Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(12): e0256973, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34879063

RESUMO

We recently identified a nuclear-encoded miRNA (miR-181c) in cardiomyocytes that can translocate into mitochondria to regulate mitochondrial gene mt-COX1 and influence obesity-induced cardiac dysfunction through the mitochondrial pathway. Because liver plays a pivotal role during obesity, we hypothesized that miR-181c might contribute to the pathophysiological complications associated with obesity. Therefore, we used miR-181c/d-/- mice to study the role of miR-181c in hepatocyte lipogenesis during diet-induced obesity. The mice were fed a high-fat (HF) diet for 26 weeks, during which indirect calorimetric measurements were made. Quantitative PCR (qPCR) was used to examine the expression of genes involved in lipid synthesis. We found that miR-181c/d-/- mice were not protected against all metabolic consequences of HF exposure. After 26 weeks, the miR-181c/d-/- mice had a significantly higher body fat percentage than did wild-type (WT) mice. Glucose tolerance tests showed hyperinsulinemia and hyperglycemia, indicative of insulin insensitivity in the miR-181c/d-/- mice. miR-181c/d-/- mice fed the HF diet had higher serum and liver triglyceride levels than did WT mice fed the same diet. qPCR data showed that several genes regulated by isocitrate dehydrogenase 1 (IDH1) were more upregulated in miR-181c/d-/- liver than in WT liver. Furthermore, miR-181c delivered in vivo via adeno-associated virus attenuated the lipogenesis by downregulating these same lipid synthesis genes in the liver. In hepatocytes, miR-181c regulates lipid biosynthesis by targeting IDH1. Taken together, the data indicate that overexpression of miR-181c can be beneficial for various lipid metabolism disorders.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hepatócitos/metabolismo , Lipogênese , Fígado/metabolismo , MicroRNAs/metabolismo , Obesidade , Triglicerídeos , Animais , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/metabolismo , Triglicerídeos/biossíntese , Triglicerídeos/genética
2.
Int J Eat Disord ; 54(4): 639-645, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33368559

RESUMO

OBJECTIVE: Patients with Anorexia Nervosa (AN) display increased levels of oxidative stress that correlates with disease severity. Unfortunately, the biological ramifications of AN-induced oxidative stress on the brain are largely unknown. Our lab uses the preclinical activity-based anorexia (ABA) paradigm to model symptoms of AN. The goal of the present study was to determine how ABA experience affects oxidative state and its consequences in adolescent female rats. METHOD: We compared systemic glutathione and cysteine plasma concentrations and medial prefrontal cortex (mPFC) mitochondrial fission in ABA animals at maximum weight loss or following 10-days of weight recovery to levels in age-matched sedentary (SED) control rats. RESULTS: ABA animals at maximum weight loss had significantly lower plasma levels of cysteine and glutathione compared to SED controls. Additionally, ABA animals at max weight loss have significantly more mPFC mitochondrial fission. There were no significant differences in plasma analyte levels or mitochondrial fission between weight recovered ABA animals and SED controls. DISCUSSION: These data suggest that ABA experience results in oxidative stress that is remedied after weight restoration. The long-lasting ramifications of transient periods of increased oxidative stress are unknown and can lead to significant consequences on brain function and behavior.


Assuntos
Anorexia Nervosa , Anorexia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Dinâmica Mitocondrial , Estresse Oxidativo , Ratos , Redução de Peso
3.
Carcinogenesis ; 41(12): 1713-1723, 2020 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-32249286

RESUMO

Children born to women who experience stress during pregnancy have an increased risk of cancer in later life, but no previous animal studies have tested such a link. We questioned whether prenatal stress (PS) in A/J mice affected the development of lung tumors after postnatal response to tobacco-specific nitrosamine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Timed-bred A/J mice were randomly assigned on gestation day 12.5 to PS by restraint for 5 consecutive days or control (no restraint). Adult offspring of control and stressed pregnancies were all treated with three NNK injections (50 mg/kg every other day) and euthanized 16 weeks later to examine their lungs. Compared with controls, PS dams exhibited significantly increased levels of plasma corticosterone, increased adrenal weights and decreased fetus weights without fetal loss. Prenatally stressed litters had a significantly higher neonatal death rate within first week of life, and surviving male and female offspring developed lung epithelial proliferations with increase multiplicity, increased area and aggressive morphology. PS also induced more advanced atypical adenomatous hyperplasia lesions. We found no difference in lung NNK-derived methyl DNA adducts, but PS did significantly enhance CD3+ T cell and Foxp3+ T cell tumor infiltration. PS significantly increases multiplicity, area of NNK-induced lung tumors and advanced morphology. PS did not affect production of NNK-derived methyl DNA adducts but did increase lymphocytic infiltration of lung tumors. To our knowledge, this is the first animal model of PS with evaluation of cancer development in offspring.


