Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 15481, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34326416

RESUMO

For many species, parental care critically affects offspring survival. But what drives animals to display parental behaviours towards young? In mammals, pregnancy-induced physiological transformations seem key in preparing the neural circuits that lead towards attraction (and reduced-aggression) to young. Beyond mammalian maternal behaviour, knowledge of the neural mechanisms that underlie young-directed parental care is severely lacking. We took advantage of a domesticated bird species, the Japanese quail, for which parental behaviour towards chicks can be induced in virgin non-reproductive adults through a sensitization procedure, a process that is not effective in all animals. We used the variation in parental responses to study neural transcriptomic changes associated with the sensitization procedure itself and with the outcome of the procedure (i.e., presence of parental behaviours). We found differences in gene expression in the hypothalamus and bed nucleus of the stria terminalis, but not the nucleus taeniae. Two genes identified are of particular interest. One is neurotensin, previously only demonstrated to be causally associated with maternal care in mammals. The other one is urocortin 3, causally demonstrated to affect young-directed neglect and aggression in mammals. Because our studies were conducted in animals that were reproductively quiescent, our results reflect core neural changes that may be associated with avian young-directed care independently of extensive hormonal stimulation. Our work opens new avenues of research into understanding the neural basis of parental care in non-placental species.


Assuntos
Comportamento Animal , Encéfalo/metabolismo , Coturnix/metabolismo , Hipotálamo/metabolismo , Transcriptoma , Animais , Mapeamento Cromossômico , Coturnix/fisiologia , Feminino , Perfilação da Expressão Gênica , Genômica , Masculino , Comportamento Materno , Neurotensina/metabolismo , Reprodução , Comportamento Social , Urocortinas/metabolismo
2.
Neuropeptides ; 80: 101994, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31740068

RESUMO

Neuropeptide Y (NPY) producing neurons in the arcuate nucleus (Arc) of the hypothalamus are essential to the regulation of food intake and energy homeostasis. Whilst they have classically been thought to co-express agouti-related peptide (AgRP), it is now clear that there is a sub-population of NPY neurons in the Arc that do not. Here, we show that a subset of AgRP-negative, NPY-positive neurons in the Arc also express neurotensin (NTS) and we use an NTS-Cre line to investigate the function of this sub-population of NPY neurons. The lack of NPY in NTS-positive neurons led to a marked reduction in fat mass and bodyweight as well as a significant reduction in food intake in male NPYlox/lox; NTScre/+ mice compared to controls. Despite the reduction in food intake, overall energy expenditure was similar between genotypes due to concomitant reduction in activity in NPYlox/lox; NTScre/+ mice. Furthermore, cortical bone mass was significantly reduced in NPYlox/lox;NTScre/+ mice with no evident alterations in the cancellous bone compartment, likely due to reduced leptin levels as a result of their reduced adiposity. Taken together, these data suggest that the sub-population of Arc NPY neurons expressing NTS are critical for regulating food intake, activity and fat mass but are not directly involved in the control of bone mass.


Assuntos
Peso Corporal/fisiologia , Metabolismo Energético/fisiologia , Neurônios/metabolismo , Neuropeptídeo Y/deficiência , Neurotensina/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Homeostase/fisiologia , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos Transgênicos , Neuropeptídeo Y/metabolismo , Fenótipo
3.
Neuron ; 103(2): 323-334.e7, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31178114

RESUMO

A crucial step in understanding the sleep-control mechanism is to identify sleep neurons. Through systematic anatomical screening followed by functional testing, we identified two sleep-promoting neuronal populations along a thalamo-amygdala pathway, both expressing neurotensin (NTS). Rabies-mediated monosynaptic retrograde tracing identified the central nucleus of amygdala (CeA) as a major source of GABAergic inputs to multiple wake-promoting populations; gene profiling revealed NTS as a prominent marker for these CeA neurons. Optogenetic activation and inactivation of NTS-expressing CeA neurons promoted and suppressed non-REM (NREM) sleep, respectively, and optrode recording showed they are sleep active. Further tracing showed that CeA GABAergic NTS neurons are innervated by glutamatergic NTS neurons in a posterior thalamic region, which also promote NREM sleep. CRISPR/Cas9-mediated NTS knockdown in either the thalamic or CeA neurons greatly reduced their sleep-promoting effect. These results reveal a novel thalamo-amygdala circuit for sleep generation in which NTS signaling is essential for both the upstream glutamatergic and downstream GABAergic neurons.


Assuntos
Tonsila do Cerebelo/citologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Neurotensina/metabolismo , Sono/fisiologia , Tálamo/citologia , Potenciais de Ação/genética , Tonsila do Cerebelo/fisiologia , Animais , Caspase 9/metabolismo , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Vias Neurais/metabolismo , Neurotensina/genética , Técnicas de Patch-Clamp , Sono/genética , Privação do Sono/fisiopatologia , Tálamo/fisiologia , Transfecção , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
4.
PLoS One ; 13(5): e0197817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29791497

RESUMO

Xenin, a highly conserved 25 amino acid peptide cleaved from the N-terminus of the coatomer protein alpha (COPA), is emerging as a food intake regulator in mammals and birds. To date, no research has been conducted on xenin biology in fish. This study aims to identify the copa mRNA encoding xenin in goldfish (Carassius auratus) as a model, to elucidate its regulation by feeding, and to describe the role of xenin on appetite. First, a partial sequence of copa cDNA, a region encoding xenin, was identified from goldfish brain. This sequence is highly conserved among both vertebrates and invertebrates. RT-qPCR revealed that copa mRNAs are widely distributed in goldfish tissues, with the highest levels detected in the brain, gill, pituitary and J-loop. Immunohistochemistry confirmed also the presence of COPA peptide in the hypothalamus and enteroendocrine cells on the J-loop mucosa. In line with its anorexigenic effects, we found important periprandial fluctuations in copa mRNA expression in the hypothalamus, which were mainly characterized by a gradually decrease in copa mRNA levels as the feeding time was approached, and a gradual increase after feeding. Additionally, fasting differently modulated the expression of copa mRNA in a tissue-dependent manner. Peripheral and central injections of xenin reduce food intake in goldfish. This research provides the first report of xenin in fish, and shows that this peptide is a novel anorexigen in goldfish.


Assuntos
Carpa Dourada/metabolismo , Neurotensina/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Ingestão de Alimentos , Células Enteroendócrinas/metabolismo , Feminino , Hipotálamo/metabolismo , Fígado/metabolismo , Masculino , Neurotensina/classificação , Neurotensina/genética , Filogenia , RNA Mensageiro/metabolismo
5.
Folia Histochem Cytobiol ; 56(1): 49-58, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29516445

RESUMO

INTRODUCTION: The distribution of the immunoreactive cell bodies and fibers containing neurotensin in the alpaca diencephalon was determined by an immunohistochemical technique. MATERIAL AND METHODS: The study was carried out in four male alpacas that lived at sea level. Brains of deeply anesthetized animals were fixed by perfusion with 4% paraformaldehyde. Cryostat sections were stained by a standard immunohistochemical method. RESULTS: Cell bodies containing neurotensin were observed in the zona incerta and hypothalamus. A low/moderate density of these cell bodies was observed in the lateral hypothalamic area, anterior and dorsal hypothalamic areas, suprachiasmatic nucleus, periventricular region of the hypothalamus and in the ventromedial hypothalamic nucleus. In both thalamus and hypothalamus, immunoreactive fibers showed a widespread distribution. In the thalamus, a high density of these fibers was mainly found in the midline nuclei, whereas in the hypothalamus a high density was in general observed in the whole structure. CONCLUSIONS: In comparison with other mammals, the thalamus of the alpaca showed the most widespread distribution of neurotensin-immunoreactive fibers. The widespread distribution of neurotensin through the alpaca diencephalon suggests that the peptide can be involved in many physiological actions.


Assuntos
Camelídeos Americanos , Diencéfalo/metabolismo , Neurotensina/metabolismo , Animais , Corpo Celular/química , Corpo Celular/metabolismo , Diencéfalo/química , Hipotálamo/química , Hipotálamo/metabolismo , Imuno-Histoquímica , Masculino , Neurotensina/química
6.
Brain Res ; 1659: 148-155, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28130052

RESUMO

Methamphetamine (METH) is a widely abused psychostimulant displaying potent addictive and neurotoxic properties. METH induces neurotoxicity of dopaminergic terminals and striatal neurons in the striatum. Despite much information on neurotransmitters, the role of neuropeptides is poorly understood. In this study, we investigated the role of the neuropeptide neurotensin on the METH-induced apoptosis of some striatal neurons in mice. We observed that a single injection of METH (30mg/kg, ip) induced the loss of approximately 15% of striatal neurons. An agonist of the neurotensin receptor 1 (PD149163, ip at various doses) attenuated the METH-induced striatal neuron apoptosis. Utilizing quantitative real time PCR, we showed that METH also up-regulated neurotensin gene expression with 96% increase in preproneurotensin mRNA levels in the striatum as compared to the control. Additionally, NTR1 agonist (ip injection) attenuated hyperthermia at 2h post-METH injection; hyperthermia is a putative and significant component of METH-induced neurotoxicity. To investigate the role of neurotensin without affecting core body temperature, we performed stereotactic injection of PD149163 into the striatum and observed that this compound maintained attenuated the METH-induced apoptosis in the striatum, while leaving core body temperature unaffected. There was no effect of NTR1 agonist on METH-induced dopamine terminal degeneration, as evidenced by tyrosine hydroxylase levels determined by Western blot. These data indicate that the neuropeptide neurotensin modulates the striatal neuronal apoptosis induced by METH through diverse mechanisms that need to be investigated. Furthermore, due to its neuroprotective properties, neurotensin receptor agonists show potential as drug candidates for the treatment of METH abuse and some neurological disorders.


Assuntos
Apoptose/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Metanfetamina/toxicidade , Neurotensina/análogos & derivados , Neurotransmissores/farmacologia , Receptores de Neurotensina/agonistas , Animais , Apoptose/fisiologia , Estimulantes do Sistema Nervoso Central/toxicidade , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Febre/induzido quimicamente , Febre/tratamento farmacológico , Febre/metabolismo , Febre/patologia , Masculino , Camundongos Endogâmicos ICR , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Neurotensina/metabolismo , Neurotensina/farmacologia , Precursores de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptores de Neurotensina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Cancer Lett ; 388: 73-84, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27914862

RESUMO

Hepatocellular carcinoma (HCC) is the third leading cause of death from cancer due to the combination of late diagnosis and a lack of curative treatments. The identification of factors which promote tumor aggressiveness, and those that predict treatment responses, are a means to optimize the management of HCC patients. The complex of Neurotensin (NTS) and its high affinity receptor (NTSR1) has been shown to induce tumor growth and metastasis process in various cancers. In this paper, we propose that NTS and NTSR1 can assist in the management of HCC. Concomitant expression of NTS/NTSR1 was correlated with poor prognosis and found in 50% of HCC patients. We show that NTSR1 expression was positively correlated with the alteration of the Wnt/ß-catenin pathway. Its activation creates EGFR driver activation which consequently enhances tumor progression, and sensitizes HCC tumor cells to TKI, such as sorafenib. The NTS/NTSR1 complex is a potential drug target for HCC, because it is an upstream regulator in the chain of cellular events involved in HCC progression. It could also be used as a theranostic biomarker for sorafenib to improve the HCC patient management.


Assuntos
Antineoplásicos/uso terapêutico , Receptores ErbB/genética , Cloridrato de Erlotinib/uso terapêutico , Neurotensina/metabolismo , Niacinamida/análogos & derivados , Compostos de Fenilureia/uso terapêutico , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/patologia , Progressão da Doença , Cloridrato de Erlotinib/farmacologia , Humanos , Neoplasias Hepáticas/patologia , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Compostos de Fenilureia/farmacologia , Prognóstico , Sorafenibe , Transfecção
9.
Behav Brain Res ; 312: 118-26, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27316340

RESUMO

Xenin is a gut hormone that reduces food intake by partly acting through the hypothalamus via neurotensin receptor 1 (Ntsr1). However, specific signaling pathways that mediate xenin-induced feeding suppression are not fully understood. Activation of Ntsr1 leads to the activation of the extracellular signal-regulated kinase (ERK). Hypothalamic ERK participates in the regulation of food intake by mediating the effect of hormonal signals. Therefore, we hypothesized that the anorectic effect of xenin is mediated by hypothalamic ERK signaling. To address this hypothesis, we compared levels of phosphorylation of ERK1/2 (pERK1/2) in the hypothalamus of both control and xenin-treated mice. The effect of xenin on ERK1/2 phosphorylation was also examined in mouse hypothalamic neuronal cell lines with or without Ntsr1. We also examined the effect of the blockade of central ERK signaling on xenin-induced feeding suppression in mice. The intraperitoneal (i.p.) injection of xenin caused a significant increase in the number of pERK1/2-immunoreactive cells in the hypothalamic arcuate nucleus. The majority of pERK1/2-positive cells expressed neuronal nuclei (NeuN), a marker for neurons. Xenin treatment increased pERK1/2 levels in one cell line expressing Ntsr1 but not another line without Ntsr1 expression. Both i.p. and intracerebroventricular (i.c.v.) injections of xenin reduced food intake in mice. The i.c.v. pre-treatment with U0126, a selective inhibitor of ERK1/2 upstream kinases, did not affect xenin-induced reduction in food intake. These findings suggest that although xenin activates ERK signaling in subpopulations of hypothalamic neurons, xenin does not require the activation of hypothalamic ERK signaling pathway to elicit feeding suppression.


Assuntos
Ingestão de Alimentos , Hipotálamo/metabolismo , Sistema de Sinalização das MAP Quinases , Neurotensina/metabolismo , Animais , Linhagem Celular , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotensina/administração & dosagem , Fosforilação
10.
Zhen Ci Yan Jiu ; 41(1): 35-9, 50, 2016 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-27141618

RESUMO

OBJECTIVE: To observe The effect of electroacupuncture (EA) stimulation of "Zusanli" (ST 36) and "Taichong" (LR 3) on intestinal motor and neurotensin (NT) levels in the plasma, hypothalamus, and gastro-antrum tissues in functional dyspepsia (FD) rats so as to reveal its mechanisms underlying improvement of FD. METHODS: Forty-eight SD rats were randomly divided into control, model and EA groups, with 16 rats in each group. The FD model was established by clamping the rats' tails and alternate day's feeding according to the related references. EA (2 Hz/100 Hz, 2 mA) was applied to unilateral ST 36 and LR 3 for 30 min, once daily for 14 days. Rats of the control group were only restricted. The gastric emptying rate and propulsive rate of the small intestine were detected. The content of NT in the plasma was assayed using ELISA, and the immunoactivity levels of NT in the hypothalamus, gastric antrum mucous membrane and ileum tissues were detected using immunohistochemistry. RESULTS: Compared with the control group, the gastric emptying rate and propulsive rate of the small intestine were considerably lowered in the model group (P < 0.01), and the content and immunoactivity levels of NT in the plasma, hypothalamus, mucous membrane of the gastric antrum and ileum tissues were significantly increased (P < 0.05). After EA intervention, the decreased gastric emptying rate and intestinal propulsive rate, as well as the increased NT content and immunoactivity levels of plasma, hypothalamus, gastric antrum and ileum were reversed (P < 0.05). CONCLUSION: EA intervention can obviously promote gastrointestinal motor in FD rats, which may be related to its function in down-regulating NT levels in the plasma, hypothalamus, gastric antrum and ileum. It suggests an involvement of NT in the brain-gut axis in EA-induced improvement of FD.


Assuntos
Dispepsia/terapia , Eletroacupuntura , Neurotensina/genética , Pontos de Acupuntura , Animais , Encéfalo/metabolismo , Dispepsia/genética , Dispepsia/metabolismo , Dispepsia/fisiopatologia , Feminino , Esvaziamento Gástrico , Mucosa Gástrica/metabolismo , Humanos , Hipotálamo/metabolismo , Masculino , Neurotensina/metabolismo , Antro Pilórico/metabolismo , Ratos , Ratos Sprague-Dawley , Estômago/fisiopatologia
11.
J Comp Neurol ; 523(5): 831-48, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25421126

RESUMO

Neurotensin (NTS) is a 13 amino acid neuropeptide that is expressed in the hypothalamus. In mammals, NTS-producing neurons that express leptin receptor (LepRb) regulate the function of hypocretin/orexin (HCRT) and dopamine neurons. Thus, the hypothalamic leptin-NTS-HCRT neuronal network orchestrates key homeostatic output, including sleep, feeding, and reward. However, the intricate mechanisms of the circuitry and the unique role of NTS-expressing neurons remain unclear. We studied the NTS neuronal networks in zebrafish and cloned the genes encoding the NTS neuropeptide and receptor (NTSR). Similar to mammals, the ligand is expressed primarily in the hypothalamus, while the receptor is expressed widely throughout the brain in zebrafish. A portion of hypothalamic nts-expressing neurons are inhibitory and some coexpress leptin receptor (lepR1). As in mammals, NTS and HCRT neurons are localized adjacently in the hypothalamus. To track the development and axonal projection of NTS neurons, the NTS promoter was isolated. Transgenesis and double labeling of NTS and HCRT neurons showed that NTS axons project toward HCRT neurons, some of which express ntsr. Moreover, another target of NTS neurons is ntsr-expressing dopaminergeric neurons. These findings suggest structural circuitry between leptin, NTS, and hypocretinergic or dopaminergic neurons and establish the zebrafish as a model to study the role of these neuronal circuits in the regulation of feeding, sleep, and reward.


Assuntos
Hipotálamo/anatomia & histologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leptina/metabolismo , Neuropeptídeos/metabolismo , Neurotensina/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/anatomia & histologia , Animais , Animais Geneticamente Modificados , Dopamina/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Vias Neurais/anatomia & histologia , Vias Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Orexinas , Receptores para Leptina/metabolismo , Receptores de Neurotensina/metabolismo , Peixe-Zebra/metabolismo
12.
J Endocrinol ; 221(2): 193-200, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24520141

RESUMO

Xenin-25, a peptide co-secreted with the incretin hormone glucose-dependent insulinotropic polypeptide (GIP), possesses promising therapeutic actions for obesity-diabetes. However, native xenin-25 is rapidly degraded by serum enzymes to yield the truncated metabolites: xenin 9-25, xenin 11-25, xenin 14-25 and xenin 18-25. This study has examined the biological activities of these fragment peptides. In vitro studies using BRIN-BD11 cells demonstrated that native xenin-25 and xenin 18-25 possessed significant (P<0.05 to P<0.001) insulin-releasing actions at 5.6 and 16.7 mM glucose, respectively, but not at 1.1  mM glucose. In addition, xenin 18-25 significantly (P<0.05) potentiated the insulin-releasing action of the stable GIP mimetic (D-Ala²)GIP. In contrast, xenin 9-25, xenin 11-25 and xenin 14-25 displayed neither insulinotropic nor GIP-potentiating actions. Moreover, xenin 9-25, xenin 11-25 and xenin 14-25 significantly (P<0.05 to P<0.001) inhibited xenin-25 (10⁻6 M)-induced insulin release in vitro. I.p. administration of xenin-based peptides in combination with glucose to high fat-fed mice did not significantly affect the glycaemic excursion or glucose-induced insulin release compared with controls. However, when combined with (D-Ala²)GIP, all xenin peptides significantly (P<0.01 to P<0.001) reduced the overall glycaemic excursion, albeit to a similar extent as (D-Ala²)GIP alone. Xenin-25 and xenin 18-25 also imparted a potential synergistic effect on (D-Ala²)GIP-induced insulin release in high fat-fed mice. All xenin-based peptides lacked significant satiety effects in normal mice. These data demonstrate that the C-terminally derived fragment peptide of xenin-25, xenin 18-25, exhibits significant biological actions that could have therapeutic utility for obesity-diabetes.


Assuntos
Neurotensina/metabolismo , Neurotensina/farmacologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Avaliação Pré-Clínica de Medicamentos , Hipoglicemiantes/química , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Secreção de Insulina , Masculino , Camundongos , Neurotensina/química , Proteólise , Saciação/efeitos dos fármacos
13.
Food Chem ; 147: 152-9, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24206699

RESUMO

In this paper the oil from seeds of Amaranthus cruentus L. (AmO) was shown to be an efficient modulator of the physical chemical properties of artificial lipid and rat hepatocyte plasma membranes. AmO improved the membrane stability, their stress resistance and the adsorption of neurotensin to plasma membranes with the distinct biphasic interactions being observed even after adrenalin stress exposure. The analysis of pro-/antioxidant balance in rat blood revealed a mild prooxidant activity after AmO intake, which was accompanied by accumulation of oxidative destruction products in plasma membranes. This prooxidant action of AmO was corroborated in vitro in an adrenalin autooxidation model. On the other hand, the observed improved resistance to adrenalin stress in AmO supplemented rats was associated with an antioxidant response in blood and plasma membrane studies. The AmO effects can be attributed to the modulation of the metabolic pathways involved into oxygen and free radical homeostasis.


Assuntos
Amaranthus/química , Antioxidantes/farmacologia , Membrana Celular/efeitos dos fármacos , Epinefrina/metabolismo , Hepatócitos/metabolismo , Lipídeos/química , Estresse Oxidativo/efeitos dos fármacos , Óleos de Plantas/farmacologia , Animais , Membrana Celular/metabolismo , Hepatócitos/efeitos dos fármacos , Bicamadas Lipídicas/metabolismo , Masculino , Neurotensina/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
14.
J Neurosci ; 33(18): 7618-26, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23637156

RESUMO

The lateral hypothalamus (LH) sends a dense glutamatergic and peptidergic projection to dopamine neurons in the ventral tegmental area (VTA), a cell group known to promote reinforcement and aspects of reward. The role of the LH to VTA projection in reward-seeking behavior can be informed by using optogenetic techniques to dissociate the actions of LH neurons from those of other descending forebrain inputs to the VTA. In the present study, we identify the effect of neurotensin (NT), one of the most abundant peptides in the LH to VTA projection, on excitatory synaptic transmission in the VTA and reward-seeking behavior. Mice displayed robust intracranial self-stimulation of LH to VTA fibers, an operant behavior mediated by NT 1 receptors (Nts1) and NMDA receptors. Whole-cell patch-clamp recordings of VTA dopamine neurons demonstrated that NT (10 nm) potentiated NMDA-mediated EPSCs via Nts1. Results suggest that NT release from the LH into the VTA activates Nts1, thereby potentiating NMDA-mediated EPSCs and promoting reward. The striking behavioral and electrophysiological effects of NT and glutamate highlight the LH to VTA pathway as an important component of reward.


Assuntos
Condicionamento Operante/fisiologia , Ácido Glutâmico/metabolismo , Hipotálamo/fisiologia , Neurotensina/metabolismo , Recompensa , Área Tegmentar Ventral/fisiologia , Animais , Proteínas de Bactérias/genética , Channelrhodopsins , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Hipotálamo/efeitos dos fármacos , Técnicas In Vitro , Luz , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Vias Neurais/fisiologia , Neurotensina/farmacologia , Pirazóis/farmacologia , Quinolinas/farmacologia , Quinoxalinas/farmacologia , Receptores de Neurotensina/antagonistas & inibidores , Receptores de Neurotensina/deficiência , Autoestimulação , Transdução de Sinais/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo , Valina/análogos & derivados , Valina/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos
15.
PLoS One ; 8(4): e61616, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23626705

RESUMO

Ciliary neurotrophic factor (CNTF) induces neurogenesis, reduces feeding, and induces weight loss. However, the central mechanisms by which CNTF acts are vague. We employed the mHypoE-20/2 line that endogenously expresses the CNTF receptor to examine the direct effects of CNTF on mRNA levels of urocortin-1, urocortin-2, agouti-related peptide, brain-derived neurotrophic factor, and neurotensin. We found that treatment of 10 ng/ml CNTF significantly increased only urocortin-1 mRNA by 1.84-fold at 48 h. We then performed intracerebroventricular injections of 0.5 mg/mL CNTF into mice, and examined its effects on urocortin-1 neurons post-exposure. Through double-label immunohistochemistry using specific antibodies against c-Fos and urocortin-1, we showed that central CNTF administration significantly activated urocortin-1 neurons in specific areas of the hypothalamus. Taken together, our studies point to a potential role for CNTF in regulating hypothalamic urocortin-1-expressing neurons to mediate its recognized effects on energy homeostasis, neuronal proliferaton/survival, and/or neurogenesis.


Assuntos
Depressores do Apetite/farmacologia , Fator Neurotrófico Ciliar/farmacologia , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Urocortinas/genética , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Apetite/efeitos dos fármacos , Apetite/fisiologia , Depressores do Apetite/metabolismo , Regulação do Apetite/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linhagem Celular , Fator Neurotrófico Ciliar/metabolismo , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/citologia , Hipotálamo/metabolismo , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/metabolismo , Neurotensina/genética , Neurotensina/metabolismo , Receptor do Fator Neutrófico Ciliar/genética , Receptor do Fator Neutrófico Ciliar/metabolismo , Urocortinas/agonistas , Urocortinas/metabolismo
16.
PLoS One ; 8(4): e62391, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23620827

RESUMO

Both orexin and neurotensin are expressed in the lateral hypothalamic area (LHA) and have been implicated in the regulation of feeding, motor activity and the reward system. A double label immunofluorescence and in situ hybridization studies showed that neurotensin colocalizes with orexin in neurons of the LHA. Pharmacological studies suggested that neurotensin excites orexin-producing neurons (orexin neurons) through activation of neurotensin receptor-2 (NTSR-2) and non-selective cation channels. In situ hybridization study showed that most orexin neurons express neurotensin receptor-2 mRNA but not neurotensin receptor-1 (Ntsr-1) mRNA. Immunohistochemical studies showed that neurotensin-immunoreactive fibers make appositions to orexin neurons. A neurotensin receptor antagonist decreased Fos expression in orexin neurons and wakefulness time in wild type mice when administered intraperitoneally. However, the antagonist did not evoke any effect on these parameters in orexin neuron-ablated mice. These observations suggest the importance of neurotensin in maintaining activity of orexin neurons. The evidence presented here expands our understanding of the regulatory mechanism of orexin neurons.


Assuntos
Hipotálamo/citologia , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Neurotensina/metabolismo , Sono , Vigília , Animais , Hipotálamo/efeitos dos fármacos , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurotensina/antagonistas & inibidores , Orexinas , Pirazóis/farmacologia , Quinolinas/farmacologia , Receptores de Neurotensina/metabolismo , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos
17.
Endocrinology ; 153(5): 2385-97, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22416082

RESUMO

Glucagon-like peptide-2 (GLP-2), a proglucagon-derived peptide, has been postulated to affect appetite at the level of the hypothalamus. To gain better insight into this process, a degradation-resistant GLP-2 analog, human (Gly(2))GLP-2(1-33) [h(Gly(2))GLP-2] was intracerebroventricularly injected into mice to examine its action on food and water intake and also activation of hypothalamic anorexigenic α-melanocyte-stimulating hormone/proopiomelanocortin, neurotensin, and orexigenic neuropeptide Y, and ghrelin neurons. Central h(Gly(2))GLP-2 administration significantly suppressed food and water intake with acute weight loss at 2 h. Further, central h(Gly(2))GLP-2 robustly induced c-Fos activation in the hypothalamic arcuate, dorsomedial, ventromedial, paraventricular, and the lateral hypothalamic nuclei. We found differential colocalization of neuropeptides with c-Fos in specific regions of the hypothalamus. To assess whether hypothalamic neuropeptides are directly regulated by GLP-2 in vitro, we used an adult-derived clonal, immortalized hypothalamic cell line, mHypoA-2/30, that endogenously expresses functional GLP-2 receptors (GLP-2R) and two of the feeding-related neuropeptides linked to GLP-2R activation in vivo: neurotensin and ghrelin. Treatment with h(Gly(2))GLP-2 stimulated c-Fos expression and phosphorylation of cAMP response element-binding protein/activating transcription factor-1. In addition, treatment with h(Gly(2))GLP-2 significantly increased neurotensin and ghrelin mRNA transcript levels by 50 and 95%, respectively, at 24 h after treatment in protein kinase A-dependent manner. Taken together, these findings implicate the protein kinase A pathway as the means by which GLP-2 can up-regulate hypothalamic neuropeptide mRNA levels and provide evidence for a link between central GLP-2R activation and specific hypothalamic neuropeptides involved in appetite regulation.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Líquidos/fisiologia , Ingestão de Alimentos/fisiologia , Grelina/genética , Grelina/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microinjeções , Neurônios/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Neurotensina/genética , Neurotensina/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
18.
Endocrinology ; 153(5): 2208-22, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22334721

RESUMO

Exendin-4, a long-acting glucagon-like peptide-1 receptor (GLP-1R) agonist, is a potential regulator of feeding behavior through its ability to inhibit gastric emptying, reduce food intake, and induce satiety. GLP-1R activation by exendin-4 induces anorexia; however, the specific populations of neuropeptidergic neurons activated by exendin-4 within the hypothalamus, the central regulator of energy homeostasis, remain unclear. This study determines whether exendin-4 regulates hypothalamic neuropeptide expression and explores the signaling mechanisms involved. The distribution and quantity of exendin-4-induced c-Fos immunoreactivity were evaluated to determine activation of α-melanocyte-stimulating hormone/proopiomelanocortin, neuropeptide Y, neurotensin (NT), and ghrelin neurons in hypothalamic nuclei during exendin-4-induced anorexia in mice. Additionally, exendin-4 action on NT and ghrelin transcript regulation was examined in immortalized hypothalamic neurons. With anorexia induced by intracerebroventricular exendin-4, α-melanocyte-stimulating hormone/proopiomelanocortin and neuropeptide Y neurons were activated in the arcuate nucleus, with simultaneous activation of NT-expressing neurons in the paraventricular nucleus, and ghrelin-expressing neurons in the arcuate nucleus, paraventricular nucleus, and periventricular hypothalamus, suggesting that neurons in one or more of these areas mediate the anorexic action of exendin-4. In the hypothalamic neuronal cell models, exendin-4 increased cAMP, cAMP response element-binding protein/activating transcription factor-1 and c-Fos activation, and via a protein kinase A-dependent mechanism regulated NT and ghrelin mRNA expression, indicating that these neuropeptides may serve as downstream mediators of exendin-4 action. These findings provide a previously unrecognized link between central GLP-1R activation by exendin-4 and the regulation of hypothalamic NT and ghrelin. Further understanding of this central GLP-1R activation may lead to safe and effective therapeutics for the treatment of metabolic disorders.


Assuntos
Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , Receptores de Glucagon/agonistas , Peçonhas/farmacologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Exenatida , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Neurotensina/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , alfa-MSH/metabolismo
19.
Schizophr Res ; 136(1-3): 88-95, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22104138

RESUMO

The neuropeptide neurotensin (NT) is closely associated with dopaminergic and glutamatergic systems in the rat brain. Central injection of NT into the nucleus accumbens (NAcc) or peripheral administration of NT receptor agonists, reduces many of the behavioral effects of psychostimulants. However, the role of endogenous NT in the behavioral effects of psychostimulants (e.g. DA agonists and NMDA receptor antagonists) remains unclear. Using a NTR antagonist, SR142948A, the current studies were designed to examine the role of endogenous NT in DA receptor agonist- and NMDA receptor antagonist-induced disruption of prepulse inhibition of the acoustic startle response (PPI), locomotor hyperactivity and brain-region specific c-fos mRNA expression. Adult male rats received a single i.p. injection of SR142948A or vehicle followed by D-amphetamine, apomorphine or dizocilpine challenge. SR142948A had no effect on baseline PPI, but dose-dependently attenuated d-amphetamine- and dizocilpine-induced PPI disruption and enhanced apomorphine-induced PPI disruption. SR142948A did not significantly affect either baseline locomotor activity or stimulant-induced hyperlocomotion. Systemic SR142948A administration prevented c-fos mRNA induction in mesolimbic terminal fields (prefrontal cortex, lateral septum, NAcc, ventral subiculum) induced by all three psychostimulants implicating the VTA as the site for NT modulation of stimulant-induced PPI disruption. Further characterization of the NT system may be valuable to find clinical useful compounds for schizophrenia and drug addiction.


Assuntos
Anfetamina/efeitos adversos , Apomorfina/efeitos adversos , Estimulantes do Sistema Nervoso Central/efeitos adversos , Transtornos Neurológicos da Marcha/induzido quimicamente , Neurotensina/metabolismo , Estimulação Acústica/métodos , Adamantano/efeitos adversos , Adamantano/análogos & derivados , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/efeitos adversos , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Desempenho Psicomotor/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neurotensina/antagonistas & inibidores , Reflexo de Sobressalto/efeitos dos fármacos
20.
Neurosci Lett ; 479(1): 18-21, 2010 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-20471454

RESUMO

Leptin signaling in the hypothalamus is required for normal food intake and body weight homeostasis. Recent evidence suggests that besides the signal transducer and activator of transcription-3 (STAT3) pathway, several non-STAT3 pathways mediate leptin signaling in the hypothalamus. We have previously demonstrated that leptin stimulates phosphodiesterase-3B (PDE3B) activity in the hypothalamus, and PDE3 inhibitor cilostamide reverses anorectic and body weight reducing effects of leptin. To establish the physiological role of PDE3B signaling in the hypothalamus, we examined if leptin signaling through the PDE3B pathway is responsible for the activation of proopiomelanocortin (POMC) and neurotensin (NT) neurons, which are known to play a critical role in energy homeostasis. To this end, we assessed the effect of cilostamide on leptin-induced POMC and NT gene expression in the rat hypothalamus. Results showed that while central injection of leptin significantly increased both POMC and NT mRNA levels in the medial basal hypothalamus, cilostamide completely reversed this effect of leptin suggesting a PDE3B-activation dependent induction of POMC and NT gene expression by leptin. This result further suggests that the PDE3B pathway plays an important role in mediating leptin action in the hypothalamus.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Neurotensina/metabolismo , Pró-Opiomelanocortina/metabolismo , Transdução de Sinais , Animais , Expressão Gênica , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Masculino , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurotensina/genética , Inibidores da Fosfodiesterase 3 , Inibidores de Fosfodiesterase/farmacologia , Pró-Opiomelanocortina/genética , Quinolonas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA