Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Pharmacol Rev ; 66(4): 984-1001, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25107984

RESUMEN

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.


Asunto(s)
Ghrelina/análogos & derivados , Ghrelina/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Humanos , Receptores de Ghrelina/biosíntesis
2.
Alcohol Alcohol ; 51(2): 121-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26330568

RESUMEN

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.


Asunto(s)
Anfetamina/administración & dosificación , Etanol/administración & dosificación , Glicina/análogos & derivados , Locomoción/efectos de los fármacos , Receptores de Ghrelina/antagonistas & inhibidores , Triazoles/administración & dosificación , Animales , Estimulantes del Sistema Nervioso Central/administración & dosificación , Glicina/administración & dosificación , Locomoción/fisiología , Masculino , Ratones , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Receptores de Ghrelina/biosíntesis , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
3.
Am J Physiol Endocrinol Metab ; 300(6): E1002-11, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21406615

RESUMEN

The adipose tissue-derived hormone leptin regulates energy balance through catabolic effects on central circuits, including proopiomelanocortin (POMC) neurons. Leptin activation of POMC neurons increases thermogenesis and locomotor activity. Protein tyrosine phosphatase 1B (PTP1B) is an important negative regulator of leptin signaling. POMC neuron-specific deletion of PTP1B in mice results in reduced high-fat diet-induced body weight and adiposity gain due to increased energy expenditure and greater leptin sensitivity. Mice lacking the leptin gene (ob/ob mice) are hypothermic and cold intolerant, whereas leptin delivery to ob/ob mice induces thermogenesis via increased sympathetic activity to brown adipose tissue (BAT). Here, we examined whether POMC PTP1B mediates the thermoregulatory response of CNS leptin signaling by evaluating food intake, body weight, core temperature (T(C)), and spontaneous physical activity (SPA) in response to either exogenous leptin or 4-day cold exposure (4°C) in male POMC-Ptp1b-deficient mice compared with wild-type controls. POMC-Ptp1b(-/-) mice were hypersensitive to leptin-induced food intake and body weight suppression compared with wild types, yet they displayed similar leptin-induced increases in T(C). Interestingly, POMC-Ptp1b(-/-) mice had increased BAT weight and elevated plasma triiodothyronine (T(3)) levels in response to a 4-day cold challenge, as well as reduced SPA 24 h after cold exposure, relative to controls. These data show that PTP1B in POMC neurons plays a role in short-term cold-induced reduction of SPA and may influence cold-induced thermogenesis via enhanced activation of the thyroid axis.


Asunto(s)
Frío , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Homeostasis/genética , Homeostasis/fisiología , Neuronas/metabolismo , Proopiomelanocortina/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Animales , Ghrelina/sangre , Hipotálamo/metabolismo , Luz , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Neuronas/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , ARN/biosíntesis , ARN/genética , ARN/aislamiento & purificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Ghrelina/biosíntesis , Transducción de Señal/fisiología , Telemetría , Termogénesis/fisiología , Hormonas Tiroideas/sangre , Tirotropina/metabolismo
4.
Cell Tissue Res ; 344(3): 481-98, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21461677

RESUMEN

Ghrelin is a pituitary growth hormone (GH)-secretagogue that also has metabolic, reproductive, proliferative, immunological and brain functions in mammals. Far less is known about its role in fish. We have therefore performed an immunohistochemical determination of its tissue distribution in the developing Atlantic halibut (Hippoglossus hippoglossus) to gain insights into its potential function. Ghrelin immunoreactivity was detected in first-feeding halibut larvae in the skin, urinary bladder, gastrointestinal (GI) tract and olfactory lobe of the brain. In subsequent stages up to metamorphosis, ghrelin immunoreactivity declined in the skin and became evident in the gills. When the stomach developed, ghrelin immunoreactivity declined throughout the GI tract with the exception of the stomach, which exhibited an intense signal. Immunoreactive ghrelin cells were also present in the olfactory lobe, nerve and epithelium and in occasional cells of the buccal cavity and oesophagus. Ghrelin immunoreactivity had an overlapping distribution with that for Na(+),K(+)-ATPase, colocalisation also being observed in some ionocytes of the gill. The co-expression of ghrelin and the GH-secretagogue receptor in the same tissue indicates that ghrelin can exert both endocrine and paracrine actions in the developing halibut. The presence of immunoreactive ghrelin in several osmoregulatory tissues, the GI tract and sensory tissue provides strong evidence that ghrelin has multiple functions during development and also suggests targets for future investigations.


Asunto(s)
Lenguado/metabolismo , Ghrelina/biosíntesis , Receptores de Ghrelina/biosíntesis , ATPasa Intercambiadora de Sodio-Potasio/biosíntesis , Animales , Lenguado/crecimiento & desarrollo , Inmunohistoquímica , Metamorfosis Biológica
5.
Cell Tissue Res ; 346(1): 135-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21956643

RESUMEN

Ghrelin, a peptide hormone from the stomach, has been recently discovered to reduce sodium excretion from the kidney. Although the effects on the kidney suggest actions in the distal nephron, the sites of expression of ghrelin receptors have not been localised. In the present work we have used a mouse that expresses green fluorescent protein under the control of the ghrelin receptor promoter to locate sites of receptor expression in the kidney. Receptor expression was confined to the straight parts of the distal tubules and the thin limbs of the loops of Henle. No expression was detected in other structures, including the glomeruli, proximal tubules and collecting ducts. Ghrelin receptors were not found in extra-renal or intra-renal arteries, despite observations that ghrelin is a vasodilator. The distribution revealed by in situ hybridisation histochemistry was the same as that revealed by the reporter. In conclusion, ghrelin receptors have a restricted distribution in the kidney. The location in the straight parts of the distal tubules accords with observations that ghrelin promotes sodium retention.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Túbulos Renales Distales/metabolismo , Asa de la Nefrona/metabolismo , Receptores de Ghrelina/biosíntesis , Animales , Transporte Iónico/fisiología , Túbulos Renales Distales/citología , Asa de la Nefrona/citología , Ratones , Ratones Transgénicos , Especificidad de Órganos/fisiología , Receptores de Ghrelina/genética , Sodio/metabolismo
6.
BMC Physiol ; 11: 12, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21801348

RESUMEN

BACKGROUND: In recent years the interest on the relationship of gut hormones to bone processes has increased and represents one of the most interesting aspects in skeletal research. The proportion of bone mass to soft tissue is a relationship that seems to be controlled by delicate and subtle regulations that imply "cross-talks" between the nutrient intake and tissues like fat. Thus, recognition of the mechanisms that integrate a gastrointestinal-fat-bone axis and its application to several aspects of human health is vital for improving treatments related to bone diseases. This work analysed the effects of gut hormones in cell cultures of three osteoblastic cell lines which represent different stages in osteoblastic development. Also, this is the first time that there is a report on the direct effects of glucagon-like peptide 2, and obestatin on osteoblast-like cells. METHODS: mRNA expression levels of five gut hormone receptors (glucose-dependent insulinotropic peptide [GIP], glucagon-like peptide 1 [GLP-1], glucagon-like peptide 2 [GLP-2], ghrelin [GHR] and obestatin [OB]) were analysed in three osteoblastic cell lines (Saos-2, TE-85 and MG-63) showing different stages of osteoblast development using reverse transcription and real time polymerase chain reaction. The responses to the gut peptides were studied using assays for cell viability, and biochemical bone markers: alkaline phosphatase (ALP), procollagen type 1 amino-terminal propeptides (P1NP), and osteocalcin production. RESULTS: The gut hormone receptor mRNA displayed the highest levels for GIP in Saos-2 and the lowest levels in MG-63, whereas GHR and GPR39 (the putative obestatin receptor) expression was higher in TE-85 and MG-63 and lower in Saos-2. GLP-1 and GLP-2 were expressed only in MG-63 and TE-85. Treatment of gut hormones to cell lines showed differential responses: higher levels in cell viability in Saos-2 after GIP, in TE-85 and MG-63 after GLP-1, GLP-2, ghrelin and obestatin. ALP showed higher levels in Saos-2 after GIP, GHR and OB and in TE-85 after GHR. P1NP showed higher levels after GIP and OB in Saos-2. Decreased levels of P1NP were observed in TE-85 and MG-63 after GLP-1, GLP-2 and OB. MG-63 showed opposite responses in osteocalcin levels after GLP-2. CONCLUSIONS: These results suggest that osteoblast activity modulation varies according to different development stage under different nutrition related-peptides.


Asunto(s)
Hormonas Gastrointestinales/farmacología , Osteoblastos/efectos de los fármacos , Fosfatasa Alcalina/biosíntesis , Línea Celular , Polipéptido Inhibidor Gástrico/farmacología , Ghrelina/farmacología , Péptido 1 Similar al Glucagón/farmacología , Péptido 2 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Receptor del Péptido 2 Similar al Glucagón , Humanos , Osteoblastos/metabolismo , Osteocalcina/biosíntesis , Fragmentos de Péptidos/biosíntesis , Procolágeno/biosíntesis , Receptores de la Hormona Gastrointestinal/biosíntesis , Receptores de Ghrelina/biosíntesis , Receptores de Glucagón/biosíntesis
7.
Mol Brain ; 14(1): 157, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34641940

RESUMEN

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.


Asunto(s)
Región CA1 Hipocampal/citología , Interneuronas/metabolismo , Memoria/fisiología , Células Piramidales/metabolismo , Receptores de Ghrelina/biosíntesis , Animales , Ghrelina/fisiología , Hibridación Fluorescente in Situ , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos , Receptores de Ghrelina/genética , Reconocimiento en Psicología , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Regulación hacia Arriba
8.
Endocrinology ; 162(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34289060

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Ghrelina/biosíntesis , Polipéptido Pancreático/metabolismo , Receptores de Ghrelina/biosíntesis , Animales , Proteínas Bacterianas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes Reporteros , Células Secretoras de Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ligandos , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Páncreas/metabolismo , Análisis de la Célula Individual , Transcriptoma
9.
Biochem Biophys Res Commun ; 393(3): 455-60, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20152815

RESUMEN

Expression of mRNA for the ghrelin receptor, GHS-R1a, was detected in various peripheral and central tissues of fetal rats, including skin, bone, heart, liver, gut, brain and spinal cord, on embryonic day (ED)15 and ED17. However, its expression in skin, bone, heart and liver, but not in gut, brain and spinal cord, became relatively weak on ED19 and disappeared after birth (ND2). Ghrelin and des-acyl ghrelin facilitated the proliferation of cultured fetal (ED17, 19), but not neonatal (ND2), skin cells. On the other hand, with regard to cells from the spinal cord and hypothalamus, the proliferative effect of ghrelin continued after birth, whereas the effect of des-acyl ghrelin on neurogenesis in these tissues was lost at the ED19 fetal and ND2 neonatal stages. Immunohistochemistry revealed that the cells in the hypothalamus induced to proliferate by ghrelin at the ND2 stage were positive for nestin and glial fibrillary acidic protein. These results suggest that in the period immediately prior to, and after birth, rat fetal cells showing proliferation in response to ghrelin and des-acyl ghrelin are at a transitional stage characterized by alteration of the expression of GHS-R1a and an undefined des-acyl ghrelin receptor, their responsiveness varying among different tissues.


Asunto(s)
Feto/citología , Feto/metabolismo , Ghrelina/metabolismo , Receptores de Ghrelina/biosíntesis , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Feto/efectos de los fármacos , Ghrelina/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Embarazo , Ratas , Ratas Wistar , Piel/efectos de los fármacos , Piel/crecimiento & desarrollo , Piel/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo
10.
Endocr J ; 56(3): 525-31, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19506321

RESUMEN

The aim of our study was to investigate the effect of the peripheral administration of ghrelin, a peptide hormone secreted from the stomach, on cellular proliferation and differentiation of progenitor cells in the adult hippocampus. Double immunohistochemical staining revealed that Ki-67-positive hippocampal progenitor cells expressed ghrelin receptors. In mice treated with ghrelin (80 microg/kg, i.p.) for 8 days, bromodeoxyuridine incorporation and doublecortin-positive neuroblasts were significantly increased in the dentate subgranular zone. We also found that the numbers of bromodeoxyuridine- and doublecortin-immunoreactive cells were significantly reduced after anti-ghrelin antibody (10 microg/kg, i.p.) treatment for 8 days. Therefore, our results indicate that ghrelin induces proliferation and differentiation of adult hippocampal progenitors, suggesting an involvement of ghrelin in hippocampal neurogenesis.


Asunto(s)
Ghrelina/fisiología , Hipocampo/fisiología , Neurogénesis/efectos de los fármacos , Animales , Anticuerpos/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ghrelina/inmunología , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Ghrelina/biosíntesis , Células Madre/fisiología
11.
Physiol Res ; 67(2): 339-346, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29303606

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Ghrelina/análogos & derivados , Ghrelina/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Glucólisis/efectos de los fármacos , Humanos , L-Lactato Deshidrogenasa/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Síndromes de Neurotoxicidad/prevención & control , Piruvaldehído/toxicidad , Receptores de Ghrelina/biosíntesis
12.
Invest Ophthalmol Vis Sci ; 58(14): 6113-6122, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29222553

RESUMEN

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.


Asunto(s)
Apoptosis , Autofagia , Regulación de la Expresión Génica , Ghrelina/genética , Glaucoma/genética , Receptores de Ghrelina/genética , Células Ganglionares de la Retina/patología , Animales , Western Blotting , Modelos Animales de Enfermedad , Ghrelina/biosíntesis , Glaucoma/metabolismo , Glaucoma/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Presión Intraocular , Masculino , ARN/genética , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/biosíntesis , Células Ganglionares de la Retina/metabolismo , Regulación hacia Arriba
13.
Gene ; 578(2): 169-76, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26692148

RESUMEN

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.


Asunto(s)
Tamaño Corporal/genética , Ghrelina/biosíntesis , Receptores de Ghrelina/biosíntesis , Caracteres Sexuales , Animales , Bagres/genética , Bagres/crecimiento & desarrollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Ghrelina/genética , Masculino , Metiltestosterona/farmacología , Receptores de Ghrelina/genética
14.
Peptides ; 69: 40-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25828736

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Ghrelina/biosíntesis , Células-Madre Neurales/metabolismo , Receptores de Ghrelina/biosíntesis , Animales , Sistema Nervioso Central/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Ghrelina/administración & dosificación , Ratones , Neurogénesis/genética , Receptores de Ghrelina/genética , Porcinos
15.
J Neuroendocrinol ; 27(12): 872-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26442444

RESUMEN

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.


Asunto(s)
Aciltransferasas/deficiencia , Aciltransferasas/fisiología , Hormona del Crecimiento/metabolismo , Aciltransferasas/genética , Animales , Hormona Liberadora de Hormona del Crecimiento/biosíntesis , Hipotálamo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Noqueados , Neuropéptido Y/biosíntesis , Receptores de Ghrelina/biosíntesis , Somatostatina/biosíntesis
16.
PLoS One ; 10(10): e0141043, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26506093

RESUMEN

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.


Asunto(s)
Ritmo Circadiano/genética , Ghrelina/biosíntesis , Receptores de Ghrelina/biosíntesis , Receptores de Ghrelina/metabolismo , Animales , Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Regulación de la Expresión Génica , Ghrelina/genética , Ghrelina/metabolismo , Carpa Dorada/genética , Hipófisis/metabolismo , Receptores de Ghrelina/genética
17.
Regul Pept ; 190-191: 18-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24809812

RESUMEN

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.


Asunto(s)
Ojo/metabolismo , Ghrelina/análisis , Receptores de Ghrelina/análisis , Ghrelina/biosíntesis , Humanos , Receptores de Ghrelina/biosíntesis
18.
Regul Pept ; 192-193: 1-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25058156

RESUMEN

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.


Asunto(s)
Vasos Coronarios/metabolismo , Vasos Coronarios/cirugía , Ghrelina/biosíntesis , Ghrelina/genética , Receptores de Ghrelina/biosíntesis , Receptores de Ghrelina/genética , Animales , Ghrelina/sangre , Inmunohistoquímica , Ligadura , Masculino , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Peptides ; 57: 59-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24780389

RESUMEN

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.


Asunto(s)
Ghrelina/biosíntesis , Glaucoma/genética , Hipertensión Ocular/genética , Receptores de Ghrelina/biosíntesis , Animales , Cilios/metabolismo , Ojo/metabolismo , Ojo/patología , Ghrelina/genética , Glaucoma/metabolismo , Glaucoma/patología , Humanos , Presión Intraocular/genética , Hipertensión Ocular/inducido químicamente , Hipertensión Ocular/patología , Conejos , Ratas , Receptores de Ghrelina/genética , Cloruro de Sodio/toxicidad , Malla Trabecular/metabolismo
20.
Circ Heart Fail ; 7(4): 643-51, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24855152

RESUMEN

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.


Asunto(s)
Metabolismo Energético/genética , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Hemodinámica , Miocitos Cardíacos/metabolismo , ARN/genética , Receptores de Ghrelina/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Perros , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocitos Cardíacos/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ghrelina/biosíntesis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda