Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 942
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 34(1-2): 72-86, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31831627

RESUMO

Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ) are key effectors of the Hippo pathway to control cell growth and organ size, of which dysregulation yields to tumorigenesis or hypertrophy. Upon activation, YAP/TAZ translocate into the nucleus and bind to TEAD transcription factors to promote transcriptional programs for proliferation or cell specification. Immediate early genes, represented by AP-1 complex, are rapidly induced and control later-phase transcriptional program to play key roles in tumorigenesis and organ maintenance. Here, we report that YAP/TAZ directly promote FOS transcription that in turn contributes to the biological function of YAP/TAZ. YAP/TAZ bind to the promoter region of FOS to stimulate its transcription. Deletion of YAP/TAZ blocks the induction of immediate early genes in response to mitogenic stimuli. FOS induction contributes to expression of YAP/TAZ downstream target genes. Genetic deletion or chemical inhibition of AP-1 suppresses growth of YAP-driven cancer cells, such as Lats1/2-deficient cancer cells as well as Gαq/11 mutated uveal melanoma. Furthermore, AP-1 inhibition almost completely abrogates the hepatomegaly induced by YAP overexpression. Our findings reveal a feed-forward interplay between immediate early transcription of AP-1 and Hippo pathway function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Regulação Neoplásica da Expressão Gênica , Transativadores/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes fos/genética , Células HEK293 , Humanos , Fígado/metabolismo , Melanoma/fisiopatologia , Camundongos , Mitógenos/farmacologia , Tamanho do Órgão/genética , Regiões Promotoras Genéticas/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Neoplasias Uveais/fisiopatologia , Proteínas de Sinalização YAP
2.
Clin Exp Pharmacol Physiol ; 49(3): 341-349, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34729812

RESUMO

An imbalance of osteoclasts and osteoblasts can result in a variety of bone-related diseases, including osteoporosis. Thus, decreasing the activity of osteoclastic bone resorption is the main therapeutic method for treating osteoporosis. 2E-Decene-4, 6-diyn-1-ol-acetate (DDA) is a natural bioactive compound with anti-inflammatory and anti-cancer properties. However, its effects on osteoclastogenesis are unknown. Murine bone marrow-derived macrophages (BMMs) or RAW264.7 cells were treated with DDA, followed by evaluation of cell viability, RANKL-induced osteoclast differentiation, and pit formation assay. Effects of DDA on RANKL-induced phosphorylation of MAPKs were assayed by western blot analysis. Expression of osteoclast-specific genes was examined with reverse transcription-PCR (RT-PCR) and western blot analysis. In this study, DDA significantly inhibited RANKL-induced osteoclast differentiation in RAW264.7 cells as well as in BMMs without cytotoxicity. DDA also strongly blocked the resorbing capacity of BMM on calcium phosphate-coated plates. DDA inhibited RANKL-induced phosphorylation of ERK, JNK and p38 MAPKs, as well as expression of c-Fos and NFATc1, which are essential transcription factors for osteoclastogenesis. In addition, DDA decreased expression levels of osteoclastogenesis-specific genes, including matrix metalloproteinase-9 (MMP-9), tartrate-resistant acid phosphatase (TRAP), and receptor activator of NF-κB (RANK) in RANKL-induced RAW264.7 cells. Collectively, these findings indicated that DDA attenuates RANKL-induced osteoclast formation by suppressing the MAPKs-c-Fos-NFATc1 signalling pathway and osteoclast-specific genes. These results indicate that DDA may be a potential candidate for bone diseases associated with abnormal osteoclast formation and function.


Assuntos
Produtos Biológicos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genes fos/fisiologia , Macrófagos/efeitos dos fármacos , Fatores de Transcrição NFATC/metabolismo , Osteogênese/efeitos dos fármacos , Animais , Aster/química , Produtos Biológicos/química , Diferenciação Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos/genética , Camundongos , Fatores de Transcrição NFATC/genética , Osteoclastos , Ligante RANK/genética , Ligante RANK/metabolismo , Células RAW 264.7
3.
J Neurochem ; 157(3): 532-549, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33454999

RESUMO

Interleukin-6 (IL-6) is a cytokine primarily known for immune regulation. There is also growing evidence that IL-6 triggers neurogenesis and impacts neural development, both life-long occurring processes that can be impaired by early-life and adult stress. Stress induces the release of glucocorticoids by activation of the hypothalamic-pituitary-adrenal (HPA) axis. On the cellular level, glucocorticoids act via the ubiquitously expressed glucocorticoid receptor. Thus, we aimed to elucidate whether glucocorticoids affect IL-6-induced neural development. Here, we show that IL-6 signalling induces neurite outgrowth in adrenal pheochromocytoma PC12 cells in a mitogen-activated protein kinase (MAPK) pathway-dependent manner, since neurite outgrowth was diminished upon Mek-inhibitor treatment. Using quantitative biochemical approaches, such as qRT-PCR analysis of Hyper-IL-6 treated PC12 cells, we show that neurite outgrowth induced by IL-6 signalling is accompanied by early and transient MAPK-dependent mRNA expression of immediate early genes coding for proteins such as early growth response protein 1 (Egr1) and c-Fos. This correlates with reduced proliferation and prolonged G0/G1 cell cycle arrest as determined by monitoring the cellular DNA content using flow cytometry. These results indicate for IL-6 signalling-induced neural differentiation. Interestingly, the glucocorticoid Dexamethasone impairs early IL-6 signalling-induced mRNA expression of c-Fos and Egr1 and restrains neurite outgrowth. Impaired Egr1 and c-Fos expression in neural development is implicated in the aetiology of neuropathologies. Thus, it appears likely that stress-induced release of glucocorticoids, as well as therapeutically administered glucocorticoids, contribute to the development of neuropathologies by reducing the expression of Egr1 and c-Fos, and by restraining IL-6-dependent neural differentiation.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/genética , Genes fos/efeitos dos fármacos , Genes fos/genética , Glucocorticoides/farmacologia , Interleucina-6/antagonistas & inibidores , Neuritos/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/antagonistas & inibidores , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Células PC12 , Ratos
4.
Mol Cell ; 52(3): 314-24, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24207025

RESUMO

Lysine acetylation regulates transcription by targeting histones and nonhistone proteins. Here we report that the central regulator of transcription, RNA polymerase II, is subject to acetylation in mammalian cells. Acetylation occurs at eight lysines within the C-terminal domain (CTD) of the largest polymerase subunit and is mediated by p300/KAT3B. CTD acetylation is specifically enriched downstream of the transcription start sites of polymerase-occupied genes genome-wide, indicating a role in early stages of transcription initiation or elongation. Mutation of lysines or p300 inhibitor treatment causes the loss of epidermal growth-factor-induced expression of c-Fos and Egr2, immediate-early genes with promoter-proximally paused polymerases, but does not affect expression or polymerase occupancy at housekeeping genes. Our studies identify acetylation as a new modification of the mammalian RNA polymerase II required for the induction of growth factor response genes.


Assuntos
Histonas/genética , Lisina/genética , RNA Polimerase II/metabolismo , Transcrição Gênica , Acetilação , Animais , Proteína 2 de Resposta de Crescimento Precoce/biossíntese , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica , Genes fos/genética , Histonas/metabolismo , Humanos , Regiões Promotoras Genéticas , RNA Polimerase II/genética , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
5.
J Neurosci ; 39(6): 1077-1087, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30509960

RESUMO

Humans with alcohol use disorder typically abstain because of the negative consequences associated with excessive drinking, and exposure to contexts previously associated with alcohol use can trigger relapse. We used a rat model that captures a characteristic of this human condition: namely voluntary abstinence from alcohol use because of contingent punishment. There is substantial variability in the propensity to relapse following extended periods of abstinence, and this is a critical feature preventing the successful treatment of alcohol use disorder. Here we examined relapse following acute or prolonged abstinence. In male alcohol preferring P rats, we found an increased propensity to relapse in Context B, the punishment context after prolonged abstinence. Next, we found that neither alcohol intake history nor the motivational strength of alcohol predicted the propensity to relapse. We next examined the putative circuitry of context-induced relapse to alcohol seeking following prolonged abstinence using Fos as a marker of neuronal activation. The anterior insular cortex (AI) was the only brain region examined where Fos expression correlated with alcohol seeking behavior in Context B after prolonged abstinence. Finally, we used local infusion of GABAA and GABAB receptor agonists (muscimol + baclofen) to show a causal role of the AI in context-induced relapse in Context B, the punishment context after prolonged abstinence. Our results show that there is substantial individual variability in the propensity to relapse in the punishment-associated context after prolonged abstinence, and this is mediated by activity in the AI.SIGNIFICANCE STATEMENT A key feature of alcohol use disorder is that sufferers show an enduring propensity to relapse throughout their lifetime. Relapse typically occurs despite the knowledge of adverse consequences including health complications or relationship breakdowns. Here we use a recently developed rodent model that recapitulates this behavior. After an extended period of abstinence, relapse propensity is markedly increased in the "adverse consequence" environment, akin to humans with alcohol use disorder relapsing in the face of adversity. From a circuitry perspective, we demonstrate a causal role of the anterior insular cortex in relapse to alcohol seeking after extended abstinence following punishment imposed voluntary cessation of alcohol use.


Assuntos
Abstinência de Álcool/psicologia , Consumo de Bebidas Alcoólicas/psicologia , Córtex Cerebral/fisiologia , Comportamento de Procura de Droga/fisiologia , Punição/psicologia , Animais , Depressores do Sistema Nervoso Central/farmacologia , Córtex Cerebral/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Etanol/farmacologia , Agonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B/farmacologia , Genes fos/genética , Masculino , Motivação , Ratos , Receptores de GABA-A/efeitos dos fármacos , Recidiva
6.
Cell Mol Neurobiol ; 40(8): 1395-1404, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32162199

RESUMO

In the present work, using in situ hybridization, we studied the expression patterns of three molluscan homologs of vertebrate immediate-early genes C/EBP, c-Fos, and c-Jun in the central nervous system (CNS) of terrestrial gastropod snail Helix. The molluscan C/EBP gene was described in literature, while c-Fos and c-Jun were studied in terrestrial snails for the first time. Localization of the expression was traced in normal conditions, and in preparations physiologically activated using stimulation of suboesophageal ganglia nerves. No expression was detected constitutively. In stimulated preparations, all three genes had individual expression patterns in Helix CNS, and the level of expression was stimulus-dependent. The number of cells expressing the gene of interest was different from the number of cells projecting to the stimulated nerve, and thus activated retrogradely. This difference depended on the ganglia studied. At the subcellular level, the labeled RNA was observed as dots (probably small clusters of RNA molecules) and shapeless mass of RNA, often seen as a circle at the internal border of the cell nuclei. The data provide a basis for further study of behavioral role of these putative immediate-early genes in snail behavior and learning.


Assuntos
Sistema Nervoso Central/metabolismo , Genes Precoces/genética , Neurônios/metabolismo , Caramujos/genética , Animais , Genes Precoces/fisiologia , Genes fos/genética , Caracois Helix/fisiologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , RNA/metabolismo
7.
FASEB J ; 33(4): 5389-5398, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30702925

RESUMO

With an increasing body of evidence regarding GPCR oligomerization and its clinical implications over the last decade, the modulation and dynamics of GPCR homo- and hetero-oligomers has more recently become an area of intense research focus. Previously, our lab showed in vitro heteromer formation between angiotensin II receptor type 1 subtype a (AT1aR) and secretin receptor (SCTR), which is involved in in vivo control of hyperosmolality-induced water drinking behavior. Because the secretin (SCT)/SCTR axis is crucial to the central actions of angiotensin II (ANGII) and both SCT and ANGII are capable of triggering vasopressin (Vp) release from hypothalamus, we investigated here the in vivo role of SCTR-AT1aR heteromer in regulating Vp release in hypothalamus using transmembrane peptides as tools. We showed that SCTR-AT1aR heteromer mediates stimulatory actions of both SCT and ANGII in hypothalamic Vp expression and release as well as neuronal activities via the immediate early gene cFos. The results from this study not only are consistent with our hypothesis that SCT and ANGII interact at the receptor level to mediate their water homeostatic activities but also provide evidence for in vivo functions of cross-class GPCR heteromers.-Mak, S. O. K., Zhang, L., Chow, B. K. C. In vivo actions of SCTR/AT1aR heteromer in controlling Vp expression and release via cFos/cAMP/CREB pathway in magnocellular neurons of PVN.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/fisiologia , Angiotensina II/metabolismo , Animais , Genes fos/genética , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Secretina/metabolismo , Vasopressinas/metabolismo
8.
Mol Biol Rep ; 46(5): 5425-5432, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31364017

RESUMO

Obesity is a complex disorder that is influenced by genetic and environmental factors. DNA methylation is an epigenetic mechanism that is involved in development of obesity and its metabolic complications. The aim of this study was to investigate the association between the RANKL and c-Fos gene methylation on obesity with body mass index (BMI), lipid parameters, homeostasis model assessment of insulin resistance (HOMA-IR), plasma leptin, adiponectin and resistin levels. The study included 68 obese and 46 non-obese subjects. Anthropometric parameters, including body weight, body mass index, waist circumference, and waist-hip ratio, were assessed. Serum glucose, triglycerides (TG), total cholesterol, high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C), plasma leptin, adiponectin and resistin levels were measured. Methylation status of RANKL and c-Fos gen were evaluated by MS-HRM. Statistically significant differences were observed between obese patients and the controls with respect to RANKL and c-Fos gene methylation status (p < 0.001). Also, statistically significant importance was observed RANKL gene methylation and increased level of leptin in obese subjects (p = 0.0081). At the same time, statistically significant association between methylation of c-Fos and increased level of adiponectin was observed in obese patients (p = 0.03) On the other hand, decreased level of resistin was observed where the c-Fos was unmetyladed in controls (p = 0.01). We conclude that methylation of RANKL and c-Fos genes have significant influences on obesity and adipokine levels. Based on literature this was the first study which shows the interactions between RANKL and c-Fos methylation and obesity.


Assuntos
Epigênese Genética/genética , Regulação da Expressão Gênica/genética , Obesidade/genética , Adiponectina/análise , Adiponectina/sangue , Adulto , Antropometria , Índice de Massa Corporal , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Metilação de DNA/genética , Feminino , Genes fos/genética , Humanos , Resistência à Insulina/genética , Leptina/análise , Leptina/sangue , Masculino , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/genética , Triglicerídeos/sangue
9.
J Biol Chem ; 292(52): 21282-21290, 2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29150442

RESUMO

Epithelioid hemangioma is a locally aggressive vascular neoplasm, found in bones and soft tissue, whose cause is currently unknown, but may involve oncogene activation. FOS is one of the earliest viral oncogenes to be characterized, and normal cellular FOS forms part of the activator protein 1 (AP-1) transcription factor complex, which plays a pivotal role in cell growth, differentiation, and survival as well as the DNA damage response. Despite this, a causal link between aberrant FOS function and naturally occurring tumors has not yet been established. Here, we describe a thorough molecular and biochemical analysis of a mutant FOS protein we identified in these vascular tumors. The mutant protein lacks a highly conserved helix consisting of the C-terminal four amino acids of FOS, which we show is indispensable for fast, ubiquitin-independent FOS degradation via the 20S proteasome. Our work reveals that FOS stimulates endothelial sprouting and that perturbation of normal FOS degradation could account for the abnormal vessel growth typical of epithelioid hemangioma. To the best of our knowledge, this is the first functional characterization of mutant FOS proteins found in tumors.


Assuntos
Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/fisiologia , Neoplasias Vasculares/genética , Indutores da Angiogênese , Carcinogênese/genética , Carcinogênese/metabolismo , Diferenciação Celular , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes fos/genética , Hemangioma/genética , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Elementos Reguladores de Transcrição/genética , Neoplasias Vasculares/metabolismo
10.
Eur J Neurosci ; 48(11): 3319-3334, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30346078

RESUMO

Mammalian circadian rhythms are entrained by photic stimuli that are relayed by retinal projections to the core of the suprachiasmatic nucleus (SCN). Neuronal activation, as demonstrated by expression of the immediate early gene c-fos, leads to transcription of the core clock gene per1. The duper mutation in hamsters shortens circadian period and amplifies light-induced phase shifts. We performed two experiments to compare the number of c-FOS immunoreactive (ir) and PER1-ir cells, and the intensity of staining, in the SCN of wild-type (WT) and duper hamsters at various intervals after presentation of a 15-min light pulse in the early subjective night. Light-induced c-FOS-ir within 1 hr in the dorsocaudal SCN of duper, but not WT hamsters. In cells that express vasoactive intestinal peptide (VIP), which plays a critical role in synchronization of SCN cellular oscillators, light-induced c-FOS-ir was greater in duper than WT hamsters. After the light pulse, PER1-ir cells were found in more medial portions of the SCN than FOS-ir, and appeared with a longer latency and over a longer time course, in VIP cells of duper than wild-type hamsters. Our results indicate that the duper allele alters SCN function in ways that may contribute to changes in free running period and phase resetting.


Assuntos
Ritmo Circadiano/fisiologia , Atividade Motora/fisiologia , Núcleo Supraquiasmático/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Cricetinae , Genes fos/genética , Imuno-Histoquímica/métodos , Masculino , Mutação/genética , Neurônios/metabolismo , Proteínas Circadianas Period/metabolismo , Estimulação Luminosa , Proteínas Proto-Oncogênicas c-fos/metabolismo
11.
J Sleep Res ; 26(3): 386-393, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28318068

RESUMO

Natural helicid (4-formylphenyl-O-ß-d-allopyranoside), a main active constituent from seeds of the Chinese herb Helicia nilagirica, has been reported to exert a sedative, analgesic and hypnotic effect, and is used clinically to treat neurasthenic syndrome, vascular headaches and trigeminal neuralgia. In the current study, mechanical allodynia tests, electroencephalograms, electromyogram recordings and c-Fos expression in neuropathic pain-like model mice of partial sciatic nerve ligation were used to investigate the effect of helicid on neuropathic pain and co-morbid insomnia. Our results showed that helicid at a dose of 100, 200 or 400 mg kg-1 could increase the mechanical threshold by 2.5-, 2.8- and 3.1-fold for 3 h after administration, respectively. Helicid at 200 and 400 mg kg-1 given at 07:00 hours increased the amount of non-rapid eye movement sleep in a 3-h period by 1.27- and 1.35-fold in partial sciatic nerve ligated mice. However, helicid (400 mg kg-1 ) given at 21:00 hours did not change the sleep pattern in normal mice. Immunohistochemical study showed that helicid (400 mg kg-1 ) administration could reverse the increase of c-Fos expression in the neurons of the rostral anterior cingulate cortex and tuberomammillary nucleus, and the decrease of c-Fos expression in the ventrolateral preoptic area caused by partial sciatic nerve ligation. These results indicate that helicid is an effective agent for both neuropathic pain and sleep disturbances in partial sciatic nerve ligated mice.


Assuntos
Benzaldeídos/uso terapêutico , Neuralgia/complicações , Neuralgia/tratamento farmacológico , Distúrbios do Início e da Manutenção do Sono/etiologia , Distúrbios do Início e da Manutenção do Sono/prevenção & controle , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Benzaldeídos/administração & dosagem , Benzaldeídos/farmacologia , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Genes fos/genética , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Hiperalgesia/diagnóstico , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/farmacologia , Hipnóticos e Sedativos/uso terapêutico , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/metabolismo , Ligadura , Masculino , Camundongos , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Nervo Isquiático , Sono/efeitos dos fármacos , Sono/fisiologia
12.
Alcohol Clin Exp Res ; 41(9): 1574-1583, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28753742

RESUMO

BACKGROUND: Chronic exposure to stress or alcohol can drive neuroadaptations that alter cognition. Alterations in cognition may contribute to alcohol use disorders by reducing cognitive control over drinking and maintenance of abstinence. Here we examined effects of combined ethanol (EtOH) and stress exposure on prefrontal cortex (PFC)-dependent cognition. METHODS: Adult male C57BL/6J mice were trained to drink EtOH (15%, v/v) on a 1 h/d 1-bottle schedule. Once stable, mice were exposed to cycles of chronic intermittent EtOH (CIE) or air-control vapor exposure (Air), followed by test cycles of 1 h/d EtOH drinking. During test drinking, mice received no stress (NS) or 10 minutes of forced swim stress (FSS) 4 hours before each test. This schedule produced 4 experimental groups: control, Air/NS; EtOH-dependent no stress, CIE/NS; nondependent stress, Air/FSS; or EtOH-dependent stress, CIE/FSS. After 2 cycles of CIE and FSS exposure, we assessed PFC-dependent cognition using object/context recognition and attentional set shifting. At the end of the study, mice were perfused and brains were collected for measurement of c-Fos activity in PFC and locus coeruleus (LC). RESULTS: CIE/FSS mice escalated EtOH intake faster than CIE/NS and consumed more EtOH than Air/NS across all test cycles. After 2 cycles of CIE/FSS, mice showed impairments in contextual learning and extradimensional set-shifting relative to other groups. In addition to cognitive dysfunction, CIE/FSS mice demonstrated widespread reductions in c-Fos activity within prelimbic and infralimbic PFC as well as LC. CONCLUSIONS: Together, these findings show that interactions between EtOH and stress exposure rapidly lead to disruptions in signaling across cognitive networks and impairments in PFC-dependent cognitive function.


Assuntos
Alcoolismo/psicologia , Depressores do Sistema Nervoso Central/toxicidade , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/psicologia , Etanol/toxicidade , Estresse Psicológico/psicologia , Animais , Atenção/efeitos dos fármacos , Disfunção Cognitiva/complicações , Genes fos/genética , Aprendizagem/efeitos dos fármacos , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/fisiopatologia , Estresse Psicológico/complicações , Natação/psicologia
13.
Nature ; 470(7333): 221-6, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21307935

RESUMO

Electrical stimulation of certain hypothalamic regions in cats and rodents can elicit attack behaviour, but the exact location of relevant cells within these regions, their requirement for naturally occurring aggression and their relationship to mating circuits have not been clear. Genetic methods for neural circuit manipulation in mice provide a potentially powerful approach to this problem, but brain-stimulation-evoked aggression has never been demonstrated in this species. Here we show that optogenetic, but not electrical, stimulation of neurons in the ventromedial hypothalamus, ventrolateral subdivision (VMHvl) causes male mice to attack both females and inanimate objects, as well as males. Pharmacogenetic silencing of VMHvl reversibly inhibits inter-male aggression. Immediate early gene analysis and single unit recordings from VMHvl during social interactions reveal overlapping but distinct neuronal subpopulations involved in fighting and mating. Neurons activated during attack are inhibited during mating, suggesting a potential neural substrate for competition between these opponent social behaviours.


Assuntos
Agressão/fisiologia , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Estimulação Elétrica , Eletrofisiologia , Feminino , Regulação da Expressão Gênica , Genes fos/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Inibição Neural/genética , Inibição Neural/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Comportamento Sexual Animal/fisiologia , Núcleo Hipotalâmico Ventromedial/anatomia & histologia , Núcleo Hipotalâmico Ventromedial/metabolismo
14.
Biol Pharm Bull ; 40(11): 1963-1967, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29093345

RESUMO

Cytosolic prostaglandin (PG) E synthase (cPGES/p23) plays a role in the biosynthesis of PGE2 and in the molecular chaperone machinery. Studies of knockout mice lacking cPGES/p23 have demonstrated that cPGES/p23 is essential in fetal mouse development. A cDNA microarray analysis revealed that a lack of cPGES/p23 decreases the expression of some immediate early genes, such as c-fos and activating transcription factor 3 (ATF3). Here we report the involvement of cPGES/p23 in c-Fos expression. A stable knockdown of cPGES/p23 in cultured fibroblasts not only reduced serum-induced c-Fos expression, but also decreased the phosphorylation of extracellular signal regulated kinase (ERK). These results suggest that cPGES/p23 is involved in the activation of ERK to promote c-Fos expression.


Assuntos
Genes fos/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Chaperonas Moleculares/metabolismo , Prostaglandina-E Sintases/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Animais , Linhagem Celular , Citosol/metabolismo , Dinoprostona/metabolismo , Fibroblastos , Técnicas de Silenciamento de Genes , Análise em Microsséries , Chaperonas Moleculares/genética , Fosforilação , Proteínas Proto-Oncogênicas c-fos/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real
15.
Genes Dev ; 23(23): 2765-77, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19952111

RESUMO

Transcription elongation factor DSIF/Spt4-Spt5 is capable of promoting and inhibiting RNA polymerase II elongation and is involved in the expression of various genes. While it has been known for many years that DSIF inhibits elongation in collaboration with the negative elongation factor NELF, how DSIF promotes elongation is largely unknown. Here, an activity-based biochemical approach was taken to understand the mechanism of elongation activation by DSIF. We show that the Paf1 complex (Paf1C) and Tat-SF1, two factors implicated previously in elongation control, collaborate with DSIF to facilitate efficient elongation. In human cells, these factors are recruited to the FOS gene in a temporally coordinated manner and contribute to its high-level expression. We also show that elongation activation by these factors depends on P-TEFb-mediated phosphorylation of the Spt5 C-terminal region. A clear conclusion emerging from this study is that a set of elongation factors plays nonredundant, cooperative roles in elongation. This study also shows unambiguously that Paf1C, which is generally thought to have chromatin-related functions, is involve directlyd in elongation control.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas Nucleares/metabolismo , RNA Polimerase II/metabolismo , Fatores de Transcrição/metabolismo , Genes fos/genética , Células HeLa , Humanos , Mutação , Fatores de Transcrição/genética , Fatores de Transcrição/isolamento & purificação , Fatores de Elongação da Transcrição
16.
J Neurosci ; 35(50): 16443-9, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26674869

RESUMO

Ischemia induces extensive temporal changes in cerebral transcriptome that influences the neurologic outcome after stroke. In addition to protein-coding RNAs, many classes of noncoding RNAs, including long noncoding RNAs (LncRNAs), also undergo changes in the poststroke brain. We currently evaluated the functional significance of an LncRNA called Fos downstream transcript (FosDT) that is cogenic with Fos gene. Following transient middle cerebral artery occlusion (MCAO) in adult rats, expression of FosDT and Fos was induced. FosDT knockdown significantly ameliorated the postischemic motor deficits and reduced the infarct volume. Focal ischemia also increased FosDT binding to chromatin-modifying proteins (CMPs) Sin3a and coREST (corepressors of the transcription factor REST). Furthermore, FosDT knockdown derepressed REST-downstream genes GRIA2, NFκB2, and GRIN1 in the postischemic brain. Thus, FosDT induction and its interactions with REST-associated CMPs, and the resulting regulation of REST-downstream genes might modulate ischemic brain damage. LncRNAs, such as FosDT, can be therapeutically targeted to minimize poststroke brain damage. SIGNIFICANCE STATEMENT: Mammalian brain is abundantly enriched with long noncoding RNAs (LncRNAs). Functional roles of LncRNAs in normal and pathological states are not yet understood. This study identified that LncRNA FosDT induced after transient focal ischemia modulates poststroke behavioral deficits and brain damage. These effects of FosDT in part are due to its interactions with chromatin-modifying proteins Sin3a and coREST (corepressors of the transcription factor REST) and subsequent derepression of REST-downstream genes GRIA2, NFκB2, and GRIN1. Therefore, LncRNA-mediated epigenetic remodeling could determine stroke outcome.


Assuntos
Isquemia Encefálica/genética , Genes fos/genética , RNA Longo não Codificante/genética , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Acidente Vascular Cerebral/genética , Animais , Isquemia Encefálica/complicações , Proteínas Correpressoras/biossíntese , Proteínas Correpressoras/genética , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Ataque Isquêmico Transitório/genética , Ataque Isquêmico Transitório/patologia , Masculino , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/genética , Desempenho Psicomotor , Ratos , Ratos Endogâmicos SHR , Complexo Correpressor Histona Desacetilase e Sin3
17.
Biochim Biophys Acta ; 1853(3): 539-48, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25523140

RESUMO

Amino acid (AA) deprivation in mammalian cells activates a collection of signaling cascades known as the AA response (AAR), which is characterized by transcriptional induction of stress-related genes, including FBJ murine osteosarcoma viral oncogene homolog (cFOS). The present study established that the signaling mechanism underlying the AA-dependent transcriptional regulation of the cFOS gene in HepG2 human hepatocellular carcinoma cells is independent of the classic GCN2-eIF2-ATF4 pathway. Instead, a RAS-RAF-MEK-ERK cascade mediates AAR signaling to the cFOS gene. Increased cFOS transcription is observed from 4-24 h after AAR-activation, exhibiting little or no overlap with the rapid and transient increase triggered by the well-known serum response. Furthermore, serum is not required for the AA-responsiveness of the cFOS gene and no phosphorylation of promoter-bound serum response factor (SRF) is observed. The ERK-phosphorylated transcription factor E-twenty six-like (p-ELK1) is increased in its association with the cFOS promoter after activation of the AAR. This research identified cFOS as a target of the AAR and further highlights the importance of AA-responsive MAPK signaling in HepG2 cells.


Assuntos
Aminoácidos/deficiência , Carcinoma Hepatocelular/genética , Genes fos/genética , Neoplasias Hepáticas/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Fator 4 Ativador da Transcrição/fisiologia , Aminoácidos/farmacologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Células Cultivadas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas Serina-Treonina Quinases/fisiologia , Ativação Transcricional/efeitos dos fármacos
18.
Nature ; 465(7295): 182-7, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20393465

RESUMO

We used genome-wide sequencing methods to study stimulus-dependent enhancer function in mouse cortical neurons. We identified approximately 12,000 neuronal activity-regulated enhancers that are bound by the general transcriptional co-activator CBP in an activity-dependent manner. A function of CBP at enhancers may be to recruit RNA polymerase II (RNAPII), as we also observed activity-regulated RNAPII binding to thousands of enhancers. Notably, RNAPII at enhancers transcribes bi-directionally a novel class of enhancer RNAs (eRNAs) within enhancer domains defined by the presence of histone H3 monomethylated at lysine 4. The level of eRNA expression at neuronal enhancers positively correlates with the level of messenger RNA synthesis at nearby genes, suggesting that eRNA synthesis occurs specifically at enhancers that are actively engaged in promoting mRNA synthesis. These findings reveal that a widespread mechanism of enhancer activation involves RNAPII binding and eRNA synthesis.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Neurônios/metabolismo , Transcrição Gênica/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína de Ligação a CREB/metabolismo , Sequência Consenso/genética , Proteínas do Citoesqueleto/genética , Genes Reporter , Genes fos/genética , Histonas/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , RNA Polimerase II/metabolismo , RNA não Traduzido/biossíntese , RNA não Traduzido/genética
19.
Biol Pharm Bull ; 39(8): 1293-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27476939

RESUMO

To develop an effective oral delivery system for plasmid DNA (pDNA) using cationic liposomes, it is necessary to clarify the characteristics of uptake and transport of cationic liposome/pDNA complexes into the intestinal epithelium. In particular, evaluation of the involvement of an unstirred water layer (UWL), which is a considerable permeability barrier, in cationic liposome transport is very important. Here, we investigated the effects of a UWL on the transfection efficiency of cationic liposome/pDNA complexes into a Caco-2 cell monolayer. When Caco-2 cells were transfected with cationic liposome/pDNA complexes in shaking cultures to reduce the thickness of the UWL, gene expression was significantly higher in Caco-2 cells compared with static cultures. We also found that this enhancement of gene expression by shaking was not attributable to activation of transcription factors such as activator protein-1 and nuclear factor-kappaB (NF-κB). In addition, the increase in gene expression by mechanical agitation was observed at all charge ratios (1.5, 2.3, 3.1, 4.5) of cationic liposome/pDNA complexes. Transport experiments using Transwells demonstrated that mechanical agitation increased the uptake of cationic liposome/pDNA complexes by Caco-2 cells, whereas transport of the complexes across a Caco-2 cell monolayer did not occurr. Moreover, the augmentation of the gene expression of cationic liposome/pDNA complexes by shaking was observed in Madin-Darby canine kidney cells. These results indicate that a UWL greatly affects the uptake and transfection efficiency of cationic liposome/pDNA complexes into an epithelial monolayer in vitro.


Assuntos
DNA/administração & dosagem , Lipossomos/administração & dosagem , Transfecção/métodos , Animais , Células CACO-2 , Cátions , Cães , Expressão Gênica , Genes fos/genética , Humanos , Células Madin Darby de Rim Canino , Plasmídeos , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/metabolismo , Fator de Transcrição RelA/metabolismo , Água
20.
Am J Med Genet B Neuropsychiatr Genet ; 171(6): 827-38, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27090961

RESUMO

Despite moderate heritability estimates, the molecular architecture of aggressive behavior remains poorly characterized. This study compared gene expression profiles from a genetic mouse model of aggression with zebrafish, an animal model traditionally used to study aggression. A meta-analytic, cross-species approach was used to identify genomic variants associated with aggressive behavior. The Rankprod algorithm was used to evaluated mRNA differences from prefrontal cortex tissues of three sets of mouse lines (N = 18) selectively bred for low and high aggressive behavior (SAL/LAL, TA/TNA, and NC900/NC100). The same approach was used to evaluate mRNA differences in zebrafish (N = 12) exposed to aggressive or non-aggressive social encounters. Results were compared to uncover genes consistently implicated in aggression across both studies. Seventy-six genes were differentially expressed (PFP < 0.05) in aggressive compared to non-aggressive mice. Seventy genes were differentially expressed in zebrafish exposed to a fight encounter compared to isolated zebrafish. Seven genes (Fos, Dusp1, Hdac4, Ier2, Bdnf, Btg2, and Nr4a1) were differentially expressed across both species 5 of which belonging to a gene-network centred on the c-Fos gene hub. Network analysis revealed an association with the MAPK signaling cascade. In human studies HDAC4 haploinsufficiency is a key genetic mechanism associated with brachydactyly mental retardation syndrome (BDMR), which is associated with aggressive behaviors. Moreover, the HDAC4 receptor is a drug target for valproic acid, which is being employed as an effective pharmacological treatment for aggressive behavior in geriatric, psychiatric, and brain-injury patients. © 2016 Wiley Periodicals, Inc.


Assuntos
Agressão/fisiologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes/genética , Genes fos/genética , Genes fos/fisiologia , Camundongos , Comportamento Social , Transcriptoma/genética , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA