Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biomolecules ; 12(12)2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36551241

RESUMEN

The growth hormone secretagogue receptor-1a (GHSR1a) is the endogenous receptor for ghrelin. Activation of GHSR1a participates in many physiological processes including energy homeostasis and eating behavior. Due to its transitory half-life, the efficacy of ghrelin treatment in patients is restricted; hence the development of new adjuvant therapy is an urgent need. This study aimed to establish a cell line stably expressing GHSR1a, which could be employed to screen potential ghrelin agonists from natural compounds. First, by means of lentiviral transduction, the genome of a human HEK293T cell was modified, and a cell platform stably overexpressing GHSR1a was successfully established. In this platform, GHSR1a was expressed as a fusion protein tagged with mCherry, which allowed the monitoring of the dynamic cellular distribution of GHSR1a by fluorescent microscopy. Subsequently, the authenticity of the GHSR1a mediated signaling was further characterized by using ghrelin and teaghrelin, two molecules known to stimulate GHSR1a. The results indicated that both ghrelin and teaghrelin readily activated GHSR1a mediated signaling pathways, presumably via increasing phosphorylation levels of ERK. The specific GHSR1a signaling was further validated by using SP-analog, an antagonist of GHSR1a as well as using a cell model with the knockdown expression of GHSR1a. Molecular modeling predicted that crocin might be a potential ghrelin agonist, and this prediction was further confirmed by the established platform.


Asunto(s)
Carotenoides , Ghrelina , Receptores de Ghrelina , Humanos , Ghrelina/agonistas , Células HEK293 , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Transducción de Señal , Carotenoides/farmacología
2.
Gen Comp Endocrinol ; 327: 114097, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35853503

RESUMEN

The Chinese alligator (Alligator sinensis) is a freshwater crocodilian endemic to China. So far, the endocrine regulation of feeding and growth in Chinese alligator is poorly understood. In this study, the molecular structure and tissue expression profiles of ghrelin and its receptor GHSR in the Chinese alligator were characterized for the first time. The full-length cDNA of ghrelin was 1770 bp, including a 37 bp 5 '-UTR (untranslated region), a 435 bp ORF (open reading frame) and a 1298 bp 3 '-UTR. The ORF encodes a ghrelin precursor, which consists of 145 amino acid residues, including a signal peptide with 52 amino acid residues at the N-terminus, a mature peptide with 28 amino acid residues, and a possibly obestain at the C-terminus. The full-length cDNA of GHSR was 3961 bp, including a 5'-UTR of 375-bp, an ORF of 1059-bp and a 3' -UTR of 2527-bp. The ORF encodes a protein of 352 amino acid residues containing seven transmembrane domains, with multiple N glycosylation modification sites and conserved cysteine residue sites. The active core "GSSF" of Chinese alligator ghrelin was identical to that of mammals and birds, and the ghrelin binding site of GHSR was similar to that of mammals. The amino acid sequences of both ghrelin and GHSR share high identity with American alligator (Alligator mississippiensis) and birds. Ghrelin was highly expressed in cerebrum, mesencephalon, hypothalamus and multiple peripheral tissues, including lung, stomach and intestine, suggesting that it could play functions in paracrine and/or autocrine manners in addition to endocrine manner. GHSR expression level was higher in hypothalamus, epencephalon and medulla oblongata, and moderate in multiple peripheral tissues including lung, kindey, stomach and oviduct, implicating that ghrelin/GHSR system may participate in the regulation of energy balance, food intake, water and mineral balance, gastrointestinal motility, gastric acid secretion and reproduction. During hibernation, the expression of ghrelin and GHSR in the brain was significantly increased, while ghrelin was significantly decreased in heart, liver, lung, stomach, pancreas and ovary, and GHSR was significantly decreased in heart, liver, spleen, lung, kindey, stomach, ovary and oviduct. These temporal changes in ghrelin and GHSR expression could facilitate the physiological adaption to the hibernation of Chinese alligator. Our study could provide basic data for further studies on the regulation of feeding, physiological metabolism and reproduction of Chinese alligator, which could also be useful for the improvement of artificial breeding of this endangered species.


Asunto(s)
Caimanes y Cocodrilos , Caimanes y Cocodrilos/genética , Caimanes y Cocodrilos/metabolismo , Aminoácidos , Animales , Clonación Molecular , ADN Complementario/genética , Femenino , Ghrelina/metabolismo , Mamíferos/metabolismo , ARN Mensajero/genética , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Distribución Tisular
3.
Psychoneuroendocrinology ; 144: 105862, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35835020

RESUMEN

Weight gain is the one of the most important factors which increases global burden of psychiatric disorder. Second-generation antipsychotics, olanzapine (Olz) and valproic acid (Vpa) in particular, are held responsible for weight gain. However, it is still uncertain how these drugs cause this. Thus, the rats selected for the experiment were randomly divided into 3 groups. The 1st group received only 0.5 ml saline solution intraperitoneally (n = 20, control group); the second group was given 200 mg / kg Vpa intraperitoneally (n = 20, Vpa group) and 2 mg / kg Olz was given intraperitoneally to the 3rd group (n = 20, Olz group) between 8 and 10 am for 30 days. We examined serum leptin, adiponectin, resistin, TNF-α, IL-6, ghrelin level and, the amount of ghrelin secreting cells in the stomach and growth hormone secretagogue receptor-1a (GHSR-1a, ghrelin receptor) expression in the hypothalamus. The hypothalamic GHS-1a receptor index was significantly higher in the Olz group compared with the control group and Vpa group (p = 0.036 and p = 0.016 respectively). Ghrelin immune positive cell index in stomach was statistically significantly lower in the Vpa group compared with the control and Olz groups (p = 0.028 and p = 0.013 respectively) There was no difference between the groups in terms of serum leptin, resistin, IL-6 and ghrelin levels. In the Vpa group, a statistically significant increase was found in serum adiponectin level compared with both the control group and the Olz group (p = 0009 and p = 0024 respectively) and, significant decrease was found in serum TNF-α level compared to Olz group (p = 0007). In conclusion, we found that the main cause of weight gain in Olz use was the increase in the number of hypothalamic ghrelin receptors. Investigating the mechanism by which Olz increases the number of ghrelin receptors may help to develop effective treatment strategies in preventing obesity in psychiatric patients.


Asunto(s)
Ghrelina , Receptores de Ghrelina , Adiponectina/metabolismo , Animales , Ghrelina/metabolismo , Ghrelina/farmacología , Hipotálamo/metabolismo , Interleucina-6/metabolismo , Leptina/metabolismo , Olanzapina/farmacología , Ratas , Receptores de Ghrelina/metabolismo , Resistina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ácido Valproico/farmacología , Aumento de Peso
4.
J Neuroendocrinol ; 34(4): e13126, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35365872

RESUMEN

Lactating rats show changes in the secretion of hormones and brain signals that promote hyperphagia and facilitate the production of milk. Little is known, however, about the role of ghrelin in the mechanisms sustaining lactational hyperphagia. Here, we used Wistar female rats that underwent surgery to sever the galactophores to prevent milk delivery (GC rats) and decrease the energetic drain of milk delivery. We compared plasma acyl-ghrelin concentrations and growth hormone secretagogue receptor (GHSR) mRNA expression in different brain regions of GC rats with those of sham operated lactating and nonlactating rats. Additional lactating and nonlactating rats were implanted with cannulae aimed at the lateral ventricles and were used to compare feeding responses to central ghrelin or GHSR antagonist infusions to those of nonlactating rats receiving similar infusions on day 14-16 postpartum (pp). Results show lower plasma acyl-ghrelin concentrations on day 15 pp sham operated lactating rats compared to GC or nonlactating rats. These changes occur in association with increased GHSR mRNA expression in the hypothalamic arcuate nucleus (ARC) and ventral tegmental area (VTA) of sham operated lactating rats. Despite lactational hyperphagia, infusions of ghrelin (0.25 or 1 µg) resulted in similar increases in food intake in lactating and nonlactating rats. In addition, infusions of the GHSR antagonist JMV3002 (4 µg in 1 µl of vehicle) produced greater suppression of food intake in lactating rats than in nonlactating rats. These data suggest that, despite lower plasma ghrelin, the energetic drain of lactation increases sensitivity to the orexigenic effects of ghrelin in brain regions important for food intake and energy balance, and these events are associated with lactational hyperphagia.


Asunto(s)
Ghrelina , Hipotálamo , Lactancia , Receptores de Ghrelina , Área Tegmental Ventral , Animales , Femenino , Ghrelina/sangre , Hiperfagia , Hipotálamo/metabolismo , Lactancia/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Ghrelina/metabolismo , Área Tegmental Ventral/metabolismo
5.
Neuroendocrinology ; 112(3): 215-234, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33774644

RESUMEN

INTRODUCTION: While the vast majority of research investigating the role of ghrelin or its receptor, GHS-R1a, in growth, feeding, and metabolism has been conducted in male rodents, very little is known about sex differences in this system. Furthermore, the role of GHS-R1a signaling in the control of pulsatile GH secretion and its link with growth or metabolic parameters has never been characterized. METHODS: We assessed the sex-specific contribution of GHS-R1a signaling in the activity of the GH/IGF-1 axis, metabolic parameters, and feeding behavior in adolescent (5-6 weeks old) or adult (10-19 weeks old) GHS-R KO (Ghsr-/-) and WT (Ghsr+/+) male and female mice. RESULTS: Adult Ghsr-/- male and female mice displayed deficits in weight and linear growth that were correlated with reduced GH pituitary contents in males only. GHS-R1a deletion was associated with reduced meal frequency and increased meal intervals, as well as reduced hypothalamic GHRH and NPY mRNA in males, not females. In adult, GH release from Ghsr-/- mice pituitary explants ex vivo was reduced independently of the sex. However, in vivo pulsatile GH secretion decreased in adult but not adolescent Ghsr-/- females, while in males, GHS-R1a deletion was associated with reduction in pulsatile GH secretion during adolescence exclusively. In males, linear growth did not correlate with pulsatile GH secretion, but rather with ApEn, a measure that reflects irregularity of the rhythmic secretion. Fat mass, plasma leptin concentrations, or ambulatory activity did not predict differences in GH secretion. DISCUSSION/CONCLUSION: These results point to a sex-dependent dimorphic effect of GHS-R1a signaling to modulate pulsatile GH secretion and meal pattern in mice with different compensatory mechanisms occurring in the hypothalamus of adult males and females after GHS-R1a deletion. Altogether, we show that GHS-R1a signaling plays a more critical role in the regulation of pulsatile GH secretion during adolescence in males and adulthood in females.


Asunto(s)
Ghrelina , Receptores de Ghrelina/metabolismo , Animales , Conducta Alimentaria , Femenino , Ghrelina/metabolismo , Hormona del Crecimiento/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Hipófisis/metabolismo , Receptores de Ghrelina/genética
6.
Front Endocrinol (Lausanne) ; 12: 681647, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276562

RESUMEN

Spexin (SPX) is a pleiotropic peptide with highly conserved protein sequence from fish to mammals and its biological actions are mediated by GalR2/GalR3 receptors expressed in target tissues. Recently, SPX has been confirmed to be a novel satiety factor in fish species but whether the peptide has a similar function in mammals is still unclear. Using the mouse as a model, the functional role of SPX in feeding control and the mechanisms involved were investigated. After food intake, serum SPX in mice could be up-regulated with elevations of transcript expression and tissue content of SPX in the glandular stomach but not in other tissues examined. As revealed by immunohistochemical staining, food intake also intensified SPX signals in the major cell types forming the gastric glands (including the foveolar cells, parietal cells, and chief cells) within the gastric mucosa of glandular stomach. Furthermore, IP injection of SPX was effective in reducing food intake with parallel attenuation in transcript expression of NPY, AgRP, NPY type 5 receptor (NPY5R), and ghrelin receptor (GHSR) in the hypothalamus, and these inhibitory effects could be blocked by GalR3 but not GalR2 antagonism. In agreement with the central actions of SPX, similar inhibition on feeding and hypothalamic expression of NPY, AgRP, NPY5R, and GHSR could also be noted with ICV injection of SPX. In the same study, in contrast to the drop in NPY5R and GHSR, SPX treatment could induce parallel rises of transcript expression of leptin receptor (LepR) and melanocortin 4 receptor (MC4R) in the hypothalamus. These findings, as a whole, suggest that the role of SPX as a satiety factor is well conserved in the mouse. Apparently, food intake can induce SPX production in glandular stomach and contribute to the postprandial rise of SPX in circulation. Through GalR3 activation, this SPX signal can act within the hypothalamus to trigger feedback inhibition on feeding by differential modulation of feeding regulators (NPY and AgRP) and their receptors (NPY5R, GHSR, LepR, and MC4R) involved in the feeding circuitry within the CNS.


Asunto(s)
Ingestión de Alimentos/fisiología , Hipotálamo/metabolismo , Hormonas Peptídicas/metabolismo , Saciedad/fisiología , Animales , Ratones , Receptores de Ghrelina/metabolismo , Regulación hacia Arriba
7.
Nat Prod Res ; 35(1): 57-62, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31135190

RESUMEN

Teaghrelins, identified originally in Chin-shin oolong tea, are unique acylated flavonoid tetraglycosides and proposed to be potential oral analogues of ghrelin. In the present study, two new teaghrelin-like compounds were characterized from tea cultivars (TTES No. 12), and their chemical structures were established by the spectroscopic and spectrometric analysis. However, due to the different location of rhamnose, these two teaghrelin-like compounds may not show significant ghrelin receptor affinity.[Figure: see text].


Asunto(s)
Camellia sinensis/química , Flavonoides/química , Acilación , Flavonoides/metabolismo , Ghrelina/química , Ghrelina/farmacología , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Estructura Molecular , Extractos Vegetales/química , Receptores de Ghrelina/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Té/química
8.
Physiol Behav ; 228: 113208, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33068562

RESUMEN

Ghrelin is a 28 amino acid peptide hormone that targets the brain to promote feeding and adiposity. The ghrowth hormone secretagogue receptor 1a (GHSR1a) is expressed within many hypothalamic nuclei, including the ventral premammillary nucleus (PMV), but the role of GHSR1a signaling in this region is unknown. In order to investigate whether GHSR1a signaling within the PMV modulates energy balance, we implanted osmotic minipumps connected to cannulae that were implanted intracranially and aiming at the PMV. The cannulae delivered either saline or ghrelin (10 µg/day at a flow rate of 0.11µL/h for 28 days) into the PMV of adult male C57BLJ6 mice. We found that chronic infusion of ghrelin into the PMV increased weight gain, promoted the oxidation of carbohydrates as a fuel source and resulted in hyperglycemia, without affecting food intake, or body fat. This suggests that ghrelin signaling in the PMV contributes to the modulation of metabolic fuel utilization and glucose homeostasis.


Asunto(s)
Ghrelina , Hipotálamo , Animales , Metabolismo Energético , Ghrelina/metabolismo , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Obesidad , Receptores de Ghrelina/metabolismo
9.
J Neuroendocrinol ; 33(1): e12913, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33169872

RESUMEN

Bariatric surgery is the most effective and durable means of treating obesity and its comorbidities. Women make up 80% of those receiving weight loss surgery and they experience improvements in fertility. Unfortunately, bariatric surgery in the context of pregnancy is associated with complications, including growth restriction and small-for-gestational age offspring (SGA). SGA offspring have a greater risk for obesity in adulthood, although the mechanism for this SGA-induced obesity is unknown. In a rat model of vertical sleeve gastrectomy (VSG), we previously identified reductions during pregnancy in ghrelin, a stomach-derived hormone that increases appetite and induces growth hormone secretion. Here, we hypothesise that VSG offspring will have altered ghrelin signalling compared to offspring of Sham dams as a result of reduced in utero ghrelin. At postnatal day (PND)21, male and female offspring of dams that have previously received VSG have an increase in mRNA expression for the ghrelin receptor in the hypothalamus compared to Sham offspring, and the expression of GOAT is lower in females compared to males. Liver expression of endogenous ghrelin antagonist, LEAP2, is elevated at PND60 in VSG offspring. Expression of other genes in the growth hormone system (growth hormone-releasing hormone and growth hormone) were not altered. Plasma levels of total ghrelin at PND21 are also not different between VSG and Sham pups. In adult pups, 1-hour chow intake of male but not female VSG offspring given is less than Sham offspring when given 50 µg kg-1 of exogenous ghrelin by i.p. injection. These results indicate that maternal VSG surgery has an impact on ghrelin signalling in offspring and that, as adults, male VSG offspring may be functionally less responsive to ghrelin than controls.


Asunto(s)
Gastrectomía , Ghrelina/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal/fisiología , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Femenino , Masculino , Obesidad/cirugía , Ratas , Factores Sexuales
10.
PLoS One ; 15(9): e0238465, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32903269

RESUMEN

A negative energy balance (NEB) is detrimental to reproduction in animals. A suggested link between NEB and reproductive failure is the gastrointestinal hormone ghrelin, because of the association between ghrelin and the hypothalamo-pituitary-gonadal axis. The [D-Lys3]-Growth Hormone Releasing Peptide-6 ([D-Lys3]-GHRP-6) is a ghrelin antagonist that acts on ghrelin receptors (GHS-R1). The objective of this study was to evaluate the effect of [D-Lys3]-GHRP-6 on reproduction variables in feed restricted ewes. Two experiments were conducted. Experiment I was conducted for 30 days; and Experiment II for 13 days. In both experiments the ewes (n = 18) were randomly assigned to: Control (CO): fed to meet maintenance requirements; Feed restriction (FR): 80% of maintenance restriction; or Ghrelin antagonist (GA): feed restricted and daily subcutaneous of 7.5µg/kg of [D-Lys3]-GHRP-6. Plasma was collected to measure hormones and metabolite concentration. In Experiment II, the hypothalamus and ovaries were collected on day 13. In both Experiments, sheep allocated to the FR and GA treatments decreased their body weight compared with sheep in the CO group (P < 0.06); progesterone however, did not differ between treatments (P > 0.10). Experiment I: Plasma ghrelin concentration was greater (P < 0.01) in FR and GA compared with CO ewes. Plasma non-esterified fatty acids concentration was greater (P < 0.01) in GA and FR than CO. Experiment II: Kisspeptin1-Receptor (Kiss1-R) mRNA expression was greater in FR (P < 0.01) and tended to be greater in GA (P = 0.10) compared with CO ewes. The neuro peptide-Y (NPY) mRNA expression was greater (P = 0.03) in FR than CO; and tended to be greater (P = 0.06) compared with GA ewes. Growth hormone releasing hormone (GhRH) mRNA expression was greater in GA (P = 0.04) and tended to be greater in FR (P = 0.07) compared with CO ewes. Feed restriction increased GhRH, NPY, and Kiss-R mRNA expression in hypothalamus without affecting reproductive variables.Ghrelin antagonist may prevent an increase inNPY expression in ewes.


Asunto(s)
Ghrelina/metabolismo , Oligopéptidos/farmacología , Reproducción/efectos de los fármacos , Animales , Peso Corporal , Femenino , Ghrelina/antagonistas & inhibidores , Hormona del Crecimiento/metabolismo , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hipotálamo/metabolismo , Neuropéptido Y/metabolismo , Oligopéptidos/metabolismo , ARN Mensajero/metabolismo , Receptores de Ghrelina/metabolismo , Ovinos/genética
11.
J Nutr Biochem ; 85: 108468, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32750410

RESUMEN

The mismatch between maternal undernutrition and adequate nutrition after birth increases the risk of developing metabolic diseases. We aimed to investigate whether the hyperghrelinemia during maternal undernourishment rewires the hypothalamic development of the offspring and contributes to the conversion to an obese phenotype when fed a high-fat diet (HFD). Pregnant C57BL/6 J, wild type (WT) and ghrelin receptor (GHSR)-/- mice were assigned to either a normal nourished (NN) group, or an undernutrition (UN) (30% food restricted) group. All pups were fostered by NN Swiss mice. After weaning, pups were fed a normal diet, followed by a HFD from week 9. Plasma ghrelin levels peaked at postnatal day 15 (P15) in both C57BL/6 J UN and NN pups. Hypothalamic Ghsr mRNA expression was upregulated at P15 in UN pups compared to NN pups and inhibited agouti-related peptide (AgRP) projections. Adequate lactation increased body weight of UN WT but not of GHSR-/- pups compared to NN littermates. After weaning with a HFD, body weight and food intake was higher in WT UN pups but lower in GHSR-/- UN pups than in NN controls. The GHSR prevented a decrease in ambulatory activity and oxygen consumption in UN offspring during ad libitum feeding. Maternal undernutrition triggers developmental changes in the hypothalamus in utero which were further affected by adequate feeding after birth during the postnatal period by affecting GHSR signaling. The GHSR contributes to the hyperphagia and the increase in body weight when maternal undernutrition is followed by an obesity prone life environment.


Asunto(s)
Hipotálamo/metabolismo , Desnutrición/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Obesidad/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Animales Recién Nacidos , Dieta Alta en Grasa/efectos adversos , Femenino , Eliminación de Gen , Hipotálamo/crecimiento & desarrollo , Masculino , Desnutrición/complicaciones , Ratones Endogámicos C57BL , Obesidad/etiología , Embarazo , Receptores de Ghrelina/genética
12.
Mol Metab ; 39: 101004, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32339772

RESUMEN

OBJECTIVE: Binding of ghrelin to its receptor, growth hormone secretagogue receptor (GHSR), stimulates GH release, induces eating, and increases blood glucose. These processes may also be influenced by constitutive (ghrelin-independent) GHSR activity, as suggested by findings in short people with naturally occurring GHSR-A204E mutations and reduced food intake and blood glucose in rodents administered GHSR inverse agonists, both of which impair constitutive GHSR activity. In this study, we aimed to more fully determine the physiologic relevance of constitutive GHSR activity. METHODS: We generated mice with a GHSR mutation that replaces alanine at position 203 with glutamate (GHSR-A203E), which corresponds to the previously described human GHSR-A204E mutation, and used them to conduct ex vivo neuronal electrophysiology and in vivo metabolic assessments. We also measured signaling within COS-7 and HEK293T cells transfected with wild-type GHSR (GHSR-WT) or GHSR-A203E constructs. RESULTS: In COS-7 cells, GHSR-A203E resulted in lower baseline IP3 accumulation than GHSR-WT; ghrelin-induced IP3 accumulation was observed in both constructs. In HEK293T cells co-transfected with voltage-gated CaV2.2 calcium channel complex, GHSR-A203E had no effect on basal CaV2.2 current density while GHSR-WT did; both GHSR-A203E and GHSR-WT inhibited CaV2.2 current in the presence of ghrelin. In cultured hypothalamic neurons from GHSR-A203E and GHSR-deficient mice, native calcium currents were greater than those in neurons from wild-type mice; ghrelin inhibited calcium currents in cultured hypothalamic neurons from both GHSR-A203E and wild-type mice. In brain slices, resting membrane potentials of arcuate NPY neurons from GHSR-A203E mice were hyperpolarized compared to those from wild-type mice; the same percentage of arcuate NPY neurons from GHSR-A203E and wild-type mice depolarized upon ghrelin exposure. The GHSR-A203E mutation did not significantly affect body weight, body length, or femur length in the first ∼6 months of life, yet these parameters were lower in GHSR-A203E mice after 1 year of age. During a 7-d 60% caloric restriction regimen, GHSR-A203E mice lacked the usual marked rise in plasma GH and demonstrated an exaggerated drop in blood glucose. Administered ghrelin also exhibited reduced orexigenic and GH secretagogue efficacies in GHSR-A203E mice. CONCLUSIONS: Our data suggest that the A203E mutation ablates constitutive GHSR activity and that constitutive GHSR activity contributes to the native depolarizing conductance of GHSR-expressing arcuate NPY neurons. Although the A203E mutation does not block ghrelin-evoked signaling as assessed using in vitro and ex vivo models, GHSR-A203E mice lack the usual acute food intake response to administered ghrelin in vivo. The GHSR-A203E mutation also blunts GH release, and in aged mice leads to reduced body length and femur length, which are consistent with the short stature of human carriers of the GHSR-A204E mutation.


Asunto(s)
Alelos , Sustitución de Aminoácidos , Metabolismo Energético/genética , Mutación , Receptores de Ghrelina/genética , Animales , Pesos y Medidas Corporales , Señalización del Calcio , Línea Celular , Fenómenos Electrofisiológicos , Regulación de la Expresión Génica , Marcación de Gen , Estudios de Asociación Genética , Células HEK293 , Hormonas/metabolismo , Humanos , Hipotálamo/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Técnicas de Placa-Clamp , Receptores de Ghrelina/metabolismo
13.
Physiol Behav ; 220: 112882, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32205145

RESUMEN

Ghrelin is a 28 amino acid peptide hormone that targets the brain to promote feeding and adiposity. The ghrelin receptor, the GHSR1a, is expressed within most hypothalamic nuclei, including the DMH, but the role of GHSR1a in this region on energy balance is unknown. In order to investigate whether GHSR1a within the DMH modulate energy balance, we implanted osmotic minipumps filled with saline, ghrelin, or the GHSR1a antagonist JMV2959, and connected it to a cannula aimed unilaterally at the DMH of adult male C57BLJ6 mice and assessed their metabolic profile. We found that chronic infusion of ghrelin in the DMH promoted an increase in caloric intake as well as a decrease in energy expenditure. This translated to an overall increase in weight gain, primarily in the form of adipose tissue in ghrelin treated animals. Further, chronic ghrelin unilateral infusion into the DMH slowed glucose clearance. These results suggest that GHSR in the DMH significantly contribute to the metabolic effects produced by ghrelin.


Asunto(s)
Adiposidad , Ghrelina , Animales , Ingestión de Alimentos , Metabolismo Energético , Ghrelina/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Obesidad , Receptores de Ghrelina/metabolismo
14.
Digestion ; 101(3): 227-238, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30889598

RESUMEN

BACKGROUND: Rikkunshito has been used to treat gastrointestinal (GI) disorders. The purpose of this study was to investigate the effects of Rikkunshito, a traditional Japanese herbal medicine, on the pacemaker potentials of interstitial cells of Cajal (ICCs) from the small intestines of mice. METHODS: We isolated ICCs from the small intestines of mice, and the whole-cell patch-clamp configuration was used to record the pacemaker potentials in cultured ICCs and membrane currents. RESULTS: Rikkunshito depolarized ICC pacemaker potentials in a dose-dependent manner. Pretreatment with GSK1614343 or (D-Lys3)-growth hormone-releasing peptide-6 inhibited Rikkunshito-induced depolarization of pacemaker potentials. Intracellular GDP-ß-S inhibited Rikkunshito-induced effects. In Ca2+-free solution or in the presence of thapsigargin, Rikkunshito did not depolarize pacemaker potentials. Moreover, in the presence of U-73122 or xestospongin C, Rikkunshito-induced effects were inhibited. However, in the presence of staurosporine, Go6976 or Rottlerin, Rikkunshito depolarized pacemaker potentials. Furthermore, Rikkunshito inhibited both transient receptor potentials melastatin 7 (TRPM7) and Ca2+-activated Cl- channels (ANO1) currents. CONCLUSION: Rikkunshito depolarized pacemaker potentials of ICCs via ghrelin receptor and G protein through internal or external Ca2+-, phospholipase C-, and inositol triphosphate-dependent and protein kinase C-, TRPM7-, and ANO1-independent pathways. The study shows that Rikkunshito may alleviate GI motility disorders through its depolarizing effects on ICCs.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Motilidad Gastrointestinal/efectos de los fármacos , Células Intersticiales de Cajal/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Receptores de Ghrelina/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Motilidad Gastrointestinal/fisiología , Células Intersticiales de Cajal/fisiología , Intestino Delgado/citología , Intestino Delgado/fisiología , Ratones , Técnicas de Placa-Clamp , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos
15.
Endocr J ; 67(4): 379-386, 2020 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-31839623

RESUMEN

Rikkunshito, a traditional Japanese herbal medicine, improves appetite via activation of gastrointestinal hormone ghrelin pathway. The function of ghrelin is mediated by growth hormone secretagogue receptor (GHSR1a), and ghrelin has been known to possess diverse physiological functions including growth suppression of some cancer cells. Considering that increased ghrelin signaling by Rikkunshito could enhance sirtuin1 (SIRT1) activity in nervous system, we aimed to investigate the effect of Rikkunshito in ovarian cancer cells. Ovarian cancer cell lines were treated with Rikkunshito, and cellular viability, gene expressions and epithelial-mesenchymal transition (EMT) status were investigated. To investigate the involvement of SIRT1 by Rikkunshito in SKOV3 cancer cells, endogenous expression of SIRT1 was depleted using small interfering RNA (siRNA). Treatment with Rikkunshito elevated ghrelin, GHSR1a and SIRT1, while cellular viability was decreased. The treatment of Rikkunshito also inhibited cellular migration and invasion status in a dose-dependent manner, and these effects were translated to the enhanced EMT status, although the role of SIRT1 was not determined. Our study revealed a novel function of Rikkunshito in enhancing EMT status of ovarian cancer cells. Therefore, we would like to propose that Rikkunshito may be used as a novel adjunctive therapy in chemotherapy of ovarian cancer because platinum-based chemotherapy frequently used for the treatment of ovarian cancer inevitably impairs appetite.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Ováricas/metabolismo , Receptores de Ghrelina/efectos de los fármacos , Sirtuina 1/efectos de los fármacos , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , Cadherinas/efectos de los fármacos , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ghrelina/efectos de los fármacos , Ghrelina/metabolismo , Humanos , Neoplasias Ováricas/genética , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Vimentina/efectos de los fármacos , Vimentina/metabolismo
16.
Cell Physiol Biochem ; 53(5): 851-864, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31714043

RESUMEN

BACKGROUND/AIMS: The growth promoting effect of lysine and betaine as well as the expression of candidate genes reflecting their efficacy, such as ghrelin, leptin, Growth Hormone Secretagogue Receptor (GHS-R), Insulin like Growth Factor (IGF- 1) and Growth Hormone Releasing Hormone (GHRH) was examined in Labeo rohita fingerlings. METHODS: One hundred eighty healthy juveniles from a homologous population were randomly distributed to 15 rectangular tanks of 150 litres capacity. The experiment was carried out for 60 days with five treatment groups consisting T1 (0.25% Betaine), T2 (0.5% Betaine), T3 (0.75% Lysine) and T4 (1.5% Lysine) and control group. The experiment was carried out for 60 days with five treatment groups consisting T1 (0.25% Betaine), T2 (0.5% Betaine), T3 (0.75% Lysine) and T4 (1.5% Lysine) and control group. At the end of trial, the growth parameters such as weight gain, SGR, PER were estimated from the weight of the triplicate groups. The digestive, metabolic and antioxidant enzymes were analysed using spectrophotometric methods. The intestine, brain and liver were sampled from the treatments and expression of different genes ghrelin, leptin, GHSR, IGF-1 and GHRH was also performed by realtime PCR. RESULTS: A significant (P<0.05) increase in weight gain, SGR, PER and lowest FCR was found in T4 group which was significantly (p < 0.05) different from other experimental groups. The highest mRNA expression levels of expression were found in T4 group which was similar to that of ghrelin gene mRNA of T2 group. The significantly (p<0.05) highest GHSR, GHRH and IGF-1 gene expression levels were found in T4 treatment group compared to other groups. CONCLUSION: The present study reveals that the lysine and betaine stimulate growth and expression of ghrelin GHRH, GHS-R and IGF-1 genes. The increase of IGF-I mRNA expression with lysine and betaine supplementation revealed that these compounds act as growth modulators. However, lysine was found to be a more potent modulator of growth compared to betaine.


Asunto(s)
Betaína/farmacología , Cyprinidae/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lisina/farmacología , Alimentación Animal , Animales , Catalasa/metabolismo , Cyprinidae/crecimiento & desarrollo , Ghrelina/genética , Ghrelina/metabolismo , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Leptina/genética , Leptina/metabolismo , Hígado/enzimología , Hígado/metabolismo , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Superóxido Dismutasa/metabolismo
17.
J Neuroendocrinol ; 31(7): e12763, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31251830

RESUMEN

Ghrelin is a gut hormone that signals to the hypothalamus to stimulate growth hormone release, increase food intake and promote fat deposition. The ghrelin receptor, also known as growth hormone secretagogue receptor (GHS-R), is highly expressed in the brain, with the highest expression in agouti-related peptide (AgRP) neurones in the hypothalamus. Compelling evidence indicates that ghrelin serves as a survival hormone with respect to maintaining blood glucose and body weight during nutritional deficiencies. Recent studies have demonstrated that AgRP neurones are involved in metabolic and behavioural adaptation to an energy deficit to improve survival. In the present study, we used a neuronal subtype-specific GHS-R knockout mouse (AgRP-Cre;Ghsrf/f ) to investigate the role of GHS-R in hypothalamic AgRP neurones in metabolic and behavioural adaptation to hypocaloric restricted feeding. We subjected the mice to a restricted feeding regimen of 40% mild calorie restriction (CR), with one-quarter of food allotment given in the beginning of the light cycle and three-quarters given at the beginning of the dark cycle, to mimic normal mouse intake pattern. The CR-fed AgRP-Cre;Ghsrf/f mice exhibited reductions in body weight, fat mass and blood glucose. Metabolic profiling of these CR-fed AgRP-Cre;Ghsrf/f mice showed a trend toward reduced basal metabolic rate, significantly reduced core body temperature and a decreased expression of thermogenic genes in brown adipose tissue. This suggests a metabolic reset to a lower threshold. Significantly increased physical activity, a trend toward increased food anticipatory behaviour and altered fuel preferences were also observed in these mice. In addition, these CR-fed AgRP-Cre;Ghsrf/f mice exhibited a decreased counter-regulatory response, showing impaired hepatic glucose production. Lastly, hypothalamic gene expression in AgRP-Cre;Ghsrf/f mice revealed increased AgRP expression and a decreased expression of genes in ß-oxidation pathways. In summary, our data suggest that GHS-R in AgRP neurones is a key component of the neurocircuitry involved in metabolic adaptation to calorie restriction.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Restricción Calórica , Metabolismo Energético , Hipotálamo/metabolismo , Neuronas/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Ingestión de Alimentos , Conducta Alimentaria/fisiología , Masculino , Ratones Noqueados
18.
PLoS One ; 14(3): e0214626, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30921423

RESUMEN

We estimated the effect of oligosaccharide supplementation and feed restriction on calves. The study was divided into two experimental periods of 28 days each with 20 crossbred calves that had initial body weight of 37 Kg and housed in individual pens. The animals were split in four experimental groups: animals fed 6 L milk/day (CON) in the two periods, animals fed milk restricted (3 L milk/day) in the first period and followed by CON feeding in the second period (RES), animals receiving supplementation of 5 g/day of mannanoligosaccharide (MOS) and animals receiving supplementation of 5 g/day mannan and frutoligosaccharide (MFOS). At the end of the study, all the animals were slaughtered. The average weight gain was lower in the restricted group when compared with CON and MFOS groups in the first period (P < 0.05) and there were no difference among the groups in the second period. Animals supplemented with MOS showed a significant increases in jejunal villus height and rumen papillae, which were not observed for MFOS group (P < 0.05) compared with RES and CON groups. There were no difference in ghrelin and leptin levels among treatments during periods 1 and 2 (P > 0.05). Also, the expression of ghrelin receptors in the paraventricular region of the hypothalamus did not differ among groups. We conclude that milk restriction during the first weeks of life in calves resulted in compensatory gain and did not modify the hormonal profile and expression of the ghrelin receptor in the hypothalamus. Moreover, a prebiotic supplementation changed the development of intestinal and ruminal epithelium.


Asunto(s)
Alimentación Animal/análisis , Peso Corporal/efectos de los fármacos , Suplementos Dietéticos/análisis , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/crecimiento & desarrollo , Leche , Oligosacáridos/farmacología , Animales , Bovinos , Ingestión de Alimentos/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Receptores de Ghrelina/metabolismo
19.
Biosci Rep ; 38(5)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30177523

RESUMEN

Ghrelin, an acylated peptide hormone of 28 amino acids, is an endogenous ligand of the released growth hormone secretagogue receptor (GHSR). Ghrelin has been isolated from human and rat stomach and is also detected in the hypothalamic arcuate nucleus. Ghrelin receptor is primarily located in the neuropeptide Y and agouti-related protein neurons. Many previous studies have shown that ghrelin and GHSR are involved in the regulation of energy homeostasis, and its administration can increase food intake and body weight gain. AMP-activated protein kinase is activated by ghrelin in the hypothalamus, which contributes to lower intracellular long-chain fatty acid level. Ghrelin appears to modulate the response to food cues via a neural network involved in the regulation of feeding and in the appetitive response to food cues. It also increases the response of brain areas involved in visual processing, attention, and memory to food pictures. Ghrelin is also an important factor linking the central nervous system with peripheral tissues that regulate lipid metabolism. It promotes adiposity by the activation of hypothalamic orexigenic neurons and stimulates the expression of fat storage-related proteins in adipocytes. Meanwhile, ghrelin exerts direct peripheral effects on lipid metabolism, including increase in white adipose tissue mass, stimulation of lipogenesis in the liver, and taste sensitivity modulation.


Asunto(s)
Ghrelina/genética , Metabolismo de los Lípidos/genética , Obesidad/genética , Receptores de Ghrelina/genética , Animales , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Ghrelina/metabolismo , Humanos , Hipotálamo/metabolismo , Neuropéptido Y/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Ratas , Receptores de Ghrelina/metabolismo
20.
Mol Metab ; 14: 130-138, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29914853

RESUMEN

OBJECTIVE: To date, early developmental ablation of leptin receptor (LepRb) expression from circumscribed populations of hypothalamic neurons (e.g., arcuate nucleus (ARC) Pomc- or Agrp-expressing cells) has only minimally affected energy balance. In contrast, removal of LepRb from at least two large populations (expressing vGat or Nos1) spanning multiple hypothalamic regions produced profound obesity and metabolic dysfunction. Thus, we tested the notion that the total number of leptin-responsive hypothalamic neurons (rather than specific subsets of cells with a particular molecular or anatomical signature) subjected to early LepRb deletion might determine energy balance. METHODS: We generated new mouse lines deleted for LepRb in ARC GhrhCre neurons or in Htr2cCre neurons (representing roughly half of all hypothalamic LepRb neurons, distributed across many nuclei). We compared the phenotypes of these mice to previously-reported models lacking LepRb in Pomc, Agrp, vGat or Nos1 cells. RESULTS: The early developmental deletion of LepRb from vGat or Nos1 neurons produced dramatic obesity, but deletion of LepRb from Pomc, Agrp, Ghrh, or Htr2c neurons minimally altered energy balance. CONCLUSIONS: Although early developmental deletion of LepRb from known populations of ARC neurons fails to substantially alter body weight, the minimal phenotype of mice lacking LepRb in Htr2c cells suggests that the phenotype that results from early developmental LepRb deficiency depends not simply upon the total number of leptin-responsive hypothalamic LepRb cells. Rather, specific populations of LepRb neurons must play particularly important roles in body energy homeostasis; these as yet unidentified LepRb cells likely reside in the DMH.


Asunto(s)
Metabolismo Energético , Hipotálamo/citología , Neuronas/metabolismo , Obesidad/metabolismo , Receptores de Leptina/genética , Animales , Femenino , Eliminación de Gen , Hipotálamo/embriología , Hipotálamo/metabolismo , Masculino , Ratones , Neuronas/clasificación , Neuronas/citología , Receptor de Serotonina 5-HT2C/genética , Receptor de Serotonina 5-HT2C/metabolismo , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Receptores de Leptina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA