PRS affects both basal antioxidant status in the hippocampus and antioxidant response after an acute ethanol exposure. These findings extend previous works showing that PRS leads to hippocampal dysfunctions and raise the question of the potential increase of the hippocampal oxidative damage in PRS rats after repeated exposure to ethanol.


Assuntos
Intoxicação Alcoólica/enzimologia , Hipocampo/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Oxirredutases/metabolismo , Efeitos Tardios da Exposição Pré-Natal/enzimologia , Estresse Psicológico/enzimologia , Fatores Etários , Intoxicação Alcoólica/complicações , Animais , Antioxidantes/metabolismo , Catalase/efeitos dos fármacos , Catalase/metabolismo , Modelos Animais de Doenças , Etanol/farmacologia , Feminino , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Glutationa Peroxidase/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Masculino , Oxirredutases/efeitos dos fármacos , Gravidez , Ratos , Restrição Física , Estresse Psicológico/complicações , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
12.
Sci Rep ; 8(1): 17492, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30504818

RESUMO

Syndromes that display craniofacial anomalies comprise a major class of birth defects. Both genetic and environmental factors, including prenatal retinoic acid (RA) exposure, have been associated with these syndromes. While next generation sequencing has allowed the discovery of new genes implicated in these syndromes, some are still poorly characterized such as Oculo-Auriculo-Vertebral Spectrum (OAVS). Due to the lack of clear diagnosis for patients, developing new strategies to identify novel genes involved in these syndromes is warranted. Thus, our study aimed to explore the link between genetic and environmental factors. Owing to a similar phenotype of OAVS reported after gestational RA exposures in humans and animals, we explored RA targets in a craniofacial developmental context to reveal new candidate genes for these related disorders. Using a proteomics approach, we detected 553 dysregulated proteins in the head region of mouse embryos following their exposure to prenatal RA treatment. This novel proteomic approach implicates changes in proteins that are critical for cell survival/apoptosis and cellular metabolism which could ultimately lead to the observed phenotype. We also identified potential molecular links between three major environmental factors known to contribute to craniofacial defects including maternal diabetes, prenatal hypoxia and RA exposure. Understanding these links could help reveal common key pathogenic mechanisms leading to craniofacial disorders. Using both in vitro and in vivo approaches, this work identified two new RA targets, Gnai3 and Eftud2, proteins known to be involved in craniofacial disorders, highlighting the power of this proteomic approach to uncover new genes whose dysregulation leads to craniofacial defects.


Assuntos
Anormalidades Craniofaciais/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal , Tretinoína/toxicidade , Feminino , Humanos , Gravidez
13.
Neuropsychopharmacology ; 43(13): 2627-2635, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29487370

RESUMO

Depression is highly prevalent worldwide, but its etiology is not fully understood. An overlooked possible contributor to the epidemic of depression is feeding styles, particularly at early age when the brain is intensely changing. We have previously reported that unlimited sucrose consumption during adolescence leads to enduring changes in brain reward function. Here, we tested the hypothesis that sucrose consumption during adolescence would lead to a 'depressive-like' phenotype. Adolescent male rats were given unlimited access to 5% sucrose in their home cages from postnatal day 30 to postnatal day 46 and their emotional behavior was subsequently examined at adulthood. Sucrose consumption during adolescence caused anhedonia, decreased motivation for saccharin, increased immobility in the forced swim test and exacerbated anxiety-like behavior. Additionally, sucrose consumption during adolescence decreased cell proliferation in the hippocampus in adulthood. Chronic treatment with imipramine (10 mg/kg) normalized behavior and restored cell proliferation in the hippocampus of adult rats with a history of sucrose consumption during adolescence. A similar sucrose consumption starting at adulthood only increases immobility in the forced swim test, suggesting that sucrose intake affects also adults' behavior but to a lesser degree. Overall, our findings reveal an unsuspected protracted effect of sucrose consumption on behavior and suggest that unlimited sucrose consumption during critical periods of brain development may play an important role in the etiology of reward-related disorders such as depression.


Assuntos
Depressão/induzido quimicamente , Depressão/psicologia , Sacarose Alimentar/administração & dosagem , Sacarose Alimentar/efeitos adversos , Fenótipo , Fatores Etários , Animais , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Masculino , Ratos , Ratos Wistar , Natação/psicologia
14.
Brain Struct Funct ; 223(2): 883-895, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29022091

RESUMO

The medial prefrontal cortex (mPFC) is a key area for the regulation of numerous brain functions including stress response and cognitive processes. This brain area is also particularly affected by adversity during early life. Using an animal model in rats, we recently demonstrated that maternal exposure to a high-fat diet (HFD) prevents maternal separation (MS)-induced gene expression alterations in the developing PFC and attenuates several long-term deleterious behavioral effects of MS. In the present study, we ask whether maternal HFD could protect mPFC neurons of pups exposed to early life stress by examining dendritic morphology and spine density in juvenile [postnatal day (PND) 21] and adult rats submitted to MS. Dams were fed either a control or an HFD throughout gestation and lactation, and pups were submitted to MS from PND2 to PND14. We report that maternal HFD prevents MS-induced spine loss at PND21 and dendritic atrophy at adulthood. Furthermore, we show in adult MS rats that PFC-dependent memory extinction deficits are prevented by maternal HFD. Finally, perinatal HFD exposure reverses gut leakiness following stress in pups and seems to exert an anti-stress effect in dams. Overall, our work demonstrates that maternal HFD affects the developing brain and suggests that nutrition, possibly through gut-brain interactions, could modulate mPFC sensitivity to early stress.


Assuntos
Envelhecimento , Dendritos/patologia , Dieta Hiperlipídica , Troca Materno-Fetal/fisiologia , Córtex Pré-Frontal/patologia , Estresse Psicológico/patologia , Estresse Psicológico/prevenção & controle , Animais , Animais Recém-Nascidos , Contagem de Células , Dendritos/ultraestrutura , Feminino , Trato Gastrointestinal/fisiopatologia , Masculino , Neurônios/patologia , Neurônios/ultraestrutura , Odorantes , Permeabilidade , Córtex Pré-Frontal/crescimento & desenvolvimento , Gravidez , Ratos , Ratos Wistar , Privação de Água
15.
Psychoneuroendocrinology ; 32 Suppl 1: S57-61, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17618061

RESUMO

The decrease in some hormones with aging, such as insulin-like growth factor-I (IGF-I) and estradiol, may have a negative impact on brain function. Estradiol and IGF-I may antagonize the damaging effects of adrenal steroids and other causes of brain deterioration. The signaling of estradiol and IGF-I interact to promote neuroprotection. Estrogen receptor alpha, in an estrogen-dependent process, can physically interact with IGF-I receptor and with the downstream signaling molecules of the phosphotidylinositol 3-kinase (PI3K)/Akt/glycogen synthase kinase 3 (GSK3) pathway. Estradiol and IGF-I have a synergistic effect on the activation of Akt, which in turn decreases the activity of GSK3. This may be one of the mechanisms used by estradiol to promote neuronal survival, since the inhibition of GSK3 is associated to the activation of surviving signaling pathways in neurons. Furthermore, estradiol may control Tau phosphorylation by modulating the interactions of estrogen receptor alpha with GSK3 and beta-catenin, another molecule involved in the regulation of neuronal survival and the reorganization of the cytoskeleton. All these actions may be involved in the neuroprotective effects of the hormone. Possible aging-associated changes in the expression or activity of these signaling molecules may affect estradiol neuroprotective effects. Therefore, it is important to determine whether aging affects the signaling of estradiol and IGF-I in the brain.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiologia , Estradiol/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Transdução de Sinais/fisiologia , Corticosteroides/fisiologia , Animais , Humanos , Receptores de Estrogênio/fisiologia
16.
Psychoneuroendocrinology ; 32(7): 803-12, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17640823

RESUMO

The transition to motherhood results in a number of hormonal, neurological and behavioral changes necessary to ensure offspring survival. However, little attention has been paid to changes not directly linked to reproductive function in the early mother. In this study, we demonstrate that spatial performances during the learning phase were impaired after the delivery in rats, while spatial retention ability was improved 2 weeks later. In addition, we also report that early motherhood reduced the cell proliferation in the dentate gyrus of the hippocampus without inducing a decrease in the newborn cells 2 weeks later. The decrease of estradiol levels and high levels of glucocorticoids after delivery could in part explain the changes in the hippocampal function. In summary, our findings suggest that early postpartum period is associated with a modification of hippocampal function. This may reflect a homeostatic form of hippocampal plasticity in response to the onset of the maternal experience.


Assuntos
Hipocampo/fisiologia , Mães/psicologia , Animais , Antimetabólitos , Bromodesoxiuridina , Proliferação de Células , Sobrevivência Celular/fisiologia , Cognição/fisiologia , Corticosterona/sangue , Giro Denteado/citologia , Giro Denteado/crescimento & desenvolvimento , Estradiol/sangue , Feminino , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Aprendizagem em Labirinto/fisiologia , Plasticidade Neuronal/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Percepção Espacial/fisiologia
17.
Psychoneuroendocrinology ; 83: 49-57, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28595087

RESUMO

Early-life exposure to calorie-dense food, rich in fat and sugar, contributes to the increasing prevalence of obesity and its associated adverse cognitive and emotional outcomes at adulthood. It is thus critical to determine the impact of such nutritional environment on neurobehavioral development. In animals, maternal high-fat diet (HFD) consumption impairs hippocampal function in adult offspring, but its impact on hippocampal neuronal morphology is unknown. Moreover, the consequences of perinatal HFD exposure on the amygdala, another important structure for emotional and cognitive processes, remain to be established. In rats, we show that adult offspring from dams fed with HFD (45% from fat, throughout gestation and lactation) exhibit atrophy of pyramidal neuron dendrites in both the CA1 of the hippocampus and the basolateral amygdala (BLA). Perinatal HFD exposure also impairs conditioned odor aversion, a task highly dependent on BLA function, without affecting olfactory or malaise processing. Neuronal morphology and behavioral alterations elicited by perinatal HFD are not associated with body weight changes but with higher plasma leptin levels at postnatal day 15 and at adulthood. Taken together, our results suggest that perinatal HFD exposure alters hippocampal and amygdala neuronal morphology which could participate to memory alterations at adulthood.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Plasticidade Neuronal/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Filhos Adultos , Tonsila do Cerebelo/fisiologia , Animais , Peso Corporal , Dendritos/metabolismo , Dendritos/fisiologia , Feminino , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Leptina/sangue , Masculino , Exposição Materna/efeitos adversos , Memória/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Obesidade/psicologia , Gravidez , Ratos , Ratos Wistar , Lobo Temporal
18.
Neurobiol Aging ; 27(1): 119-27, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16298247

RESUMO

Stress during the prenatal period can induce permanent abnormalities in adult life such as increased anxiety-like behavior and hyperactivity of hypothalamo-pituitary-adrenal (HPA) axis system. The present study was designed to investigate whether prenatal stress could induce spatial learning impairment in aged female rats. Furthermore, since it has been recently reported that insulin-like growth factor 1 (IGF-1) attenuates spatial learning deficits in aged rats and promotes neurogenesis in the hippocampus, we assessed the impact of a chronic infusion of IGF-1 on age-related disorders. Our results show that females stressed during prenatal life exhibit learning impairments in the water maze task. Chronic IGF-1 treatment restores their spatial abilities, reduces their HPA axis dysfunction and increases plasma estradiol levels. Parallel to these effects, chronic IGF-1 up-regulates neural proliferation in the dentate gyrus of the hippocampus. These findings support the hypothesis of an early programming of the vulnerability to some neurological diseases during senescence and reinforce the potential therapeutic interest of IGF-1 during brain aging.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/fisiopatologia , Fator de Crescimento Insulin-Like I/administração & dosagem , Aprendizagem em Labirinto/efeitos dos fármacos , Complicações na Gravidez , Estresse Fisiológico/complicações , Animais , Transtornos Cognitivos/etiologia , Estrogênios/sangue , Feminino , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Infusões Parenterais , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiopatologia , Gravidez , Ratos , Ratos Sprague-Dawley
19.
Psychoneuroendocrinology ; 31(1): 92-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16081221

RESUMO

Gender is an important factor in the vulnerability to develop psychopathologies. At the biological level, stress-related pathologies such as depression or post-traumatic stress disorder (PTSD) are associated with profound disturbances of the hypothalamo-pituitary-adrenal (HPA) axis. The aim of the present study was to assess sex-differences in the long-term effect of an intense stressful procedure on HPA function and behaviour in the aversive context in rats. Female and male rats experienced an aversive procedure consisting in an electric footshock (2mA, 10s) in a dark chamber followed by 3 weekly situational reminders (SR, 2min in the white chamber close to the footshock chamber). Our results indicate that 41 days after the end of the aversive procedure, female rats showed an increase of the corticosterone negative feedback in response to restraint stress, whereas such effect was not observed in males. Despite this change in the hormonal response, glucocorticoid receptors mRNA expression in the hippocampus was not affected in shocked females. In contrast, a significant increase of the mineralocorticoid receptors mRNA was observed in the CA2 of the hippocampus in shocked males. Finally, CRH mRNA levels in the paraventricular nucleus of the hypothalamus (PVN) were decreased in both female and male animals exposed to the aversive procedure. Behavioural observation revealed that shocked males and shocked females showed a high level of avoidance. However, the latency to visit the shock box was lower in females, which spent also more time in this area than males. In conclusion, our results suggest that gender might be a key factor impacting the direction of the effects induced by an intense stress. Interestingly, only females exhibited an increased negative feedback of the HPA axis response to stress, which could parallel endocrine changes of PTSD.


Assuntos
Comportamento Animal/fisiologia , Eletrochoque , Sistema Hipotálamo-Hipofisário/fisiopatologia , Estresse Psicológico/fisiopatologia , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/biossíntese , Feminino , Hibridização In Situ , Masculino , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores de Esteroides/biossíntese , Restrição Física , Caracteres Sexuais
20.
Psychoneuroendocrinology ; 31(6): 769-80, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16624492

RESUMO

Prenatal stress (PS) durably influences responses of rats from birth throughout life by inducing deficits of the hypothalamo-pituitary-adrenal (HPA) axis feedback. The neuronal mechanisms sustaining such alterations are still unknown. The purpose of the present study was to determine whether in PS and control rats, the exposure to a mild stressor differentially induces Fos protein in hippocampus and locus coeruleus, brain areas involved in the feedback control of the HPA axis. Moreover, Fos protein expression was also evaluated in the hypothalamic paraventricular nucleus (PVN) that reflect the magnitude of the hormonal response to stress. Basal plasma corticosterone levels were not different between the groups, while, PS rats exhibited higher number of Fos-immunoreactive neurons than controls, in the hippocampus and locus coeruleus in basal condition. A higher basal expression of a marker of GABAergic synapses, the vGAT, was also observed in the hypothalamus of PS rats. Fifteen minutes after the end of the exposure to the open arm of the elevated plus-maze (mild stress) a similar increased plasma corticosterone levels was observed in both groups in parallel with an increased number of Fos-immunoreactive neurons in the PVN. Return to basal plasma corticosterone values was delayed only in the PS rats. On the contrary, after stress, no changes in Fos-immunoreactivity were observed in the hippocampus and locus coeruleus of PS rats compared to basal condition. After stress, only PS rats presented an elevation of the number of activated catecholaminergic neurons in the locus coeruleus. In conclusion, these results suggest for the first time that PS alters the neuronal activation of hippocampus and locus coeruleus implicated in the feedback mechanism of the HPA axis. These data give anatomical substrates to sustain the HPA axis hyperactivity classically described in PS rats after stress exposure.


Assuntos
Hipocampo/metabolismo , Locus Cerúleo/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Psicológico/metabolismo , Animais , Corticosterona/sangue , Feminino , Hipocampo/citologia , Sistema Hipotálamo-Hipofisário/citologia , Sistema Hipotálamo-Hipofisário/metabolismo , Locus Cerúleo/citologia , Masculino , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Gravidez , Ratos , Distribuição Tecidual , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA