Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 39(7): 866-881, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33621399

RESUMO

A key challenge for clinical application of induced pluripotent stem cells (iPSC) to accurately model and treat human pathologies depends on developing a method to generate genetically stable cells to reduce long-term risks of cell transplant therapy. Here, we hypothesized that CYCLIN D1 repairs DNA by highly efficient homologous recombination (HR) during reprogramming to iPSC that reduces genetic instability and threat of neoplastic growth. We adopted a synthetic mRNA transfection method using clinically compatible conditions with CYCLIN D1 plus base factors (OCT3/4, SOX2, KLF4, LIN28) and compared with methods that use C-MYC. We demonstrate that CYCLIN D1 made iPSC have (a) lower multitelomeric signal, (b) reduced double-strand DNA breaks, (c) correct nuclear localization of RAD51 protein expression, and (d) reduced single-nucleotide polymorphism (SNP) changes per chromosome, compared with the classical reprogramming method using C-MYC. CYCLIN D1 iPSC have reduced teratoma Ki67 cell growth kinetics and derived neural stem cells successfully engraft in a hostile spinal cord injury (SCI) microenvironment with efficient survival, differentiation. We demonstrate that CYCLIN D1 promotes double-stranded DNA damage repair predominantly through HR during cell reprogramming to efficiently produce iPSC. CYCLIN D1 reduces general cell stress associated with significantly lower SIRT1 gene expression and can rescue Sirt1 null mouse cell reprogramming. In conclusion, we show synthetic mRNA transfection of CYCLIN D1 repairs DNA during reprogramming resulting in significantly improved genetically stable footprint in human iPSC, enabling a new cell reprogramming method for more accurate and reliable generation of human iPSC for disease modeling and future clinical applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Reprogramação Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Reparo do DNA/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Eur J Neurosci ; 50(5): 2773-2785, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31231836

RESUMO

Chronic stress is a major risk factor for developing Alzheimer's disease (AD) and promotes the processing of amyloid precursor protein (APP) to ß-amyloid (Aß). However, the precise relationship of stress and disease-typical cognitive decline is presently not well understood. The aim of this study was to investigate how early life stress may affect cognition in adult mice with and without soluble Aß pathology typical for the early stages of the disease. We focussed on sustained attention and response control, aspects of cognition mediated by the prefrontal cortex that are consistently impaired both in early AD and after chronic stress exposure. Young wild-type mice as well as transgenic arcAß mice overexpressing the hAPParc/swe transgene were exposed to a chronic unpredictable stress paradigm (age 3-8 weeks). At 15 weeks, these mice were tested on the 5-choice serial reaction time task, a test of sustained attention and executive control. We found that, expectedly, chronic stress increased impulsive choices and impaired sustained attention in wild-type mice. However, the same treatment reduced impulsivity and did not interfere with sustained attention in arcAß mice. These findings suggest an unexpected interaction between chronic stress and Aß whereby Aß-pathology caused by the hAPParc/swe mutation prevented and/or reversed stress-induced cognitive changes through mechanisms that deserve further investigation. They also indicate that Aß, in modest amounts, may have a beneficial role for cognitive stability, for example by protecting neural networks from the impact of further physiological or behavioural stress.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Cognição/fisiologia , Função Executiva/fisiologia , Estresse Psicológico/genética , Doença de Alzheimer/genética , Animais , Atenção/fisiologia , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Comportamento Impulsivo/fisiologia , Masculino , Camundongos , Mutação , Tempo de Reação/fisiologia
3.
Stem Cells ; 37(4): 476-488, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30664289

RESUMO

When considering the clinical applications of autologous cell replacement therapy of human induced pluripotent stem cells (iPSC)-derived cells, there is a clear need to better understand what the immune response will be before we embark on extensive clinical trials to treat or model human disease. We performed a detailed assessment comparing human fibroblast cell lines (termed F1) reprogrammed into human iPSC and subsequently differentiated back to fibroblast cells (termed F2) or other human iPSC-derived cells including neural stem cells (NSC) made from either retroviral, episomal, or synthetic mRNA cell reprogramming methods. Global proteomic analysis reveals the main differences in signal transduction and immune cell protein expression between F1 and F2 cells, implicating wild type (WT) toll like receptor protein 3 (TLR3). Furthermore, global methylome analysis identified an isoform of the human TLR3 gene that is not epigenetically reset correctly upon differentiation to F2 cells resulting in a hypomethylated transcription start site in the TLR3 isoform promoter and overexpression in most human iPSC-derived cells not seen in normal human tissue. The human TLR3 isoform in human iPSC-NSC functions to suppress NF-KB p65 signaling pathway in response to virus (Poly IC), suggesting suppressed immunity of iPSC-derived cells to viral infection. The sustained WT TLR3 and TLR3 isoform overexpression is central to understanding the altered immunogenicity of human iPSC-derived cells calling for screening of human iPSC-derived cells for TLR3 expression levels before applications. Stem Cells 2019;37:476-488.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Proteômica/métodos , Receptor 3 Toll-Like/metabolismo , Epigenoma , Humanos , Imunidade Inata , Células-Tronco Pluripotentes Induzidas/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/imunologia
4.
Front Cell Neurosci ; 12: 131, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867364

RESUMO

The locus coeruleus (LC), which contains the largest group of noradrenergic neurons in the central nervous system innervating the telencephalon, is an early and constantly vulnerable region to neurofibrillary tangle (NFT) pathology in aging and Alzheimer's disease (AD). The present study using whole genome bisulfite sequencing and Infinium Human Methylation 450 BeadChip was designed to learn about DNA methylation profiles in LC with age and NFT pathology. This method identified decreased DNA methylation of Katanin-Interacting Protein gene (KIAA0566) linked to age and presence of NFT pathology. KIAA0566 mRNA expression demonstrated with RT-qPCR significantly decreased in cases with NFT pathology. Importantly, KIAA0566 immunoreactivity was significantly decreased only in LC neurons with NFTs, but not in neurons without tau pathology when compared with neurons of middle-aged individuals. These changes were accompanied by a similar pattern of selective p80-katanin reduced protein expression in neurons with NFTs. In contrast, p60-katanin subunit expression levels in the neuropil were similar in MA cases and cases with NFT pathology. Since katanin is a major microtubule-severing protein and KIAA0566 binds and interacts with katanin, de-regulation of the katanin-signaling pathway may have implications in the regulation of microtubule homeostasis in LC neurons with NFTs, thereby potentially interfering with maintenance of the cytoskeleton and transport.

5.
Cell Rep ; 23(6): 1665-1677, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29742424

RESUMO

Rett syndrome (RTT) is the second leading cause of mental impairment in girls and is currently untreatable. RTT is caused, in more than 95% of cases, by loss-of-function mutations in the methyl CpG-binding protein 2 gene (MeCP2). We propose here a molecular target involved in RTT: the glycogen synthase kinase-3b (Gsk3b) pathway. Gsk3b activity is deregulated in Mecp2-knockout (KO) mice models, and SB216763, a specific inhibitor, is able to alleviate the clinical symptoms with consequences at the molecular and cellular levels. In vivo, inhibition of Gsk3b prolongs the lifespan of Mecp2-KO mice and reduces motor deficits. At the molecular level, SB216763 rescues dendritic networks and spine density, while inducing changes in the properties of excitatory synapses. Gsk3b inhibition can also decrease the nuclear activity of the Nfkb1 pathway and neuroinflammation. Altogether, our findings indicate that Mecp2 deficiency in the RTT mouse model is partially rescued following treatment with SB216763.


Assuntos
Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Proteína 2 de Ligação a Metil-CpG/deficiência , Subunidade p50 de NF-kappa B/metabolismo , Síndrome de Rett/metabolismo , Síndrome de Rett/patologia , Transdução de Sinais , Sinapses/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Cerebelo/metabolismo , Cerebelo/patologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Indóis/farmacologia , Inflamação/patologia , Longevidade , Maleimidas/farmacologia , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Análise de Sobrevida , Regulação para Cima/efeitos dos fármacos
6.
Clin Epigenetics ; 9: 67, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28670349

RESUMO

Ageing is the main risk factor for human neurological disorders. Among the diverse molecular pathways that govern ageing, epigenetics can guide age-associated decline in part by regulating gene expression and also through the modulation of genomic instability and high-order chromatin architecture. Epigenetic mechanisms are involved in the regulation of neural differentiation as well as in functional processes related to memory consolidation, learning or cognition during healthy lifespan. On the other side of the coin, many neurodegenerative diseases are associated with epigenetic dysregulation. The reversible nature of epigenetic factors and, especially, their role as mediators between the genome and the environment make them exciting candidates as therapeutic targets. Rather than providing a broad description of the pathways epigenetically deregulated in human neurological disorders, in this review, we have focused on the potential use of epigenetic enzymes as druggable targets to ameliorate neural decline during normal ageing and especially in neurological disorders. We will firstly discuss recent progress that supports a key role of epigenetic regulation during healthy ageing with an emphasis on the role of epigenetic regulation in adult neurogenesis. Then, we will focus on epigenetic alterations associated with ageing-related human disorders of the central nervous system. We will discuss examples in the context of psychiatric disorders, including schizophrenia and posttraumatic stress disorders, and also dementia or Alzheimer's disease as the most frequent neurodegenerative disease. Finally, methodological limitations and future perspectives are discussed.


Assuntos
Envelhecimento/genética , Encefalopatias/genética , Metilação de DNA , Neurogênese , Epigênese Genética , Redes Reguladoras de Genes , Humanos
7.
Adv Exp Med Biol ; 978: 443-475, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28523560

RESUMO

Despite the enormous efforts of the scientific community over the years, effective therapeutics for many (epi)genetic brain disorders remain unidentified. The common and persistent failures to translate preclinical findings into clinical success are partially attributed to the limited efficiency of current disease models. Although animal and cellular models have substantially improved our knowledge of the pathological processes involved in these disorders, human brain research has generally been hampered by a lack of satisfactory humanized model systems. This, together with our incomplete knowledge of the multifactorial causes in the majority of these disorders, as well as a thorough understanding of associated (epi)genetic alterations, has been impeding progress in gaining more mechanistic insights from translational studies. Over the last years, however, stem cell technology has been offering an alternative approach to study and treat human brain disorders. Owing to this technology, we are now able to obtain a theoretically inexhaustible source of human neural cells and precursors in vitro that offer a platform for disease modeling and the establishment of therapeutic interventions. In addition to the potential to increase our general understanding of how (epi)genetic alterations contribute to the pathology of brain disorders, stem cells and derivatives allow for high-throughput drugs and toxicity testing, and provide a cell source for transplant therapies in regenerative medicine. In the current chapter, we will demonstrate the validity of human stem cell-based models and address the utility of other stem cell-based applications for several human brain disorders with multifactorial and (epi)genetic bases, including Parkinson's disease (PD), Alzheimer's disease (AD), fragile X syndrome (FXS), Angelman syndrome (AS), Prader-Willi syndrome (PWS), and Rett syndrome (RTT).


Assuntos
Encefalopatias/terapia , Avaliação Pré-Clínica de Medicamentos/métodos , Epigênese Genética , Doenças Genéticas Inatas/terapia , Doenças Neurodegenerativas/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos , Animais , Encefalopatias/genética , Transplante de Tecido Encefálico , Modelos Animais de Doenças , Transplante de Tecido Fetal , Previsões , Doenças Genéticas Inatas/genética , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/genética , Medicina Regenerativa/tendências , Pesquisa com Células-Tronco , Transplante de Células-Tronco/métodos
8.
Synapse ; 71(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28105729

RESUMO

The brain's neocortex is anatomically organized into grey and white matter, which are mainly composed by neuronal and glial cells, respectively. The neocortex can be further divided in different Brodmann areas according to their cytoarchitectural organization, which are associated with distinct cortical functions. There is increasing evidence that brain development and function are governed by epigenetic processes, yet their contribution to the functional organization of the neocortex remains incompletely understood. Herein, we determined the DNA methylation patterns of grey and white matter of dorsolateral prefrontal cortex (Brodmann area 9), an important region for higher cognitive skills that is particularly affected in various neurological diseases. For avoiding interindividual differences, we analyzed white and grey matter from the same donor using whole genome bisulfite sequencing, and for validating their biological significance, we used Infinium HumanMethylation450 BeadChip and pyrosequencing in ten and twenty independent samples, respectively. The combination of these analysis indicated robust grey-white matter differences in DNA methylation. What is more, cell type-specific markers were enriched among the most differentially methylated genes. Interestingly, we also found an outstanding number of grey-white matter differentially methylated genes that have previously been associated with Alzheimer's, Parkinson's, and Huntington's disease, as well as Multiple and Amyotrophic lateral sclerosis. The data presented here thus constitute an important resource for future studies not only to gain insight into brain regional as well as grey and white matter differences, but also to unmask epigenetic alterations that might underlie neurological and neurodegenerative diseases.


Assuntos
Metilação de DNA , Substância Cinzenta/metabolismo , Córtex Pré-Frontal/metabolismo , Substância Branca/metabolismo , Idoso , Feminino , Genoma Humano , Humanos , Masculino , Pessoa de Meia-Idade , Especificidade de Órgãos
9.
Psychoneuroendocrinology ; 63: 94-101, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26433325

RESUMO

There have been numerous studies into the interaction between stress and addictive drugs, yet few have specifically addressed how the organism responds to stress when under the influence of psychostimulants. Thus, we studied the effects of different acute stressors (immobilization, interleukin-1ß and forced swimming) in young adult male rats simultaneously exposed to amphetamine (AMPH, 4 mg/kg SC), evaluating classic biological markers. AMPH administration itself augmented the plasma hypothalamic-pituitary-adrenal (HPA) hormones, adrenocorticotropin (ACTH) and corticosterone, without affecting plasma glucose levels. By contrast, this drug dampened the peripheral HPA axis, as well as the response of glucose to the three stressors. We also found that AMPH administration completely blocked the forced swim-induced expression of the corticotropin-releasing hormone (hnCRH) and it partially reduced c-fos expression in the paraventricular nucleus of the hypothalamus (PVN). Indeed, this negative synergy in the forced swim test could even be observed with a lower dose of AMPH (1mg/kg, SC), a dose that is usually received in self-administration experiments. In conclusion, when rats that receive AMPH are subjected to stress, a negative synergy occurs that dampens the prototypic peripheral physiological response to stress and activation of the PVN.


Assuntos
Hormônio Adrenocorticotrópico/efeitos dos fármacos , Anfetamina/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Interleucina-1beta/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Estresse Psicológico/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Sinergismo Farmacológico , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Restrição Física , Natação
10.
Genet Med ; 18(4): 378-85, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26181491

RESUMO

PURPOSE: Autism spectrum disorders are associated with defects in social response and communication that often occur in the context of intellectual disability. Rett syndrome is one example in which epilepsy, motor impairment, and motor disturbance may co-occur. Mutations in histone demethylases are known to occur in several of these syndromes. Herein, we aimed to identify whether mutations in the candidate histone demethylase JMJD1C (jumonji domain containing 1C) are implicated in these disorders. METHODS: We performed the mutational and functional analysis of JMJD1C in 215 cases of autism spectrum disorders, intellectual disability, and Rett syndrome without a known genetic defect. RESULTS: We found seven JMJD1C variants that were not present in any control sample (~ 6,000) and caused an amino acid change involving a different functional group. From these, two de novo JMJD1C germline mutations were identified in a case of Rett syndrome and in a patient with intellectual disability. The functional study of the JMJD1C mutant Rett syndrome patient demonstrated that the altered protein had abnormal subcellular localization, diminished activity to demethylate the DNA damage-response protein MDC1, and reduced binding to MECP2. We confirmed that JMJD1C protein is widely expressed in brain regions and that its depletion compromises dendritic activity. CONCLUSIONS: Our findings indicate that mutations in JMJD1C contribute to the development of Rett syndrome and intellectual disability.Genet Med 18 1, 378-385.


Assuntos
Deficiência Intelectual/genética , Histona Desmetilases com o Domínio Jumonji/genética , Mutação , Oxirredutases N-Desmetilantes/genética , Síndrome de Rett/genética , Adulto , Motivos de Aminoácidos , Sequência de Aminoácidos , Transtorno do Espectro Autista/diagnóstico , Transtorno do Espectro Autista/genética , Encéfalo/metabolismo , Encéfalo/patologia , Sequência Conservada , Análise Mutacional de DNA , Feminino , Expressão Gênica , Ordem dos Genes , Estudos de Associação Genética , Loci Gênicos , Humanos , Deficiência Intelectual/diagnóstico , Histona Desmetilases com o Domínio Jumonji/química , Histona Desmetilases com o Domínio Jumonji/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Neurônios/metabolismo , Oxirredutases N-Desmetilantes/química , Oxirredutases N-Desmetilantes/metabolismo , Matrizes de Pontuação de Posição Específica , Conformação Proteica , Transporte Proteico , Síndrome de Rett/diagnóstico
11.
Acta Neuropathol Commun ; 3: 76, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26621506

RESUMO

INTRODUCTION: Parkinson's disease (PD) is characterized by the accumulation of abnormal α-synuclein in selected regions of the brain following a gradient of severity with disease progression. Whether this is accompanied by globally altered protein synthesis is poorly documented. The present study was carried out in PD stages 1-6 of Braak and middle-aged (MA) individuals without alterations in brain in the substantia nigra, frontal cortex area 8, angular gyrus, precuneus and putamen. RESULTS: Reduced mRNA expression of nucleolar proteins nucleolin (NCL), nucleophosmin (NPM1), nucleoplasmin 3 (NPM3) and upstream binding transcription factor (UBF), decreased NPM1 but not NPM3 nucleolar protein immunostaining in remaining neurons; diminished 18S rRNA, 28S rRNA; reduced expression of several mRNAs encoding ribosomal protein (RP) subunits; and altered protein levels of initiation factor eIF3 and elongation factor eEF2 of protein synthesis was found in the substantia nigra in PD along with disease progression. Although many of these changes can be related to neuron loss in the substantia nigra, selective alteration of certain factors indicates variable degree of vulnerability of mRNAs, rRNAs and proteins in degenerating sustantia nigra. NPM1 mRNA and 18S rRNA was increased in the frontal cortex area 8 at stage 5-6; modifications were less marked and region-dependent in the angular gyrus and precuneus. Several RPs were abnormally regulated in the frontal cortex area 8 and precuneus, but only one RP in the angular gyrus, in PD. Altered levels of eIF3 and eIF1, and decrease eEF1A and eEF2 protein levels were observed in the frontal cortex in PD. No modifications were found in the putamen at any time of the study except transient modifications in 28S rRNA and only one RP mRNA at stages 5-6. These observations further indicate marked region-dependent and stage-dependent alterations in the cerebral cortex in PD. Altered solubility and α-synuclein oligomer formation, assessed in total homogenate fractions blotted with anti-α-synuclein oligomer-specific antibody, was demonstrated in the substantia nigra and frontal cortex, but not in the putamen, in PD. Dramatic increase in α-synuclein oligomers was also seen in fluorescent-activated cell sorter (FACS)-isolated nuclei in the frontal cortex in PD. CONCLUSIONS: Altered machinery of protein synthesis is altered in the substantia nigra and cerebral cortex in PD being the frontal cortex area 8 more affected than the angular gyrus and precuneus; in contrast, pathways of protein synthesis are apparently preserved in the putamen. This is associated with the presence of α-synuclein oligomeric species in total homogenates; substantia nigra and frontal cortex are enriched, albeit with different band patterns, in α-synuclein oligomeric species, whereas α-synuclein oligomers are not detected in the putamen.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Fator de Iniciação 3 em Eucariotos/metabolismo , Feminino , Humanos , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosfoproteínas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , alfa-Sinucleína/genética , Nucleolina
12.
Eur Neuropsychopharmacol ; 25(8): 1248-59, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26092203

RESUMO

There is evidence that endogenous cannabinoids (eCBs) play a role in the control of the hypothalamic-pituitary-adrenal (HPA) axis, although they appear to have dual, stimulatory and inhibitory, effects. Recent data in rats suggest that eCBs, acting through CB1 receptors (CB1R), may be involved in adaptation of the HPA axis to daily repeated stress. In the present study we analyze this issue in male mice and rats. Using a knock-out mice for the CB1 receptor (CB1-/-) we showed that mutant mice presented similar adrenocorticotropic hormone (ACTH) response to the first IMO as wild-type mice. Daily repeated exposure to 1h of immobilization reduced the ACTH response to the stressor, regardless of the genotype, demonstrating that adaptation occurred to the same extent in absence of CB1R. Prototypical changes observed after repeated stress such as enhanced corticotropin releasing factor (CRH) gene expression in the paraventricular nucleus of the hypothalamus, impaired body weight gain and reduced thymus weight were similarly observed in both genotypes. The lack of effect of CB1R in the expression of HPA adaptation to another similar stressor (restraint) was confirmed in wild-type CD1 mice by the lack of effect of the CB1R antagonist AM251 just before the last exposure to stress. Finally, the latter drug did not blunt the HPA, glucose and behavioral adaptation to daily repeated forced swim in rats. Thus, the present results indicate that CB1R is not critical for overall effects of daily repeated stress or proper adaptation of the HPA axis in mice and rats.


Assuntos
Adaptação Psicológica/fisiologia , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Estresse Psicológico/metabolismo , Adaptação Psicológica/efeitos dos fármacos , Hormônio Adrenocorticotrópico/metabolismo , Animais , Peso Corporal , Antagonistas de Receptores de Canabinoides/farmacologia , Corticosterona/sangue , Modelos Animais de Doenças , Glucose/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Camundongos Knockout , Piperidinas/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Pirazóis/farmacologia , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/genética , Restrição Física , Natação
13.
Neuropharmacology ; 79: 75-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24212060

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis is activated by a wide range of stimuli, including drugs. Here we report that in male rats, a dose of sodium butyrate (NaBu) that is typically used to inhibit histone deacetylation (1200 mg/kg) increased the peripheral levels of HPA hormones and glucose. In a further experiment, we compared the effects of two different doses of NaBu (200 and 1200 mg/kg) and equimolar saline solutions on peripheral neuroendocrine markers and brain c-Fos expression to demonstrate a specific stress-like effect of NaBu that is not related to hypertonicity and to localise putatively involved brain areas. Only the high dose of NaBu increased the plasma levels of stress markers. The equimolar (hypertonic) saline solution also activated the HPA axis and the c-Fos expression in the paraventricular nucleus of the hypothalamus (PVN), a key area for the control of the HPA axis, but the effects were of a lower magnitude than those of NaBu. Regarding other brain areas, group differences in c-Fos expression were not observed in the medial prefrontal cortex or the medial amygdala, but they were observed in the central amygdala and the lateral ventral septum. However, only the latter area of the NaBu group showed enhanced c-Fos expression that was significantly higher than that after hypertonic saline. The present data indicate that high doses of NaBu appear to act as a pharmacological stressor, and this fact should be taken into account when using this drug to study the role of epigenetic processes in learning and emotional behaviour.


Assuntos
Encéfalo/efeitos dos fármacos , Ácido Butírico/administração & dosagem , Inibidores de Histona Desacetilases/administração & dosagem , Estresse Fisiológico/efeitos dos fármacos , Acetilação , Hormônio Adrenocorticotrópico/sangue , Animais , Glicemia/efeitos dos fármacos , Encéfalo/fisiopatologia , Ácido Butírico/efeitos adversos , Corticosterona/sangue , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Inibidores de Histona Desacetilases/efeitos adversos , Histonas/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiopatologia , Masculino , Camundongos Endogâmicos A , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Sódio/sangue , Estresse Fisiológico/fisiologia
14.
Stress ; 16(6): 698-705, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23924206

RESUMO

Comparison of exposure to certain predominantly emotional stressors reveals a qualitatively similar neuroendocrine response profile as well as a reduction of physiological responses after daily repeated exposure (adaptation). However, particular physical components of the stressor may interfere with adaptation. As defective adaptation to stress can enhance the probability to develop pathologies, we studied in adult male rats (n = 10/group) swimming behavior (struggling, immobility and mild swim) and physiological responses (ACTH, corticosterone and rectal temperature) to daily repeated exposure to forced swim (20 min, 13 d) at 25 or 36 °C (swim25 or swim36). Rats were repeatedly blood-sampled by tail-nick and hormones measured by radioimmunoassay. Some differences were observed between the two swim temperature groups after the first exposure to forced swim: (a) active behaviors were greater in swim25 than swim36 groups; (b) swim25 but not swim36 caused hypothermia; and (c) swim36 elicited the same ACTH response as swim25, but plasma corticosterone concentration was lower for swim36 at 30 min post-swim. After daily repeated exposure, adaptation in ACTH secretion was observed with swim36 already on day 4, whereas with swim25 adaptation was not observed until day 13 and was of lower magnitude. Nevertheless, after repeated exposure to swim25 a partial protection from hypothermia was observed and the two swim conditions resulted in progressive reduction of active behaviors. Thus, daily repeated swim at 25 °C impairs adaptation of the hypothalamic-pituitary-adrenal axis as compared to swim at 36 °C, supporting the hypothesis that certain physical components of predominantly emotional stressors can interfere with the process of adaptation.


Assuntos
Hormônio Adrenocorticotrópico/sangue , Corticosterona/sangue , Sistema Hipófise-Suprarrenal/fisiologia , Estresse Psicológico/sangue , Natação/fisiologia , Temperatura , Adaptação Fisiológica/fisiologia , Animais , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotermia/etiologia , Masculino , Ratos , Ratos Sprague-Dawley , Natação/psicologia , Água
15.
Front Behav Neurosci ; 6: 69, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23112767

RESUMO

Exposure of animals to footshocks (FS) in absence of any specific cue results in the development of fear to the compartment where shocks were given (contextual fear conditioning), and this is usually evaluated by time spent freezing. However, the extent to which contextual fear conditioning always develops when animals are exposed to other stressors is not known. In the present work we firstly demonstrated, using freezing, that exposure of adult rats to a single session of FS resulted in short-term and long-term contextual fear conditioning (freezing) that was paralleled by increased hypothalamic-pituitary-adrenal (HPA) activation. In contrast, using a similar design, no HPA or behavioral evidence for such conditioning was found after exposure to immobilization on boards (IMO), despite this stressor being of similar severity as FS on the basis of standard physiological measures of stress, including HPA activation. In a final experiment we directly compared the exposure to the two stressors in the same type of context and tested for the development of conditioning to the context and to a specific cue for IMO (the board). We observed the expected high levels of freezing and the conditioned HPA activation after FS, but not after IMO, regardless of the presence of the board during testing. Therefore, it can be concluded that development of fear conditioning to context or particular cues, as evaluated by either behavioral or endocrine measures, appears to be dependent on the nature of the aversive stimuli, likely to be related to biologically preparedness to establish specific associations.

16.
Physiol Behav ; 105(3): 669-75, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22019828

RESUMO

Brain substance P and its receptor (neurokinin-1, NK1) have a widespread brain distribution and are involved in an important number of behavioural and physiological responses to emotional stimuli. However, the role of NK1 receptors in the consequences of exposure to chronic stress has not been explored. The present study focused on the role of these receptors in the hypothalamic-pituitary-adrenal (HPA) response to daily repeated restraint stress (evaluated by plasma corticosterone levels), as well as on the effect of this procedure on anxiety-like behaviour, spatial learning and memory in the Morris water maze (MWM), a hippocampus-dependent task. Adult null mutant NK1-/- mice, with a C57BL/6J background, and the corresponding wild-type mice showed similar resting corticosterone levels and, also, did not differ in corticosterone response to a first restraint. Nevertheless, adaptation to the repeated stressor was faster in NK1-/- mice. Chronic restraint modestly increased anxiety-like behaviour in the light-dark test, irrespective of genotype. Throughout the days of the MWM trials, NK1-/- mice showed a similar learning rate to that of wild-type mice, but had lower levels of thigmotaxis and showed a better retention in the probe trial. Chronic restraint stress did not affect these variables in either genotype. These results indicate that deletion of the NK1 receptor does not alter behavioural susceptibility to chronic repeated stress in mice, but accelerates adaptation of the HPA axis. In addition, deletion may result in lower levels of thigmotaxis and improved short-term spatial memory, perhaps reflecting a better learning strategy in the MWM.


Assuntos
Comportamento Animal/fisiologia , Corticosterona/sangue , Receptores da Neurocinina-1/deficiência , Estresse Psicológico , Adaptação Ocular , Animais , Distribuição de Qui-Quadrado , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Radioimunoensaio/métodos , Restrição Física/métodos , Estresse Psicológico/sangue , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Fatores de Tempo
17.
Behav Brain Res ; 217(1): 232-9, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20937327

RESUMO

Repeated exposure to the same stressor very often results in a reduction of some prototypical stress responses, namely those related to the hypothalamic-pituitary-adrenal (HPA) and sympatho-medullo-adrenal (SMA) axes. This reduced response to repeated exposure to the same (homotypic) stressor (adaptation) is usually considered as a habituation-like process, and therefore, a non-associative type of learning. However, there is some evidence that contextual cues and therefore associative processes could contribute to adaptation. In the present study we demonstrated in two experiments using adult male rats that repeated daily exposure to restraint (REST) or immobilization on boards (IMO) reduced the HPA (plasma levels of ACTH and corticosterone) and glucose responses to the homotypic stressor and such reduced responses remained intact when all putative cues associated to the procedure (experimenter, way of transporting to the stress room, stress boxes, stress room and colour of the restrainer in the case of REST) were modified on the next day. Therefore, the present results do not favour the view that adaptation after repeated exposure to a stressor may involve associative processes related to signals predicting the imminence of the stressors, but more studies are needed on this issue.


Assuntos
Adaptação Fisiológica/fisiologia , Condicionamento Clássico/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Imobilização/métodos , Sistema Hipófise-Suprarrenal/fisiologia , Restrição Física/métodos , Estresse Psicológico/fisiopatologia , Hormônio Adrenocorticotrópico/sangue , Animais , Glicemia/metabolismo , Corticosterona/sangue , Sistema Hipotálamo-Hipofisário/metabolismo , Imobilização/fisiologia , Masculino , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/sangue
18.
J Psychiatr Res ; 44(3): 157-67, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19698958

RESUMO

Stressful life events increase the susceptibility for subsequent onset of psychiatric disorders in humans. Previous research has implicated neurotrophins in the onset of some stress-related diseases, such as major depression disorder, post-traumatic stress disorder or panic disorder. We have tested the hypothesis that the neurotrophin-3 (NT-3)/TrkC system is a genetic interface mediating the deleterious effects of stress on the initiation of panic disorder and other pathologies. To this aim, we have analyzed the functionality of HPA axis and the behavioral consequences of different types of stressful conditions in a mouse model of panic disorder, which overexpresses TrkC, the high affinity-receptor for NT-3 (TgNTRK3). Our results reveal that TgNTRK3 mice exhibit an altered circadian corticosterone rhythm that is reversed by clonidine treatment, but normal expression of genes involved in the control of the hypothalamus-pituitary-adrenal (HPA) axis (CRH, GR) and normal corticosterone response to acute and chronic stressors. In contrast, they exhibit an altered pattern of activation of stress-related brain areas and showed enhanced anxiety-related behavior and more passive strategies than wild types under some chronic stress conditions. We conclude that TgNTRK3 mice present differences in their response to stress characterized by subtle changes in the HPA axis, marked changes in acute stress-induced brain activation and altered coping strategies, suggesting a key role of TrkC receptor in the stress neural circuitry and in the behavioral consequences of chronic stress.


Assuntos
Ansiedade/genética , Predisposição Genética para Doença , Fatores de Crescimento Neural/metabolismo , Transtorno de Pânico/genética , Receptor trkC/genética , Estresse Psicológico/genética , Animais , Comportamento Animal/fisiologia , Ritmo Circadiano/fisiologia , Corticosterona/fisiologia , Modelos Animais de Doenças , Sistema Hipotálamo-Hipofisário/fisiopatologia , Camundongos , Camundongos Transgênicos , Transtorno de Pânico/fisiopatologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Receptor trkC/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...