Assuntos
Neoplasias Pulmonares/patologia , Nitrosaminas/toxicidade , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Estresse Psicológico , Animais , Feminino , Neoplasias Pulmonares/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos A , Gravidez , Restrição Física
4.
Proc Natl Acad Sci U S A ; 114(39): 10479-10484, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28894008

RESUMO

Genome-wide association studies have implicated the ANK3 locus in bipolar disorder, a major human psychotic illness. ANK3 encodes ankyrin-G, which organizes the neuronal axon initial segment (AIS). We generated a mouse model with conditional disruption of ANK3 in pyramidal neurons of the adult forebrain (Ank-G cKO). This resulted in the expected loss of pyramidal neuron AIS voltage-gated sodium and potassium channels. There was also dramatic loss of markers of afferent GABAergic cartridge synapses, resembling the cortical microcircuitry changes in brains from psychotic patients, and suggesting disinhibition. Expression of c-fos was increased in cortical pyramidal neurons, consistent with increased neuronal activity due to disinhibition. The mice showed robust behavioral phenotypes reminiscent of aspects of human mania, ameliorated by antimania drugs lithium and valproate. Repeated social defeat stress resulted in repeated episodes of dramatic behavioral changes from hyperactivity to "depression-like" behavior, suggestive of some aspects of human bipolar disorder. Overall, we suggest that this Ank-G cKO mouse model recapitulates some of the core features of human bipolar disorder and indicates that cortical microcircuitry alterations during adulthood may be involved in pathogenesis. The model may be useful for studying disease pathophysiology and for developing experimental therapeutics.


Assuntos
Anquirinas/genética , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/genética , Prosencéfalo/fisiopatologia , Sinapses/patologia , Animais , Transtorno Bipolar/fisiopatologia , Modelos Animais de Doenças , Neurônios GABAérgicos/patologia , Lítio/farmacologia , Metilfenidato/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Proto-Oncogênicas c-fyn/biossíntese , Ácido Valproico/farmacologia , Canais de Sódio Disparados por Voltagem/genética
5.
Epigenetics ; 10(7): 581-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25985232

RESUMO

Methyl-Seq was recently developed as a targeted approach to assess DNA methylation (DNAm) at a genome-wide level in human. We adapted it for mouse and sought to examine DNAm differences across liver and 2 brain regions: cortex and hippocampus. A custom hybridization array was designed to isolate 99 Mb of CpG islands, shores, shelves, and regulatory elements in the mouse genome. This was followed by bisulfite conversion and sequencing on the Illumina HiSeq2000. The majority of differentially methylated cytosines (DMCs) were present at greater than expected frequency in introns, intergenic regions, near CpG islands, and transcriptional enhancers. Liver-specific enhancers were observed to be methylated in cortex, while cortex specific enhancers were methylated in the liver. Interestingly, commonly shared enhancers were differentially methylated between the liver and cortex. Gene ontology and pathway analysis showed that genes that were hypomethylated in the cortex and hippocampus were enriched for neuronal components and neuronal function. In contrast, genes that were hypomethylated in the liver were enriched for cellular components important for liver function. Bisulfite-pyrosequencing validation of 75 DMCs from 19 different loci showed a correlation of r = 0.87 with Methyl-Seq data. We also identified genes involved in neurodevelopment that were not previously reported to be differentially methylated across brain regions. This platform constitutes a valuable tool for future genome-wide studies involving mouse models of disease.


Assuntos
Encéfalo/crescimento & desenvolvimento , Metilação de DNA , Genoma , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Animais , Encéfalo/metabolismo , Ilhas de CpG , Citidina/análogos & derivados , Citidina/química , Elementos Facilitadores Genéticos , Córtex Entorrinal/química , Córtex Entorrinal/metabolismo , Hipocampo/química , Hipocampo/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Sulfitos/química
6.
Biochem Biophys Res Commun ; 420(3): 570-5, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22445894

RESUMO

Glucocorticoids may play a significant role in the etiology of neuropsychiatric illnesses. Abnormalities in plasma cortisol levels, glucocorticoid sensitivity, and HPA-axis function often accompany clinical symptoms of stress-related illnesses such as PTSD and depression. Of particular interest are genetic association studies that link single nucleotide polymorphisms of HPA-axis genes with illnesses only in the context of an early-life trauma exposure such as child abuse. These studies suggest that dysregulation of HPA-axis function can have lasting repercussions in shaping mood and anxiety, long after termination of the traumatic experience. As persistent glucocorticoid-induced loss of DNA methylation in FK506 binding protein 5 (Fkbp5) was previously observed in the hippocampus and blood and in the neuronal cell line HT-22, we asked whether these epigenetic alterations occur in non-neuronal, HPA-axis relevant cells. We used the pituitary adenoma cell line AtT-20 to demonstrate that the intronic enhancer region of Fkbp5 undergoes loss of DNA methylation in response to dexamethasone treatment in a dose-dependent manner. We also focused on the mouse hippocampal dentate gyrus to test whether these changes would be enriched in a region implicated in the HPA-axis stress response, neurogenesis, and synaptic plasticity. We observed an increase in enrichment of DNA methylation loss in the dentate gyrus, as compared to whole hippocampal tissues that were similarly treated with glucocorticoids. We then asked whether DNA methyltransferase 1 (Dnmt1), a methyltransferase enzyme involved in maintaining DNA methylation following cell division, is involved in the observed epigenetic alterations. We found a dose-dependent decrease of Dnmt1 expression in the AtT-20 cells following dexamethasone treatment, and a similar decrease in corticosterone-treated mouse hippocampus. Taken together, we provide evidence that these glucocorticoid-induced epigenetic alterations have a broader validity in non-neuronal cells and that they may involve the DNA methylation machinery.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/fisiologia , Giro Denteado/metabolismo , Epigênese Genética , Glucocorticoides/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Animais , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA/efeitos dos fármacos , Giro Denteado/efeitos dos fármacos , Dexametasona/farmacologia , Elementos Facilitadores Genéticos , Glucocorticoides/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Nat Med ; 18(1): 153-8, 2011 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-22179319

RESUMO

Huntington's disease is a fatal neurodegenerative disorder caused by an expanded polyglutamine repeat in huntingtin (HTT) protein. We previously showed that calorie restriction ameliorated Huntington's disease pathogenesis and slowed disease progression in mice that model Huntington's disease (Huntington's disease mice). We now report that overexpression of sirtuin 1 (Sirt1), a mediator of the beneficial metabolic effects of calorie restriction, protects neurons against mutant HTT toxicity, whereas reduction of Sirt1 exacerbates mutant HTT toxicity. Overexpression of Sirt1 improves motor function, reduces brain atrophy and attenuates mutant-HTT-mediated metabolic abnormalities in Huntington's disease mice. Further mechanistic studies suggested that Sirt1 prevents the mutant-HTT-induced decline in brain-derived neurotrophic factor (BDNF) concentrations and the signaling of its receptor, TrkB, and restores dopamine- and cAMP-regulated phosphoprotein, 32 kDa (DARPP32) concentrations in the striatum. Sirt1 deacetylase activity is required for Sirt1-mediated neuroprotection in Huntington's disease cell models. Notably, we show that mutant HTT interacts with Sirt1 and inhibits Sirt1 deacetylase activity, which results in hyperacetylation of Sirt1 substrates such as forkhead box O3A (Foxo3a), thereby inhibiting its pro-survival function. Overexpression of Sirt1 counteracts the mutant-HTT-induced deacetylase deficit, enhances the deacetylation of Foxo3a and facilitates cell survival. These findings show a neuroprotective role for Sirt1 in mammalian Huntington's disease models and open new avenues for the development of neuroprotective strategies in Huntington's disease.


Assuntos
Encéfalo/metabolismo , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Sirtuína 1/metabolismo , Animais , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Ratos , Receptor trkB/metabolismo , Transdução de Sinais , Sirtuína 1/genética
8.
J Neurosci ; 29(1): 179-90, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19129396

RESUMO

Previous studies have suggested that neuropeptide Y (NPY) in the dorsomedial hypothalamus (DMH) serves as an important signaling peptide in the regulation of energy balance. To elucidate such actions, we used the adenoassociated virus (AAV) system to alter Npy gene expression in the DMH and examined the effects of these alterations on food intake and energy balance as well as explored its downstream signaling pathway. We found that AAV-mediated overexpression of NPY in the DMH of lean rats increased food intake and body weight, and exacerbated high-fat diet-induced obesity. Knockdown of NPY expression in the DMH via AAV-mediated RNA interference ameliorated the hyperphagia, obesity, and diabetes of Otsuka Long-Evans Tokushima Fatty (OLETF) rats. NPY knockdown in the DMH produced a nocturnal and meal size-specific feeding effect. Moreover, we found that knockdown of DMH NPY expression in intact rats reduced NPY content in the nucleus of the solitary tract (NTS) and the dorsal motor nucleus of the vagus and affected within-meal satiation. DMH NPY knockdown increased the feeding inhibitory and NTS c-Fos responses to peripheral administration of cholecystokinin. Together, these results indicate that DMH NPY plays an important role in modulating food intake and energy balance and its dysregulation causes disordered energy balance leading to obesity.


Assuntos
Núcleo Hipotalâmico Dorsomedial/metabolismo , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Neuropeptídeo Y/metabolismo , Análise de Variância , Animais , Peso Corporal/fisiologia , Linhagem Celular Transformada , Colecistocinina/farmacologia , Dependovirus/fisiologia , Núcleo Hipotalâmico Dorsomedial/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Regulação da Expressão Gênica/fisiologia , Teste de Tolerância a Glucose/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Interferência de RNA/fisiologia , Ratos , Ratos Endogâmicos OLETF/metabolismo , Ratos Sprague-Dawley , Núcleo Solitário/metabolismo , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 105(7): 2325-30, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18287083

RESUMO

Caloric restriction (CR) is the most potent intervention known to both protect against carcinogenesis and extend lifespan in laboratory animals. A variety of anticarcinogens and CR mimetics induce and activate the NF-E2-related factor 2 (Nrf2) pathway. Nrf2, in turn, induces a number of antioxidative and carcinogen-detoxifying enzymes. Thus, Nrf2 offers a promising target for anticarcinogenesis and antiaging interventions. We used Nrf2-disrupted (KO) mice to examine its role on the biological effects of CR. Here, we show that Nrf2 is responsible for most of the anticarcinogenic effects of CR, but is dispensable for increased insulin sensitivity and lifespan extension. Nrf2-deficient mice developed tumors more readily in response to carcinogen exposure than did WT mice, and CR was ineffective in suppressing tumors in the KO mice. However, CR extended lifespan and increased insulin sensitivity similarly in KO and WT mice. These findings identify a molecular pathway that dissociates the prolongevity and anticarcinogenic effects of CR.


Assuntos
Restrição Calórica , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Animais , Regulação da Expressão Gênica , Insulina/metabolismo , Longevidade/fisiologia , Camundongos , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona) , NADPH Desidrogenase/genética , NADPH Desidrogenase/metabolismo , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Neoplasias/genética , Sensibilidade e Especificidade , Taxa de Sobrevida
10.
Appetite ; 49(2): 368-75, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17619065

RESUMO

Curt Richter's seminal contributions to the field of psychobiology continue to enlighten us today. His approaches to scientific questions, his ability to design ways of allowing his research subjects (usually rats) to answer his questions and his unique style of data presentation inform current investigations into the biological bases of behavior. One of Richter's earliest interests was identifying the causes of rhythmic spontaneous activity. Richter demonstrated an important relationship between bouts of activity and food intake in the laboratory rat. In his search for a causal link, he invoked contemporary thinking about the relationships between gastric contractions and hunger. Although his idea for a causal role for stomach contractions in eliciting spontaneous activity and food intake was mistaken, this early work identified themes for many of Richter's subsequent contributions and his behavioral observations continue to be relevant for current investigations.


Assuntos
Ingestão de Alimentos/fisiologia , Atividade Motora/fisiologia , Animais , Relógios Biológicos , Motilidade Gastrointestinal , História do Século XX , Ratos
11.
Neurosci Lett ; 403(1-2): 125-30, 2006 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-16687211

RESUMO

Stress response is an important neuroendocrine function. Overt or prolonged stress hormone secretion can lead to various disease states. The hippocampus plays an important role in the negative feedback onto the major player in the stress response, the hypothalamo-pituitary-adrenal axis. The transcription factor c-Fos is activated in the hippocampus following a number of stressors, including restraint stress. To determine whether c-fos modulates stress response, we previously generated mutant mice carrying a hippocampal mutation of the c-fos gene. In the current study, we found that female mutant mice display lower anxiety-like behavior than female wild-type mice in the elevated plus maze, whereas male mice are apparently normal. While both male and female mutant mice exhibit normal diurnal glucocorticoid (CORT) production and normal responses to acute restraint stress, female mutant mice habituated faster than female wild-type mice in response to chronic restraint stress. These findings suggest that hippocampal c-fos plays a role in gender-dependent response to stress.


Assuntos
Ansiedade/psicologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Estresse Psicológico/psicologia , Animais , Ansiedade/genética , Ritmo Circadiano , Ciclo Estral , Feminino , Habituação Psicofisiológica , Hipocampo/metabolismo , Hidrocortisona/sangue , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Mutantes , Proteínas Proto-Oncogênicas c-fos/genética , Restrição Física , Fatores Sexuais , Estresse Psicológico/metabolismo
12.
Brain Res ; 960(1-2): 252-8, 2003 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-12505679

RESUMO

Tachykinins are a family of neuropeptides that inhibit salt appetite. Although decreased tachykinin-mRNA levels are observed in natriorexic sodium-deplete rats, no decrease is seen in natriorexic sodium-replete rats that are administered the aldosterone-mimetic deoxycorticosterone acetate (DOCA). Since reduced synthesis of tachykinins could not account for increased appetite, we hypothesized that increased salt appetite was due to a downregulation of tachykinin receptors. Thus, we injected rats with DOCA once daily for 11 days and analyzed tachykinin receptor subtype, neurokinin 3 (NK3r)-immunoreactivity by Western blot analysis since intracerebroventricular injection of senktide (NK3r agonist) attenuates salt intake in DOCA-treated animals. We examined NK3r-immunoreactivity in several brain regions thought to be associated with the control of water and electrolyte balance including the bed nucleus of the stria terminalis, central nucleus of the amygdala, diagonal band of Broca, hippocampus, nucleus tractus solitarius, parabrachial nucleus, paraventricular nucleus of the hypothalamus, and supraoptic nucleus. Consistent with our hypothesis, we found decreased NK3r-immunoreactivity in all brain regions analyzed except for increases in the amygdala and no changes in the paraventricular nucleus of the hypothalamus. To examine whether DOCA's effects on NK3r synthesis are direct, we used differentiated N1E-115 neuroblastoma cells that express NK3r and treated them with a range of concentrations of DOCA and found a dose-dependent decrease in NK3r-mRNA abundance via Northern blotting. The present results suggest that the tachykinin receptors are downregulated after subchronic DOCA treatment and this finding is consistent with the hypothesis that suppressed inhibition of salt appetite as mediated through the tachykininergic system.


Assuntos
Apetite , Desoxicorticosterona/farmacologia , RNA Mensageiro/biossíntese , Receptores da Neurocinina-3/metabolismo , Sódio na Dieta/farmacologia , Animais , Autorradiografia , Northern Blotting , Western Blotting , Neoplasias Encefálicas/metabolismo , Membrana Celular/metabolismo , Depressão Química , Eletroforese em Gel de Poliacrilamida , Humanos , Masculino , Camundongos , Neuroblastoma/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-3/biossíntese , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA