Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
World J Biol Psychiatry ; 24(7): 578-586, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36748398

RESUMO

OBJECTIVES: The ability to identify persons at elevated risk for post-traumatic stress disorder (PTSD) soon after exposure to trauma, could aid clinical decision-making and treatment. In this study, we explored whether cytosine methylation of the 1 F promoter of the NR3C1 (glucocorticoid receptor [GR]) gene obtained immediately following a trauma could predict PTSD. METHODS: Our sample comprised 52 trauma survivors (28 women, 24 men), presenting to the Emergency Department (ED) within six hours of a traumatic event and followed for 13 months. Blood samples were taken at intake (n = 42) and again at the end of the study (13 months later, n = 27) to determine NR3C1-1F promoter methylation as well as plasma levels of cortisol, adrenocorticotropic-hormone (ACTH), and neuropeptide-Y (NPY). RESULTS: At the 13-month follow-up, participants who met the PTSD criteria (n = 4) showed significantly lower NR3C1-1F promoter sum percent methylation compared to the non-PTSD group (n = 38). Further, NR3C1-1F methylation at ED intake was inversely correlated with PTSD severity 13 months later, indicating that lower NR3C1-1F promoter methylation in the immediate aftermath of trauma was associated with the development of PTSD. CONCLUSION: To the extent that reduced promoter methylation is associated with greater GR expression and responsivity, this finding is consistent with the hypothalamic-pituitary-adrenal dysregulation previously described for PTSD.


Assuntos
Transtornos de Estresse Pós-Traumáticos , Masculino , Humanos , Feminino , Transtornos de Estresse Pós-Traumáticos/genética , Receptores de Glucocorticoides/genética , Metilação de DNA , Hidrocortisona/metabolismo
2.
Nat Neurosci ; 25(11): 1434-1445, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36266471

RESUMO

Post-traumatic stress disorder (PTSD) can develop following severe trauma, but the extent to which genetic and environmental risk factors contribute to individual clinical outcomes is unknown. Here, we compared transcriptional responses to hydrocortisone exposure in human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons and peripheral blood mononuclear cells (PBMCs) from combat veterans with PTSD (n = 19 hiPSC and n = 20 PBMC donors) and controls (n = 20 hiPSC and n = 20 PBMC donors). In neurons only, we observed diagnosis-specific glucocorticoid-induced changes in gene expression corresponding with PTSD-specific transcriptomic patterns found in human postmortem brains. We observed glucocorticoid hypersensitivity in PTSD neurons, and identified genes that contribute to this PTSD-dependent glucocorticoid response. We find evidence of a coregulated network of transcription factors that mediates glucocorticoid hyper-responsivity in PTSD. These findings suggest that induced neurons represent a platform for examining the molecular mechanisms underlying PTSD, identifying biomarkers of stress response, and conducting drug screening to identify new therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas , Transtornos de Estresse Pós-Traumáticos , Humanos , Transtornos de Estresse Pós-Traumáticos/genética , Glucocorticoides/farmacologia , Leucócitos Mononucleares , Interação Gene-Ambiente , Células-Tronco Pluripotentes Induzidas/metabolismo , Expressão Gênica , Neurônios/metabolismo
3.
Mol Psychiatry ; 27(4): 2225-2246, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35177824

RESUMO

Despite experiencing a significant trauma, only a subset of World Trade Center (WTC) rescue and recovery workers developed posttraumatic stress disorder (PTSD). Identification of biomarkers is critical to the development of targeted interventions for treating disaster responders and potentially preventing the development of PTSD in this population. Analysis of gene expression from these individuals can help in identifying biomarkers of PTSD. We established a well-phenotyped sample of 371 WTC responders, recruited from a longitudinal WTC responder cohort using stratified random sampling, by obtaining blood, self-reported and clinical interview data. Using bulk RNA-sequencing from whole blood, we examined the association between gene expression and WTC-related PTSD symptom severity on (i) highest lifetime Clinician-Administered PTSD Scale (CAPS) score, (ii) past-month CAPS score, and (iii) PTSD symptom dimensions using a 5-factor model of re-experiencing, avoidance, emotional numbing, dysphoric arousal and anxious arousal symptoms. We corrected for sex, age, genotype-derived principal components and surrogate variables. Finally, we performed a meta-analysis with existing PTSD studies (total N = 1016), using case/control status as the predictor and correcting for these variables. We identified 66 genes significantly associated with total highest lifetime CAPS score (FDR-corrected p < 0.05), and 31 genes associated with total past-month CAPS score. Our more granular analyses of PTSD symptom dimensions identified additional genes that did not reach statistical significance in our analyses with total CAPS scores. In particular, we identified 82 genes significantly associated with lifetime anxious arousal symptoms. Several genes significantly associated with multiple PTSD symptom dimensions and total lifetime CAPS score (SERPINA1, RPS6KA1, and STAT3) have been previously associated with PTSD. Geneset enrichment of these findings has identified pathways significant in metabolism, immune signaling, other psychiatric disorders, neurological signaling, and cellular structure. Our meta-analysis revealed 10 genes that reached genome-wide significance, all of which were downregulated in cases compared to controls (CIRBP, TMSB10, FCGRT, CLIC1, RPS6KB2, HNRNPUL1, ALDOA, NACA, ZNF429 and COPE). Additionally, cellular deconvolution highlighted an enrichment in CD4 T cells and eosinophils in responders with PTSD compared to controls. The distinction in significant genes between total lifetime CAPS score and the anxious arousal symptom dimension of PTSD highlights a potential biological difference in the mechanism underlying the heterogeneity of the PTSD phenotype. Future studies should be clear about methods used to analyze PTSD status, as phenotypes based on PTSD symptom dimensions may yield different gene sets than combined CAPS score analysis. Potential biomarkers implicated from our meta-analysis may help improve therapeutic target development for PTSD.


Assuntos
Ataques Terroristas de 11 de Setembro , Transtornos de Estresse Pós-Traumáticos , Ansiedade , Canais de Cloreto , Expressão Gênica , Humanos , Proteínas de Ligação a RNA , Autorrelato , Ataques Terroristas de 11 de Setembro/psicologia , Transtornos de Estresse Pós-Traumáticos/diagnóstico
4.
Transl Psychiatry ; 11(1): 398, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34282125

RESUMO

Epigenetic changes are currently invoked as explanations for both the chronicity and tenacity of post-traumatic stress disorder (PTSD), a heterogeneous condition showing varying, sometimes idiosyncratic responses to treatment. This study evaluated epigenetic markers in the context of a randomized clinical trial of PTSD patients undergoing prolonged-exposure psychotherapy with and without a hydrocortisone augmentation prior to each session. The purpose of the longitudinal epigenome-wide analyses was to identify predictors of recovery (from pretreatment data) or markers associated with symptom change (based on differences between pre- and post-therapy epigenetic changes). The results of these analyses identified the CREB-BDNF signaling pathway, previously linked to startle reaction and fear learning and memory processes, as a convergent marker predicting both symptom change and severity. Several previous-reported resilience markers were also identified (FKBP5, NR3C1, SDK1, and MAD1L1) to associate with PTSD recovery in this study. Especially, the methylation levels of FKBP5 in the gene body region decreased significantly as CAPS score decreased in responders, while no changes occurred in nonresponders. These biomarkers may have future utility in understanding clinical recovery in PTSD and potential applications in predicting treatment effects.


Assuntos
Transtornos de Estresse Pós-Traumáticos , Metilação de DNA , Epigênese Genética , Epigenoma , Humanos , Hidrocortisona , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Transtornos de Estresse Pós-Traumáticos/genética
5.
Biol Psychiatry ; 77(4): 356-64, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24661442

RESUMO

BACKGROUND: Enhanced glucocorticoid receptor (GR) sensitivity is present in people with posttraumatic stress disorder (PTSD), but the molecular mechanisms of GR sensitivity are not understood. Epigenetic factors have emerged as one potential mechanism that account for how trauma exposure leads to sustained PTSD symptoms given that PTSD develops in only a subset of trauma survivors. METHODS: Cytosine methylation of a relevant promoter of the GR gene (NR3C1-1F promoter) and three functional neuroendocrine markers of hypothalamic-pituitary-adrenal axis function were examined in a sample of 122 combat veterans. RESULTS: Lower NR3C1-1F promoter methylation in peripheral blood mononuclear cells (PBMCs) was observed in combat veterans with PTSD compared with combat-exposed veterans who did not develop PTSD. NR3C1-1F promoter methylation was also associated with three functional measures of glucocorticoid activity that have been associated with PTSD in combat veterans: PBMCs' lysozyme inhibition on the lysozyme suppression test, plasma cortisol decline on the low-dose (.50 mg) dexamethasone suppression test, and 24-hour urinary cortisol excretion. Finally, NR3C1-1F promoter methylation was inversely correlated with clinical markers and symptoms associated with PTSD. CONCLUSIONS: Alterations in NR3C1-1F promoter methylation may reflect enduring changes resulting from combat exposure that lead to functional neuroendocrine alterations. Because epigenetic measures are thought to reflect enduring effects of environmental exposures, they may be useful in distinguishing combat-exposed veterans who do or do not develop PTSD.


Assuntos
Monócitos/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transtornos de Estresse Pós-Traumáticos/genética , Transtornos de Estresse Pós-Traumáticos/metabolismo , Veteranos/psicologia , Adulto , Biomarcadores/metabolismo , Biomarcadores/urina , Citosina/química , Metilação de DNA , Transtorno Depressivo Maior/complicações , Transtorno Depressivo Maior/genética , Dexametasona/metabolismo , Epigênese Genética , Humanos , Hidrocortisona/urina , Hipotálamo/metabolismo , Masculino , Muramidase/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Regiões Promotoras Genéticas , Transtornos de Estresse Pós-Traumáticos/complicações
6.
Am J Psychiatry ; 171(8): 872-880, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24832930

RESUMO

OBJECTIVE: Differential effects of maternal and paternal posttraumatic stress disorder (PTSD) have been observed in adult offspring of Holocaust survivors in both glucocorticoid receptor sensitivity and vulnerability to psychiatric disorder. The authors examined the relative influences of maternal and paternal PTSD on DNA methylation of the exon 1F promoter of the glucocorticoid receptor (GR-1F) gene (NR3C1) in peripheral blood mononuclear cells and its relationship to glucocorticoid receptor sensitivity in Holocaust offspring. METHOD: Adult offspring with at least one Holocaust survivor parent (N=80) and demographically similar participants without parental Holocaust exposure or parental PTSD (N=15) completed clinical interviews, self-report measures, and biological procedures. Blood samples were collected for analysis of GR-1F promoter methylation and of cortisol levels in response to low-dose dexamethasone, and two-way analysis of covariance was performed using maternal and paternal PTSD as main effects. Hierarchical clustering analysis was used to permit visualization of maternal compared with paternal PTSD effects on clinical variables and GR-1F promoter methylation. RESULTS: A significant interaction demonstrated that in the absence of maternal PTSD, offspring with paternal PTSD showed higher GR-1F promoter methylation, whereas offspring with both maternal and paternal PTSD showed lower methylation. Lower GR-1F promoter methylation was significantly associated with greater postdexamethasone cortisol suppression. The clustering analysis revealed that maternal and paternal PTSD effects were differentially associated with clinical indicators and GR-1F promoter methylation. CONCLUSIONS: This is the first study to demonstrate alterations of GR-1F promoter methylation in relation to parental PTSD and neuroendocrine outcomes. The moderation of paternal PTSD effects by maternal PTSD suggests different mechanisms for the intergenerational transmission of trauma-related vulnerabilities.


Assuntos
Filhos Adultos , Filho de Pais com Deficiência , Metilação de DNA , Epigênese Genética/genética , Holocausto/psicologia , Receptores de Glucocorticoides/genética , Transtornos de Estresse Pós-Traumáticos/genética , Sobreviventes/psicologia , Filhos Adultos/psicologia , Estudos de Casos e Controles , Filho de Pais com Deficiência/psicologia , Dexametasona , Pai , Feminino , Hereditariedade , Humanos , Hidrocortisona/sangue , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Mães , Testes de Função Adreno-Hipofisária , Regiões Promotoras Genéticas/genética
7.
Front Psychiatry ; 4: 118, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098286

RESUMO

Epigenetic alterations offer promise as diagnostic or prognostic markers, but it is not known whether these measures associate with, or predict, clinical state. These questions were addressed in a pilot study with combat veterans with PTSD to determine whether cytosine methylation in promoter regions of the glucocorticoid related NR3C1 and FKBP51 genes would predict or associate with treatment outcome. Veterans with PTSD received prolonged exposure (PE) psychotherapy, yielding responders (n = 8), defined by no longer meeting diagnostic criteria for PTSD, and non-responders (n = 8). Blood samples were obtained at pre-treatment, after 12 weeks of psychotherapy (post-treatment), and after a 3-month follow-up. Methylation was examined in DNA extracted from lymphocytes. Measures reflecting glucocorticoid receptor (GR) activity were also obtained (i.e., plasma and 24 h-urinary cortisol, plasma ACTH, lymphocyte lysozyme IC50-DEX, and plasma neuropeptide-Y). Methylation of the GR gene (NR3C1) exon 1F promoter assessed at pre-treatment predicted treatment outcome, but was not significantly altered in responders or non-responders at post-treatment or follow-up. In contrast, methylation of the FKBP5 gene (FKBP51) exon 1 promoter region did not predict treatment response, but decreased in association with recovery. In a subset, a corresponding group difference in FKBP5 gene expression was observed, with responders showing higher gene expression at post-treatment than non-responders. Endocrine markers were also associated with the epigenetic markers. These preliminary observations require replication and validation. However, the results support research indicating that some glucocorticoid related genes are subject to environmental regulation throughout life. Moreover, psychotherapy constitutes a form of "environmental regulation" that may alter epigenetic state. Finally, the results further suggest that different genes may be associated with prognosis and symptom state, respectively.

8.
Cancer Biol Ther ; 11(2): 204-12, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21057205

RESUMO

The molecular mechanisms conferring resistance to docetaxel in prostate cancer patients remain partially understood. We generated docetaxel resistant derivatives of the androgen independent prostate cancer cell lines PC-3 and DU-145. Docetaxel rapidly induces DU-145 cell death via apoptosis and the drug resistant cells were produced by periodically exposing proliferating DU-145 cultures to small doses of docetaxel. In PC-3 cells docetaxel induces delayed cell death via mitotic catastrophe evident by profound multinucleation and formation of giant cells. Mononucleated progeny of the giant PC-3 cells shows significant resistance to docetaxel. Gene expression profiling of these docetaxel resistant PC-3 cells revealed sets of docetaxel inducible and constitutively expressed genes associated with major cancer pathways. A contradictory overlap with DU-145 docetaxel resistant cells was also found. Analyses suggested significant changes associated with apoptotic function, DNA repair, cell growth, survival and proliferation, metabolism, maintenance of cytoskeleton and extracellular matrix formation. These cellular processes often contribute to drug resistance and our study identified a set of genes managing this phenotype. Additional analyses of the drug resistant PC-3 cells using shRNA constructs determined direct relevance of Cyclin G2 to docetaxel resistance as well as prevention of multinucleation, whereas the knockdown of upregulated CYP1B1 showed no effect on either of these processes. Downregulated GBP1 was explored by ectopic overexpression and even though GBP1 has a potential to mediate resistance to docetaxel, it was not utilized in PC-3 cells. The results suggest complex combination of gene expression pattern changes that enables resistance to docetaxel while preventing death via multinucleation.


Assuntos
Antineoplásicos/farmacologia , Perfilação da Expressão Gênica , Neoplasias da Próstata/tratamento farmacológico , Taxoides/farmacologia , Androgênios/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Docetaxel , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Concentração Inibidora 50 , Masculino , Análise em Microsséries , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
J Neurochem ; 116(4): 577-87, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21166678

RESUMO

Considerable evidence suggests that receptor-mediated excitation and inhibition of brainstem pedunculopontine tegmental (PPT) neurons are critically involved in the regulation of sleep-wake states. However, the molecular mechanisms operating within the PPT-controlling sleep-wake states remain relatively unknown. This study was designed to examine sleep-wake state-associated extracellular-signal-regulated kinase 1 and 2 (ERK1/2) transduction changes in the PPT of freely moving rats. The results of this study demonstrate that the levels of ERK1/2 expression, phosphorylation, and activity in the PPT increased with increased amount of time spent in sleep. The sleep-associated increases in ERK1/2 expression, phosphorylation, and activity were not observed in the cortex, or in the immediately adjacent medial pontine reticular formation. The results of regression analyses revealed significant positive relationships between the levels of ERK1/2 expression, phosphorylation, and activity in the PPT and amounts of time spent in slow-wave sleep, rapid eye movement sleep, and total sleep. Additionally, these regression analyses revealed significant negative relationships between the levels of ERK1/2 expression, phosphorylation, and activity in the PPT and amounts of time spent in wakefulness. Collectively, these results, for the first time, suggest that the increased ERK1/2 signaling in the PPT is associated with maintenance of sleep via suppression of wakefulness.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Núcleo Tegmental Pedunculopontino/enzimologia , Transdução de Sinais/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Núcleo Tegmental Pedunculopontino/citologia , Ratos , Ratos Wistar , Fases do Sono/fisiologia
10.
J Neurosci ; 30(37): 12263-73, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844122

RESUMO

Intracellular signaling mechanisms within the pedunculopontine tegmental (PPT) nucleus for the regulation of recovery rapid eye movement (REM) sleep following REM sleep deprivation remain unknown. This study was designed to determine the role of PPT intracellular cAMP-dependent protein kinase A (cAMP-PKA) in the regulation of recovery REM sleep in freely moving rats. The results show that a brief period (3 h) of selective REM sleep deprivation caused REM sleep rebound associated with increased PKA activity and expression of the PKA catalytic subunit protein (PKA-CU) in the PPT. Local application of a cAMP-PKA-activation-selective inhibitor, RpCAMPS (0.55, 1.1, and 2.2 nmol/100 nl; n = 8 rats/group), bilaterally into the PPT, reduced PKA activity and PKA-CU expression in the PPT, and suppressed the recovery REM sleep, in a dose-dependent manner. Regression analyses revealed significant positive relationships between: PPT levels of PKA activity and the total percentages of REM sleep recovery (Rsqr = 0.944; n = 40 rats); PPT levels of PKA-CU expression and the total percentages of REM sleep recovery (Rsqr = 0.937; n = 40 rats); PPT levels of PKA-CU expression and PKA activity (Rsqr = 0.945; n = 40 rats). Collectively, these results provide evidence that activation of intracellular PKA in the PPT contributes to REM sleep recovery following REM sleep deprivation.


Assuntos
Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Núcleo Tegmental Pedunculopontino/enzimologia , Subunidades Proteicas/fisiologia , Privação do Sono/enzimologia , Sono REM/fisiologia , Animais , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/antagonistas & inibidores , Ativação Enzimática/fisiologia , Líquido Intracelular/enzimologia , Masculino , Neurônios/enzimologia , Núcleo Tegmental Pedunculopontino/citologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/fisiologia , Privação do Sono/fisiopatologia
11.
J Neurochem ; 112(1): 271-81, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19860859

RESUMO

Considerable evidence suggests that the brainstem pedunculopontine tegmentum (PPT) neurons are critically involved in the regulation of rapid eye movement (REM) sleep and wakefulness (W); however, the molecular mechanisms operating within the PPT to regulate these two behavioral states remain relatively unknown. Here we demonstrate that the levels of calcium/calmodulin kinase II (CaMKII) and phosphorylated CaMKII expression in the PPT decreased and increased with 'low W with high REM sleep' and 'high W/low REM sleep' periods, respectively. These state-specific expression changes were not observed in the cortex, or in the immediately adjacent medial pontine reticular formation. Next, we demonstrate that CaMKII activity in the PPT is negatively and positively correlated with the 'low W with high REM sleep' and 'high W/low REM sleep' periods, respectively. These differences in correlations were not seen in the medial pontine reticular formation CaMKII activity. Finally, we demonstrate that with increased PPT CaMKII activity observed during high W/low REM sleep, there were marked shifts in the expression of genes that are involved in components of various signal transduction pathways. Collectively, these results for the first time suggest that the increased CaMKII activity within PPT neurons is associated with increased W at the expense of REM sleep, and this process is accomplished through the activation of a specific gene expression profile.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Núcleo Tegmental Pedunculopontino/enzimologia , Sono REM/fisiologia , Vigília/fisiologia , Animais , Tronco Encefálico/enzimologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Masculino , Ratos , Ratos Wistar
12.
Cell Adh Migr ; 2(1): 38-44, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19262122

RESUMO

Stress is an important physiological regulator of brain function in young and adult mammals. The mechanisms underlying regulation of the consequences of stress, and in particular severe chronic stress, are thus important to investigate. These consequences most likely involve changes in synaptic function of brain areas being part of neural networks that regulate responses to stress. Cell adhesion molecules have been shown to regulate synaptic function in the adult and we were thus interested to investigate a regulatory mechanism that could influence expression of three adhesion molecules of the immunoglobulin superfamily (NCAM, L1 and CHL1) after exposure of early postnatal and adult mice to repeated stress. We hypothesized that reduction of adhesion molecule expression after chronic stress, as observed previously in vivo, could be due to gene silencing of the three molecules by DNA methylation. Although adhesion molecule expression was reduced after exposure of C57BL/6 mice to stress, thus validating our stress paradigm as imposing changes in adhesion molecule expression, we did not observe differences in methylation of CpG islands in the promoter regions of NCAM, L1 and CHL1, nor in the promoter region of the glucocorticoid receptor in the hippocampus, the expression of which at the protein level was also reduced after stress. We must therefore infer that severe stress in mice of the C57BL/6 strain downregulates adhesion molecule levels by mechanisms that do not relate to DNA methylation.


Assuntos
Antígeno CD56/metabolismo , Moléculas de Adesão Celular/metabolismo , Regulação para Baixo , Hipocampo/metabolismo , Estresse Fisiológico , Animais , Antígeno CD56/genética , Moléculas de Adesão Celular/genética , Ilhas de CpG/genética , Metilação de DNA , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética
13.
Life Sci ; 79(1): 30-7, 2006 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-16434061

RESUMO

The diurnal variations of the endocannabinoid arachidonoylethanolamine (anandamide, ANA) as well as palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) were detected and quantified in cerebrospinal fluid (CSF), pons, hippocampus, and hypothalamus in the rat over 24 h using HPLC/MS. In CSF, the 3 compounds presented an increase in their concentration during the lights-on period and a remarkable decrease in their values during the lights-off period. In the pons, ANA, PEA and OEA showed the maximum values during the dark phase. On the other hand, we found that in the hippocampus, ANA increased its concentration during the lights-off period and PEA showed the highest peak at the beginning of the same period. OEA concentration showed no diurnal variations in the hippocampus. Finally, in the hypothalamus, ANA rose during the lights-on period whereas PEA and OEA presented the highest concentration at the end of the lights-off period. We postulate that all compounds are likely to be accumulated in parenchyma during the lights-off period (when animal is awake) and then, released into the CSF in order to reach target regions in turn to modulate diverse behaviors, such as feeding and sleep.


Assuntos
Ácidos Araquidônicos/metabolismo , Química Encefálica/fisiologia , Ritmo Circadiano/fisiologia , Ácidos Oleicos/metabolismo , Ácidos Palmíticos/metabolismo , Amidas , Animais , Ácidos Araquidônicos/líquido cefalorraquidiano , Cromatografia Líquida de Alta Pressão , Endocanabinoides , Etanolaminas , Hipocampo/metabolismo , Hipotálamo/metabolismo , Masculino , Espectrometria de Massas , Ácidos Oleicos/líquido cefalorraquidiano , Ácidos Palmíticos/líquido cefalorraquidiano , Alcamidas Poli-Insaturadas , Ponte/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Sleep ; 27(5): 851-6, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15453542

RESUMO

STUDY OBJECTIVES: Hypocretins (HCRT-1 and HCRT-2), also known as orexins, are neuropeptides localized in neurons surrounding the perifornical region of the posterior hypothalamus. These neurons project to major arousal centers in the brain and are implicated in regulating wakefulness. In young rats and monkeys, levels of HCRT-1 are highest at the end of the wake-active period and lowest toward the end of the sleep period. However, the effects of age on the diurnal rhythm of HCRT-1 are not known. DESIGN: To provide such data, cerebrospinal fluid (CSF) was collected from the cisterna magna of young (2-month-old, n = 9), middle-aged (12 months, n = 10), and old (24 months, n = 10) F344 rats at 4-hour intervals, (beginning at zeitgeber [ZT]0, lights on). CSF was collected once from each rat every 4 days at 1 ZT point. After collecting the CSF at all of the time points, the rats were kept awake by gentle handling for 8 hours (ZT 0-ZT8), and the CSF was collected again at the end of the sleep-deprivation procedure. HCRT-1 levels in the CSF were determined by radioimmunoassay SETTINGS: Basic neuroscience research lab. MEASUREMENTS AND RESULTS: Old rats had significantly less HCRT-1 in the CSF versus young and middle-aged rats (P < .002) during the lights-on and lights-off periods and over the 24-hour period. In old rats, significantly low levels of HCRT-1 were evident at the end of the lights-off period (predominantly wake-active period). The old rats continued to have less HCRT-1 even after 8 hours of prolonged waking. Northern blot analysis did not show a difference in pre-proHCRT mRNA between age groups. CONCLUSIONS: In old rats there is a 10% decline in CSF HCRT-1 over the 24-hour period. Functionally, if there is less HCRT-1, which our findings indicated, and there is also a decline in HCRT receptor mRNA, as has been previously found, then the overall consequence would be diminished action of HCRT at target sites. This would diminish the waking drive, which in the elderly could contribute to the increased tendency to fall asleep during the normal wake period.


Assuntos
Envelhecimento/fisiologia , Proteínas de Transporte/biossíntese , Ritmo Circadiano/fisiologia , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/biossíntese , Vigília/fisiologia , Animais , Proteínas de Transporte/líquido cefalorraquidiano , Hipotálamo/anatomia & histologia , Neuropeptídeos/líquido cefalorraquidiano , Orexinas , RNA Mensageiro/análise , Radioimunoensaio , Ratos , Ratos Endogâmicos F344 , Sono/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA