Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Brain ; 14(1): 157, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34641940

RESUMO

AIM: Growth hormone secretagogue receptor 1a (GHS-R1a) is widely distributed in brain including the hippocampus. Studies have demonstrated the critical role of hippocampal ghrelin/GHS-R1a signaling in synaptic physiology, memory and cognitive dysfunction associated with Alzheimer's disease (AD). However, current reports are inconsistent, and the mechanism underlying memory modulation of GHS-R1a signaling is uncertain. In this study, we aim to investigate the direct impact of selective increase of GHS-R1a expression in dCA1 excitatory/inhibitory neurons on learning and memory. METHODS: Endogenous GHS-R1a distribution in dCA1 excitatory/inhibitory neurons was assessed by fluorescence in situ hybridization. Cre-dependent GHS-R1a overexpression in excitatory or inhibitory neurons was done by stereotaxic injection of aav-hSyn-DIO-hGhsr1a-2A-eGFP virus in dCA1 region of vGlut1-Cre or Dlx5/6-Cre mice respectively. Virus-mediated GHS-R1a upregulation in dCA1 neurons was confirmed by quantitative RT-PCR. Different behavioral paradigms were used to evaluate long-term memory performance. RESULTS: GHS-R1a is distributed both in dCA1 excitatory pyramidal neurons (αCaMKII+) and in inhibitory interneurons (GAD67+). Selective increase of GHS-R1a expression in dCA1 pyramidal neurons impaired spatial memory and object-place recognition memory. In contrast, selective increase of GHS-R1a expression in dCA1 interneurons enhanced long-term memory performance. Our findings reveal, for the first time, a neuronal type-specific role that hippocampal GHS-R1a signaling plays in regulating memory. Therefore, manipulating GHS-R1a expression/activity in different subpopulation of neurons may help to clarify current contradictory findings and to elucidate mechanism of memory control by ghrelin/GHS-R1a signaling, under both physiological and pathological conditions such as AD.


Assuntos
Região CA1 Hipocampal/citologia , Interneurônios/metabolismo , Memória/fisiologia , Células Piramidais/metabolismo , Receptores de Grelina/biossíntese , Animais , Grelina/fisiologia , Hibridização in Situ Fluorescente , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Receptores de Grelina/genética , Reconhecimento Psicológico , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Regulação para Cima
2.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34289060

RESUMO

Islets represent an important site of direct action of the hormone ghrelin, with expression of the ghrelin receptor (growth hormone secretagogue receptor; GHSR) having been localized variably to alpha cells, beta cells, and/or somatostatin (SST)-secreting delta cells. To our knowledge, GHSR expression by pancreatic polypeptide (PP)-expressing gamma cells has not been specifically investigated. Here, histochemical analyses of Ghsr-IRES-Cre × Cre-dependent ROSA26-yellow fluorescent protein (YFP) reporter mice showed 85% of GHSR-expressing islet cells coexpress PP, 50% coexpress SST, and 47% coexpress PP + SST. Analysis of single-cell transcriptomic data from mouse pancreas revealed 95% of Ghsr-expressing cells coexpress Ppy, 100% coexpress Sst, and 95% coexpress Ppy + Sst. This expression was restricted to gamma-cell and delta-cell clusters. Analysis of several single-cell human pancreatic transcriptome data sets revealed 59% of GHSR-expressing cells coexpress PPY, 95% coexpress SST, and 57% coexpress PPY + SST. This expression was prominent in delta-cell and beta-cell clusters, also occurring in other clusters including gamma cells and alpha cells. GHSR expression levels were upregulated by type 2 diabetes mellitus in beta cells. In mice, plasma PP positively correlated with fat mass and with plasma levels of the endogenous GHSR antagonist/inverse agonist LEAP2. Plasma PP also elevated on LEAP2 and synthetic GHSR antagonist administration. These data suggest that in addition to delta cells, beta cells, and alpha cells, PP-expressing pancreatic cells likely represent important direct targets for LEAP2 and/or ghrelin both in mice and humans.


Assuntos
Regulação da Expressão Gênica , Grelina/biossíntese , Polipeptídeo Pancreático/metabolismo , Receptores de Grelina/biossíntese , Animais , Proteínas de Bactérias/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genes Reporter , Células Secretoras de Glucagon/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Ligantes , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Pâncreas/metabolismo , Análise de Célula Única , Transcriptoma
3.
Physiol Res ; 67(2): 339-346, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29303606

RESUMO

Neurodegenerative disorders, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are increasing in prevalence. Currently, there are no effective and specific treatments for these disorders. Recently, positive effects of the orexigenic hormone ghrelin on memory and learning were demonstrated in mouse models of AD and PD. In this study, we tested the potential neuroprotective properties of a stable and long-lasting ghrelin analog, Dpr(3)ghrelin (Dpr(3)ghr), in SH-SY5Y neuroblastoma cells stressed with 1.2 mM methylglyoxal (MG), a toxic endogenous by-product of glycolysis, and we examined the impact of Dpr(3)ghr on apoptosis. Pre-treatment with both 10(-5) and 10(-7) M Dpr(3)ghr resulted in increased viability in SH-SY5Y cells (determined by MTT staining), as well as reduced cytotoxicity of MG in these cells (determined by LDH assay). Dpr(3)ghr increased viability by altering pro-apoptotic and viability markers: Bax was decreased, Bcl-2 was increased, and the Bax/Bcl-2 ratio was attenuated. The ghrelin receptor GHS-R1 and Dpr(3)ghr-induced activation of PBK/Akt were immuno-detected in SH-SY5Y cells to demonstrate the presence of GHS-R1 and GHS-R1 activation, respectively. We demonstrated that Dpr(3)ghr protected SH-SY5Y cells against MG-induced neurotoxicity and apoptosis. Our data suggest that stable ghrelin analogs may be candidates for the effective treatment of neurodegenerative disorders.


Assuntos
Apoptose/efeitos dos fármacos , Grelina/análogos & derivados , Grelina/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , Glicólise/efeitos dos fármacos , Humanos , L-Lactato Desidrogenase/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Síndromes Neurotóxicas/prevenção & controle , Aldeído Pirúvico/toxicidade , Receptores de Grelina/biossíntese
4.
Invest Ophthalmol Vis Sci ; 58(14): 6113-6122, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29222553

RESUMO

Purpose: Ghrelin, a natural ligand for the growth hormone secretagogue receptor type 1a (GHSR-1a), may protect retinal neurons against glaucomatous injury. We therefore characterized the underlying mechanism of the ghrelin/GHSR-1a-mediated neuroprotection with a rat chronic intraocular hypertension (COH) model. Methods: The rat COH model was produced by blocking episcleral veins. A combination of immunohistochemistry, Western blot, TUNEL assay, and retrograde labeling of retinal ganglion cells (RGCs) was used. Results: Elevation of intraocular pressure induced a significant increase in ghrelin and GHSR-1a expression in retinal cells, including RGCs and Müller cells. Western blot confirmed that the protein levels of ghrelin exhibited a transient upregulation at week 2 after surgery (G2w), while the GHSR-1a protein levels were maintained at high levels from G2w to G4w. In COH retinas, the ratio of LC3-II/LC-I and beclin1, two autophagy-related proteins, were increased from G1w to G4w, and the cleavage product of caspase3, an apoptotic executioner, was detected from G2w to G4w. Intraperitoneal injection of ghrelin significantly increased the number of surviving RGCs; inhibited the changes of LC3-II/LC-I, beclin1, and the cleavage products of caspase3; and reduced the number of TUNEL-positive cells in COH retinas. Ghrelin treatment also reversed the decreased levels of p-Akt and p-mTOR, upregulated GHSR-1a protein levels, and attenuated glial fibrillary acidic protein levels in COH retinas. Conclusions: All these results suggest that ghrelin may provide neuroprotective effect in COH retinas through activating ghrelin/GHSR-1a system, which was mediated by inhibiting retinal autophagy, ganglion cell apoptosis, and Müller cell gliosis.


Assuntos
Apoptose , Autofagia , Regulação da Expressão Gênica , Grelina/genética , Glaucoma/genética , Receptores de Grelina/genética , Células Ganglionares da Retina/patologia , Animais , Western Blotting , Modelos Animais de Doenças , Grelina/biossíntese , Glaucoma/metabolismo , Glaucoma/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Pressão Intraocular , Masculino , RNA/genética , Ratos , Ratos Sprague-Dawley , Receptores de Grelina/biossíntese , Células Ganglionares da Retina/metabolismo , Regulação para Cima
5.
Gene ; 578(2): 169-76, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26692148

RESUMO

Sexual size dimorphism has been observed in many cultivable fish species including yellow catfish, in which male fish grow much faster than female fish. Ghrelin is a potent stimulator of pituitary growth hormone (GH) release and known to potentially promote food intake and body weight gain. In order to investigate the molecular mechanism of sexual size dimorphism in yellow catfish (Pelteobagrus fulvidraco), ghrelin and its functional receptor, growth hormone secretagogue receptor (GHSR) cDNAs were cloned. Real-time PCR indicated that both ghrelin and GHSR were more highly expressed in hypothalamus and gut of male fish than female. During normal larval development, expression of ghrelin and GHSR genes was significantly higher in males than in females. 17a-Methyltestosterone (MT) treatment enhanced the expression of ghrelin in female larval fish and GHSR in both sexes, whereas the expression of ghrelin in male larval fish increased in the beginning, then decreased as the treatment time prolonged. Furthermore, the expression of ghrelin and GHSR in male juvenile was significantly increased compared with female juvenile, in short and long term fasting periods, suggesting that male fish may have a better appetite than female during fasting. Our results demonstrate that sex difference in the expression of ghrelin and GHSR may be involved in sexual size dimorphism by regulating feeding and GH/IGF signaling in yellow catfish.


Assuntos
Tamanho Corporal/genética , Grelina/biossíntese , Receptores de Grelina/biossíntese , Caracteres Sexuais , Animais , Peixes-Gato/genética , Peixes-Gato/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Grelina/genética , Masculino , Metiltestosterona/farmacologia , Receptores de Grelina/genética
6.
Alcohol Alcohol ; 51(2): 121-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26330568

RESUMO

AIMS: Ghrelin initially emerged as a gut-brain hormone controlling food intake, meal initiation and appetite mainly via hypothalamic circuits in both rodents and humans. The findings that ghrelin receptors (GHS-R1A) are expressed in reward-related areas, including the nucleus accumbens (NAc) and ventral tegmental area (VTA), suggest that ghrelin is a novel reward regulator. Indeed, ghrelin signalling mediates the rewarding and motivational properties of addictive drugs. In addition, daily co-administration of a GHS-R1A antagonist and various addictive drugs prevents the drug-induced locomotor sensitization in rats. METHODS: The present series of experiment were designed to evaluate the effect of repeated pharmacological GHS-R1A suppression on drug-induced locomotor stimulation in more detail. RESULTS: We showed that sub-chronic pre-treatment of the GHS-R1A antagonist, JMV2959, attenuated the ability of acute administration of alcohol as well as of amphetamine to stimulate locomotion. However, there was no effect of sub-chronic JMV2959 treatment on locomotor activity per se or on the expression of the GHS-R1A gene (Ghsr) in the VTA or the NAc compared with vehicle treatment. In addition, sub-chronic ghrelin treatment caused a locomotor sensitization. CONCLUSIONS: While previous research has pinpointed ghrelin as an appetite regulator the present study together with previous studies suggest that ghrelin signalling modulates various reward-mediated behaviours in rodents. Collectively, this suggests that the GHS-R1A could be a key target for novel treatment strategies for addiction.


Assuntos
Anfetamina/administração & dosagem , Etanol/administração & dosagem , Glicina/análogos & derivados , Locomoção/efeitos dos fármacos , Receptores de Grelina/antagonistas & inibidores , Triazóis/administração & dosagem , Animais , Estimulantes do Sistema Nervoso Central/administração & dosagem , Glicina/administração & dosagem , Locomoção/fisiologia , Masculino , Camundongos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de Grelina/biossíntese , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
7.
PLoS One ; 10(10): e0141043, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26506093

RESUMO

Ghrelin is a gut-brain peptide hormone, which binds to the growth hormone secretagogue receptor (GHS-R) to regulate a wide variety of biological processes in fish. Despite these prominent physiological roles, no studies have reported the anatomical distribution of preproghrelin transcripts using in situ hybridization in a non-mammalian vertebrate, and its mapping within the different encephalic areas remains unknown. Similarly, no information is available on the possible 24-h variations in the expression of preproghrelin and its receptor in any vertebrate species. The first aim of this study was to investigate the anatomical distribution of ghrelin and GHS-R1a ghrelin receptor subtype in brain and gastrointestinal tract of goldfish (Carassius auratus) using immunohistochemistry and in situ hybridization. Our second aim was to characterize possible daily variations of preproghrelin and ghs-r1 mRNA expression in central and peripheral tissues using real-time reverse transcription-quantitative PCR. Results show ghrelin expression and immunoreactivity in the gastrointestinal tract, with the most abundant signal observed in the mucosal epithelium. These are in agreement with previous findings on mucosal cells as the primary synthesizing site of ghrelin in goldfish. Ghrelin receptor was observed mainly in the hypothalamus with low expression in telencephalon, pineal and cerebellum, and in the same gastrointestinal areas as ghrelin. Daily rhythms in mRNA expression were found for preproghrelin and ghs-r1 in hypothalamus and pituitary with the acrophase occurring at nighttime. Preproghrelin, but not ghs-r1a, displayed a similar daily expression rhythm in the gastrointestinal tract with an amplitude 3-fold higher than the rest of tissues. Together, these results described for the first time in fish the mapping of preproghrelin and ghrelin receptor ghs-r1a in brain and gastrointestinal tract of goldfish, and provide the first evidence for a daily regulation of both genes expression in such locations, suggesting a possible connection between the ghrelinergic and circadian systems in teleosts.


Assuntos
Ritmo Circadiano/genética , Grelina/biossíntese , Receptores de Grelina/biossíntese , Receptores de Grelina/metabolismo , Animais , Encéfalo/metabolismo , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica , Grelina/genética , Grelina/metabolismo , Carpa Dourada/genética , Hipófise/metabolismo , Receptores de Grelina/genética
8.
J Neuroendocrinol ; 27(12): 872-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26442444

RESUMO

Ghrelin, a gut hormone originating from the post-translational cleavage of preproghrelin, is the endogenous ligand of growth hormone secretagogue receptor 1a (GHS-R1a). Within the growth hormone (GH) axis, the biological activity of ghrelin requires octanoylation by ghrelin-O-acyltransferase (GOAT), conferring selective binding to the GHS-R1a receptor via acylated ghrelin. Complete loss of preproghrelin-derived signalling (through deletion of the Ghrl gene) contributes to a decline in peak GH release; however, the selective contribution of endogenous acyl-ghrelin to pulsatile GH release remains to be established. We assessed the pulsatile release of GH in ad lib. fed male germline goat(-/-) mice, extending measures to include mRNA for key hypothalamic regulators of GH release, and peripheral factors that are modulated relative to GH release. The amount of GH released was reduced in young goat(-/-) mice compared to age-matched wild-type mice, whereas pulse frequency and irregularity increased. Altered GH release did not coincide with alterations in hypothalamic Ghrh, Srif, Npy or Ghsr mRNA expression, or pituitary GH content, suggesting that loss of Goat does not compromise canonical mechanisms that contribute to pituitary GH production and release. Although loss of Goat resulted in an irregular pattern of GH release (characterised by an increase in the number of GH pulses observed during extended secretory events), this did not contribute to a change in the expression of sexually dimorphic GH-dependent liver genes. Of interest, circulating levels of insulin-like growth factor (IGF)-1 were elevated in goat(-/-) mice. This rise in circulating levels of IGF-1 was correlated with an increase in GH pulse frequency, suggesting that sustained or increased IGF-1 release in goat(-/-) mice may occur in response to altered GH release patterning. Our observations demonstrate that germline loss of Goat alters GH release and patterning. Although the biological relevance of altered GH secretory patterning remains unclear, we propose that this may contribute to sustained IGF-1 release and growth in goat(-/-) mice.


Assuntos
Aciltransferases/deficiência , Aciltransferases/fisiologia , Hormônio do Crescimento/metabolismo , Aciltransferases/genética , Animais , Hormônio Liberador de Hormônio do Crescimento/biossíntese , Hipotálamo/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Neuropeptídeo Y/biossíntese , Receptores de Grelina/biossíntese , Somatostatina/biossíntese
9.
Peptides ; 69: 40-6, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25828736

RESUMO

Although considerable progress has been made in understanding how the temporal and regional control of neural progenitor cells (NPCs) dictates their fate, their key regulators during neural development are still unknown. Ghrelin, which is isolated from porcine stomach extract, is an endogenous ligand for the growth hormone secretagogue receptor (GHS-R). The widespread expression of ghrelin and GHS-R in the central nervous system during development suggests that ghrelin may be involved in developmental neural growth. However, its role in regulating fetal NPCs is still unclear. In this study, we investigated the effects of ghrelin on primary cultured NPCs derived from fetal mouse telencephalon. The expressions of both ghrelin and its receptor were observed in NPCs using RT-PCR, immunoblotting and immunocytostaining. Interestingly, the exposure of fetal NPCs to ghrelin at concentrations of 10(-7) and 10(-9)M suppressed their proliferation, and caused them to differentiate into neurons and to extend neurites. These results strongly suggest that ghrelin plays an autocrine modulatory role in fetal neural development.


Assuntos
Diferenciação Celular/genética , Grelina/biossíntese , Células-Tronco Neurais/metabolismo , Receptores de Grelina/biossíntese , Animais , Sistema Nervoso Central/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Grelina/administração & dosagem , Camundongos , Neurogênese/genética , Receptores de Grelina/genética , Suínos
11.
Pharmacol Rev ; 66(4): 984-1001, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25107984

RESUMO

The only molecularly identified ghrelin receptor is the growth hormone secretagogue receptor GHSR1a. Its natural ligand, ghrelin, is an acylated peptide whose unacylated counterpart (UAG) is almost inactive at GHSR1a. A truncated, nonfunctional receptor, GHSR1b, derives from the same gene. We have critically evaluated evidence for effects of ghrelin receptor ligands that are not consistent with actions at GHSR1a. Effects of ghrelin are observed in cells or tissues where the expression of GHSR1a is not detectable or after the Ghsr gene has been inactivated. In several, effects of ghrelin are mimicked by UAG, and ghrelin binding is competitively reduced by UAG. Effects in the absence of GHSR1a and sites at which ghrelin and UAG have similar potency suggest the presence of novel nonspecific ghrelin receptors (ghrelin receptor-like receptors [GRLRs]). A third class of receptor, the UAG receptors, at which UAG, but not ghrelin, is an agonist has been proposed. None of the novel receptors, with the exception of the glycoprotein CD36, which accounts for ghrelin action at a limited number of sites, have been identified. GHSR1a and GHSR1b combine with other G protein-coupled receptors to form heterodimers, whose pharmacologies differ from their components. Thus, it is feasible some GRLRs and some UAG receptors are heterodimers. Effects mediated through GRLRs or UAG receptors include adipocyte lipid accumulation, myoblast differentiation, osteoblast proliferation, insulin release, cardioprotection, coronary artery constriction, vascular endothelial cell proliferation, and tumor cell proliferation. The molecular identification and pharmacologic characterization of novel ghrelin receptors are thus important objectives.


Assuntos
Grelina/análogos & derivados , Grelina/metabolismo , Receptores de Grelina/metabolismo , Animais , Humanos , Receptores de Grelina/biossíntese
12.
Regul Pept ; 192-193: 1-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25058156

RESUMO

Ghrelin is a novel growth hormone-releasing peptide, which has been shown to exert beneficial effects on cardiac function and ventricular remodeling. The present study aimed to investigate the expression of ghrelin and the growth hormone (GH) secretagogue receptor 1a (GHSR-1a), and the association with cardiac remodeling in rats with myocardial infarction (MI). Twenty-four hours after ligation of the anterior descending artery (LAD), adult male Sprague-Dawley rats were randomized to 3 d, 7 d and 28 d group. Sham animals underwent thoracotomy and pericardiotomy, but not LAD ligation. Expression of both ghrelin and GHSR-1a was assessed by means of immunohistochemistry and real-time PCR. Plasma ghrelin levels were measured by ELISA kit. In addition, cardiac remodeling was assessed by echocardiographic and hemodynamic measurements. Plasma and cardiac expression of ghrelin decreased on days 3, 7 and 28 compared with the sham group (P<0.05). In contrast the GHSR-1a mRNA levels increased during the same days (P<0.05). Decreased positive immunoreaction for ghrelin and increased positive GHSR-1a were also observed in the infarcted heart. Interestingly, plasma ghrelin correlated negatively with left ventricular end-diastolic pressure (r=-0.59, P=0.002) and left ventricular end-diastolic dimension (r=-0.73, P<0.01). The ghrelin system may play an important role regulating cardiac remodeling after MI and present as a potential significant target for pharmacological modulation and treating cardiac remodeling.


Assuntos
Vasos Coronários/metabolismo , Vasos Coronários/cirurgia , Grelina/biossíntese , Grelina/genética , Receptores de Grelina/biossíntese , Receptores de Grelina/genética , Animais , Grelina/sangue , Imuno-Histoquímica , Ligadura , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
13.
Front Horm Res ; 43: 125-33, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24943303

RESUMO

Ghrelin is a peptide that was originally isolated from the stomach. It exerts potent growth hormone (GH)-releasing and orexigenic activities. Several studies have highlighted the therapeutic benefits of ghrelin for the treatment of cardiovascular disease. In animal models of chronic heart failure, the administration of ghrelin improved cardiac function and remodeling; these findings were replicated in human patients with heart failure. Moreover, in an animal study, ghrelin administration effectively reduced pulmonary hypertension induced by chronic hypoxia. In addition, repeated administration of ghrelin to cachectic patients with chronic obstructive pulmonary disease had positive effects on overall body function, including muscle wasting, functional capacity and sympathetic activity. The administration of ghrelin early after myocardial infarction (MI) reduced fatal arrhythmia and related mortality. In ghrelin-deficient mice, both exogenous and endogenous ghrelin were protective against fatal arrhythmia and promoted remodeling after MI. Although the mechanisms underlying the effects of ghrelin on the cardiovascular system remain unclear, there are indications that its beneficial effects are mediated through both direct physiological actions, including increased GH levels, improved energy balance and direct actions on cardiovascular cells, and regulation of autonomic nervous system activity. Therefore, ghrelin is a promising novel therapeutic agent for cardiovascular disease.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Grelina/fisiologia , Animais , Sistema Cardiovascular , Mucosa Gástrica/metabolismo , Grelina/biossíntese , Grelina/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Infarto do Miocárdio/tratamento farmacológico , Receptores de Grelina/biossíntese , Sistema Nervoso Simpático/efeitos dos fármacos
14.
Peptides ; 57: 59-66, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24780389

RESUMO

Recent studies evidenced a decrease in ghrelin's aqueous humor levels in patients with glaucoma. The goal of our investigation was to study the effect of the ghrelin-GHSR-1a system in the modulation of intraocular pressure in acute ocular hypertension models and its expression and distribution in ocular tissues. Two animal models of acute ocular hypertension were used to study the effect of the ghrelin-GHSR-1a system in the modulation of intraocular pressure: the rabbit and the rat. Ocular hypertension was induced by an intravitreal injection of 20% NaCl. Ghrelin or des-acyl ghrelin were delivered subconjunctivally and the intraocular pressure was assessed by a rebound tonometer that was calibrated for each species. In addition, we have studied the influence of nitric oxide and prostaglandins on ghrelin's effect in the rabbit animal model. Finally, we determined by immunofluorescence the expression of ghrelin and GHSR-1 in the rat's ocular tissue. Ghrelin decreased the intraocular pressure in both animal models (maximum decrease: 43.8±12.0% in the rabbit and 29.0±7.46% in the rat). In the rabbit, this effect was blunted in the presence of l-NAME and ketorolac. Des-acyl ghrelin only decreased the intraocular pressure in the rat (maximum decrease: 34.9±8.15%). Ghrelin expression was detected in the ciliary processes and GHSR-1 expression was detected in the trabecular meshwork and ciliary body. The ghrelin-GHSR-1 system is expressed in the anterior segment of the eye. Ghrelin and des-acyl ghrelin are responsible for a hypotensive effect in acute ocular hypertension animal models.


Assuntos
Grelina/biossíntese , Glaucoma/genética , Hipertensão Ocular/genética , Receptores de Grelina/biossíntese , Animais , Cílios/metabolismo , Olho/metabolismo , Olho/patologia , Grelina/genética , Glaucoma/metabolismo , Glaucoma/patologia , Humanos , Pressão Intraocular/genética , Hipertensão Ocular/induzido quimicamente , Hipertensão Ocular/patologia , Coelhos , Ratos , Receptores de Grelina/genética , Cloreto de Sódio/toxicidade , Malha Trabecular/metabolismo
15.
Regul Pept ; 190-191: 18-24, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809812

RESUMO

AIM OF THE STUDY: The gastrointestinal peptide hormone ghrelin (Ghr) was discovered in 1999 as the endogenous ligand for the growth hormone secretagogue receptor (GHSR-1a). It is a pleiotropic peptide that modulates a wide spectrum of biological activities, such as growth hormone (GH) release, feeding stimulation, adiposity and cardiovascular actions. The presence of Ghr mRNA in the iris and ciliary body (CB) epithelium was recently demonstrated in animal models, where a possible myorelaxing effect on the iris muscles has been suggested. Based on these observations, the aim of our study was to investigate the Ghr and GHSR-1a expression and localization in the normal human eye. MATERIAL: Five different ciliary body/iris samples from normal eyes were subjected to Western blot analysis. Immunohistochemical detection was performed on three enucleated eyes. Twenty aqueous humor (AqH) samples obtained from patients submitted to cataract surgery were analyzed with an ELISA for the presence of Ghr. RESULTS: Ghr and GHSR-1a were co-expressed by the pigmented epithelium (PE) of the CB, by the retinal pigmented epithelium (RPE) and by the anterior limiting layer (ALL) of the iris. No reaction was detected at the subepithelial level in the ciliary or pupillae smooth muscle cells. The AqH samples were positive for the presence of Ghr. CONCLUSION: This study provides the first evidence that Ghr and GHSR-1a are expressed in the human eye by specific cells. The understanding of the functional role of Ghr at the human eye level needs more efforts and investigation, but a hypothetical action on the GH retinal synthesis and/or on the circadian clock system could be suggested.


Assuntos
Olho/metabolismo , Grelina/análise , Receptores de Grelina/análise , Grelina/biossíntese , Humanos , Receptores de Grelina/biossíntese
16.
Circ Heart Fail ; 7(4): 643-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24855152

RESUMO

BACKGROUND: The gut-derived hormone ghrelin, especially its acylated form, plays a major role in the regulation of systemic metabolism and exerts also relevant cardioprotective effects; hence, it has been proposed for the treatment of heart failure (HF). We tested the hypothesis that ghrelin can directly modulate cardiac energy substrate metabolism. METHODS AND RESULTS: We used chronically instrumented dogs, 8 with pacing-induced HF and 6 normal controls. Human des-acyl ghrelin [1.2 nmol/kg per hour] was infused intravenously for 15 minutes, followed by washout (rebaseline) and infusion of acyl ghrelin at the same dose. (3)H-oleate and (14)C-glucose were coinfused and arterial and coronary sinus blood sampled to measure cardiac free fatty acid and glucose oxidation and lactate uptake. As expected, cardiac substrate metabolism was profoundly altered in HF because baseline oxidation levels of free fatty acids and glucose were, respectively, >70% lower and >160% higher compared with control. Neither des-acyl ghrelin nor acyl ghrelin significantly affected function and metabolism in normal hearts. However, in HF, des-acyl and acyl ghrelin enhanced myocardial oxygen consumption by 10.2±3.5% and 9.9±3.7%, respectively (P<0.05), and cardiac mechanical efficiency was not significantly altered. This was associated, respectively, with a 41.3±6.7% and 32.5±10.9% increase in free fatty acid oxidation and a 31.3±9.2% and 41.4±8.9% decrease in glucose oxidation (all P<0.05). CONCLUSIONS: Acute increases in des-acyl or acyl ghrelin do not interfere with cardiac metabolism in normal dogs, whereas they enhance free fatty acid oxidation and reduce glucose oxidation in HF dogs, thus partially correcting metabolic alterations in HF. This novel mechanism might contribute to the cardioprotective effects of ghrelin in HF.


Assuntos
Metabolismo Energético/genética , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Hemodinâmica , Miócitos Cardíacos/metabolismo , RNA/genética , Receptores de Grelina/genética , Animais , Western Blotting , Modelos Animais de Doenças , Cães , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Masculino , Miócitos Cardíacos/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Grelina/biossíntese
17.
Peptides ; 57: 1-11, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24768902

RESUMO

The aim of our study was to investigate the appearance, density and distribution of ghrelin cells and GHS-R1a and GHS-R1b in the human stomach and duodenum during prenatal and early postnatal development. We examined chromogranin-A and ghrelin cells in duodenum, and GHS-R1a and GHS-R1b expression in stomach and duodenum by immunohistochemistry in embryos, fetuses, and infants. Chromogranin-A and ghrelin cells were identified in the duodenum at weeks 10 and 11 of gestation. Ghrelin cells were detected individually or clustered within the base of duodenal crypts and villi during the first trimester, while they were presented separately within the basal and apical parts of crypts and villi during the second and third trimesters. Ghrelin cells were the most numerous during the first (∼11%) and third (∼10%) trimesters of gestation development. GHS-R1a and GHS-R1b were detected at 11 and 16 weeks of gestation, showed the highest level of expression in Brunner's gland and in lower parts of duodenal crypts and villi during the second trimester in antrum, and during the third trimester in corpus and duodenum. Our findings demonstrated for the first time abundant duodenal expression of ghrelin cells and ghrelin receptors during human prenatal development indicating a role of ghrelin in the regulation of growth and differentiation of human gastrointestinal tract.


Assuntos
Desenvolvimento Embrionário/genética , Trato Gastrointestinal/metabolismo , Receptores de Grelina/biossíntese , Glândulas Duodenais/metabolismo , Cromogranina A , Duodeno/crescimento & desenvolvimento , Duodeno/metabolismo , Feminino , Feto , Mucosa Gástrica/metabolismo , Trato Gastrointestinal/crescimento & desenvolvimento , Grelina/biossíntese , Humanos , Recém-Nascido , Gravidez , RNA Mensageiro/biossíntese
18.
Oncol Rep ; 30(5): 2063-70, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23982275

RESUMO

The only orexigenic peptide, ghrelin, which is primarily produced by the gastrointestinal tract, has been implicated in malignant cell proliferation and invasion. Ghrelin is a natural ligand of the growth hormone secretagogue receptor 1a (GHSR1a). However, the role of ghrelin in ovarian epithelial carcinoma remains unknown since the expression of GHSR1a in ovary is not confirmed. The aim of the present study was to assess expression of ghrelin and its receptor in human ovarian epithelial carcinoma and to examine the effect of ghrelin on carcinoma cell proliferation. Frozen sections of ovarian samples and the human ovarian epithelial carcinoma cell line, HO-8910, were used to characterize the expression of ghrelin/GHSR1a axis and the effect of ghrelin on proliferation. We found that ghrelin and GHSR1a are expressed in ovarian epithelial carcinoma in vivo and in vitro. Ghrelin inhibits the proliferation and growth of HO-8910 cells by G1 phase arrest, and this inhibition may be abolished by the ghrelin receptor antagonist D-Lys-3-GH-releasing peptide-6 and ghrelin neutralizing antibody. Ghrelin enhances HO-8910 cell apoptosis and autophagy. The activation of mammalian target of rapamycin (mTOR) signaling pathway blocks the effects of ghrelin-induced autophagy and apoptosis, therefore reverses the inhibition of HO-8910 cell proliferation induced by ghrelin. In conclusion, the present study demonstrates that ghrelin inhibits the proliferation of human HO-8910 ovarian epithelial carcinoma cells by inducing apoptosis and autophagy via the mTOR signaling pathway. This study provides a novel regulatory signaling pathway of ghrelin-regulated ovarian epithelial carcinoma growth and may contribute to ovarian cancer prevention and therapy.


Assuntos
Carcinoma/genética , Grelina/farmacologia , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Receptores de Grelina/biossíntese , Animais , Apoptose/genética , Carcinoma/patologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Grelina/genética , Humanos , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Receptores de Grelina/antagonistas & inibidores , Receptores de Grelina/genética , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética
19.
J Endocrinol ; 218(1): 49-59, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23608221

RESUMO

Ghrelin, an endogenous ligand for the GH secretagogue receptor (GHS-R) receptor 1a (GHS-R1a), has been implicated in several physiologic processes involving the hippocampus. The aim of this study was to investigate the molecular mechanisms of ghrelin-stimulated neurogenesis using cultured adult rat hippocampal neural stem cells (NSCs). The expression of GHS-R1a was detected in hippocampal NSCs, as assessed by western blot analysis and immunocytochemistry. Ghrelin treatment increased the proliferation of cultured hippocampal NSCs assessed by BrdU incorporation. The exposure of cells to the receptor-specific antagonist d-Lys-3-GHRP-6 abolished the proliferative effect of ghrelin. By contrast, ghrelin showed no significant effect on cell differentiation. The expression of GHS-R1a was significantly increased by ghrelin treatment. The analysis of signaling pathways showed that ghrelin caused rapid activation of ERK1/2 and Akt, which were blocked by the GHS-R1a antagonist. In addition, ghrelin stimulated the phosphorylation of Akt downstream effectors, such as glycogen synthase kinase (GSK)-3ß, mammalian target of rapamycin (mTOR), and p70(S6K). The activation of STAT3 was also caused by ghrelin treatment. Furthermore, pretreatment of cells with specific inhibitors of MEK/ERK1/2, phosphatidylinositol-3-kinase (PI3K)/Akt, mTOR, and Jak2/STAT3 attenuated ghrelin-induced cell proliferation. Taken together, our results support a role for ghrelin in adult hippocampal neurogenesis and suggest the involvement of the ERK1/2, PI3K/Akt, and STAT3 signaling pathways in the mediation of the actions of ghrelin on neurogenesis. Our data also suggest that PI3K/Akt-mediated inactivation of GSK-3ß and activation of mTOR/p70(S6K) contribute to the proliferative effect of ghrelin.


Assuntos
Células-Tronco Adultas/metabolismo , Grelina/metabolismo , Hipocampo/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Receptores de Grelina/metabolismo , Transdução de Sinais , Células-Tronco Adultas/citologia , Células-Tronco Adultas/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Grelina/antagonistas & inibidores , Grelina/genética , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Receptores de Grelina/antagonistas & inibidores , Receptores de Grelina/biossíntese , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
20.
Horm Res Paediatr ; 79: 130-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23485550

RESUMO

The preproghrelin gene is responsible for generating ghrelin and obestatin, two gastric peptides with opposite effects on food intake. Obestatin suppresses food intake and digestive motility through interaction with GPR39 (GPCR). Ghrelin is supposed to be a link connecting metabolism and energy homeostasis with growth as the result of activation of the growth hormone secretagogue receptor (GHSR).The aim of the current study was to assess the expression of preproghrelin, GPR39 and GHSR in thyroid tissues from patients with Graves' disease (GD; n = 15), non-toxic nodular goiter (NTNG; n = 10) and toxic nodular goiter (TNG; n = 10). GPR39 and GHSR in thyroid tissues were detected by immunohistochemistry and Western blot, revealing higher expression of both proteins in GD patients (+++; ++) in comparison with NTNG (+; +) and TNG (++; +) patients. GPR39 was present in thyroid autoimmune disease, NTNG and TNG at band p51 (kDa). The ghrelin receptor was identified in all study groups at p70. mRNA expression for preproghrelin was found in thyroid tissues from patients with immune and non-immune thyroid diseases. We conclude that the expression of the ghrelin receptor family in thyroid tissues may suggest a role of gastric peptides in thyroid functions. mRNA of preproghrelin expression is a proof of ghrelin gene-derived peptide presence in thyroid tissues.


Assuntos
Grelina/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Grelina/biossíntese , Doenças da Glândula Tireoide/metabolismo , Adolescente , Criança , Feminino , Bócio/metabolismo , Bócio Nodular/metabolismo , Doença de Graves/metabolismo , Humanos , Masculino , RNA Mensageiro/metabolismo , Glândula Tireoide/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...