Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Redox Biol ; 76: 103322, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39180981

RESUMO

In Parkinson's disease (PD), exogenous ghrelin protects dopaminergic neurons through its receptor, growth hormone secretagogue receptor (GHSR). However, in contrast to the strikingly low levels of ghrelin, GHSR is highly expressed in the substantia nigra (SN). What role does GHSR play in dopaminergic neurons is unknown. In this study, using GHSR knockout mice (Ghsr-/- mice) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model, we found that GHSR deletion aggravated dopaminergic neurons degeneration, and the expression and activity of GHSR were significantly reduced in PD. Furthermore, we explored the potential mechanism that GHSR deficiency aggregated PD-related neurodegeneration. We showed that DEPTOR, a subunit of mTORC1, was overexpressed in Ghsr-/- mice, positively regulating autophagy and enhancing autophagy initiation. The expression of lysosomal markers was abnormal, implying lysosomal dysfunction. As a result, the damaged mitochondria could not be effectively eliminated, which ultimately exacerbated the injury of nigral dopaminergic neurons. In particular, we demonstrated that DEPTOR could be transcriptionally regulated by KLF4. Specific knockdown of KLF4 in dopaminergic neurons effectively alleviated neurodegeneration in Ghsr-/- mice. In summary, our results suggested that endogenous GHSR deletion-compromised autophagy by impairing lysosomal function, is a key contributor to PD, which provided ideas for therapeutic approaches involving the manipulation of GHSR.


Assuntos
Autofagia , Modelos Animais de Doenças , Neurônios Dopaminérgicos , Fator 4 Semelhante a Kruppel , Camundongos Knockout , Doença de Parkinson , Receptores de Grelina , Animais , Autofagia/genética , Camundongos , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/patologia , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Receptores de Grelina/deficiência , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Fator 4 Semelhante a Kruppel/metabolismo , Substância Negra/metabolismo , Substância Negra/patologia , Masculino , Lisossomos/metabolismo
2.
Biochem Biophys Res Commun ; 727: 150270, 2024 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-38917617

RESUMO

Neuroinflammation has been implicated in cognitive deficits of neurological and neurodegenerative diseases. There is abundant evidence that the application of ghrelin, an orexigenic hormone regulating appetite and energy balance, abrogates neuroinflammation and rescues associated memory impairment. However, the underlying mechanism is uncertain. In this study, we find that both intraperitoneal (i.p.) and intracerebroventricular (i.c.v.) administration of lipopolysaccharide (LPS) impairs spatial memory in mice. LPS treatment causes neuroinflammation and microglial activation in the hippocampus. Ghsr1a deletion suppresses LPS-induced microglial activation and neuroinflammation, and rescued LPS-induced memory impairment. Our findings thus suggest that GHS-R1a signaling may promote microglial immunoactivation and contribute to LPS-induced neuroinflammation. GHS-R1a may be a new therapeutic target for cognitive dysfunction associated with inflammatory conditions.


Assuntos
Lipopolissacarídeos , Transtornos da Memória , Camundongos Endogâmicos C57BL , Microglia , Receptores de Grelina , Memória Espacial , Animais , Memória Espacial/efeitos dos fármacos , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Transtornos da Memória/genética , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Camundongos , Masculino , Microglia/metabolismo , Microglia/efeitos dos fármacos , Microglia/patologia , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Camundongos Knockout , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia
3.
Cereb Cortex ; 33(6): 2612-2625, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35797708

RESUMO

Growth hormone secretagogue receptor 1a (GHSR1a)-the receptor for orexigenic hormone ghrelin-is a G protein-coupled receptor that is widely distributed in the brain, including the hippocampus. Studies have demonstrated that genetic deletion of GHSR1a affects memory, suggesting the importance of ghrelin/GHSR1a signaling in cognitive control. However, current reports are controversial, and the mechanism underlying GHSR1a modulation of memory is uncertain. Here, we first report that global GHSR1a knockout enhances hippocampus-dependent memory, facilitates initial LTP in dorsal hippocampal Schaffer Collateral-CA1 synapses, and downregulates Akt activity in the hippocampus. Moreover, we show that the intrinsic excitability of GAD67+ interneurons-rather than neighboring pyramidal neurons in the dCA1-is suppressed by GHSR1a deletion, an effect that is antagonized by acute application of the Akt activator SC79. In addition, the inhibitory postsynaptic currents (IPSCs) on dCA1 pyramidal neurons are selectively reduced in mice with a GHSR1a deficiency. Finally, we demonstrate that selectively increasing the excitability of parvalbumin-expressing interneurons by hM3Dq-DREADDs increases IPSCs on dCA1 pyramidal neurons and normalizes memory in Ghsr1a KO mice. Our findings thus reveal a novel mechanism underlying memory enhancement of GHSR1a deficiency and herein support an adverse effect of GHSR1a signaling in hippocampus-dependent memory processes.


Assuntos
Região CA1 Hipocampal , Grelina , Memória , Células Piramidais , Receptores de Grelina , Colaterais de Schaffer , Animais , Camundongos , Grelina/genética , Grelina/metabolismo , Hipocampo/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Memória/fisiologia , Região CA1 Hipocampal/metabolismo , Colaterais de Schaffer/metabolismo
4.
Biochem Biophys Res Commun ; 589: 260-266, 2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-34929449

RESUMO

Acute respiratory distress syndrome (ARDS) is a critical illness syndrome characterized by dysregulated pulmonary inflammation. Currently, effective pharmacological treatments for ARDS are unavailable. Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor type 1a (GHS-R1a), has a pivotal role in regulating energy metabolism and immunomodulation. The role of endogenous ghrelin in ARDS remains unresolved. Herein, we investigated the role of endogenous ghrelin signaling by using GHS-R1a-null (ghsr-/-) mice and lipopolysaccharide (LPS)-induced ARDS model. Ghsr-/- mice survived longer than controls after LPS-induced lung injury. Ghsr-/- mice showed lower levels of pro-inflammatory cytokines and higher oxygenation levels after lung injury. The peritoneal macrophages isolated from ghsr-/- mice exhibited lower levels of cytokines production and oxygen consumption rate after LPS stimulation. Our results indicated that endogenous ghrelin plays a pivotal role in initiation and continuation in acute inflammatory response in LPS-induced ARDS model by modulating macrophage activity, and highlighted endogenous GHS-R1a signaling in macrophage as a potential therapeutic target in this relentless disease.


Assuntos
Regulação para Baixo , Lesão Pulmonar/patologia , Macrófagos Peritoneais/patologia , Receptores de Grelina/deficiência , Animais , Respiração Celular , Citocinas/genética , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Lesão Pulmonar/complicações , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Pneumonia/complicações , Pneumonia/patologia , Alvéolos Pulmonares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Grelina/metabolismo
5.
J Endocrinol ; 250(2): 37-48, 2021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-34060474

RESUMO

Chronic exposure to high circulating glucocorticoid or ghrelin concentrations increases food intake, weight gain and adiposity, suggesting that ghrelin could contribute to the metabolic effects of chronic glucocorticoids. In male mice, however, blocking ghrelin receptor (GHSR) signaling increased the weight gain and adiposity induced by chronic corticosterone (CORT), rather than attenuating them. In the current study, we investigated the role of GHSR signaling in the metabolic effects of chronic exposure to high circulating CORT in female mice. To do this, female WT and GHSR KO mice were treated with either CORT in a 1% ethanol (EtOH) solution or 1% EtOH alone in their drinking water for 32 days (n = 5-8/group). Body weight, food, and water intake as well as vaginal cyclicity were assessed daily. As expected, CORT treatment-induced significant increases in body weight, food intake, adiposity and also impaired glucose tolerance. In contrast to results observed in male mice, WT and GHSR KO female mice did not differ on any of these parameters. Neither plasma levels of ghrelin, LEAP-2, the endogenous GHSR antagonist produced by the liver, nor their ratio were altered by chronic glucocorticoid exposure. In addition, CORT treatment disrupted vaginal cyclicity, produced a reduction in sucrose consumption and increased locomotor activity regardless of genotype. Chronic CORT also decreased exploration in WT but not GHSR KO mice. Collectively, these data suggest that most metabolic, endocrine, reproductive and behavioral effects of chronic CORT exposure are independent of GHSR signaling in female mice.


Assuntos
Corticosterona/administração & dosagem , Obesidade/etiologia , Receptores de Grelina/fisiologia , Transdução de Sinais/fisiologia , Adiposidade/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/sangue , Comportamento Animal/efeitos dos fármacos , Corticosterona/sangue , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Grelina/sangue , Camundongos , Camundongos Knockout , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Aumento de Peso/efeitos dos fármacos
6.
Mol Metab ; 53: 101258, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34023483

RESUMO

OBJECTIVE: Acyl-ghrelin regulates eating, body weight, blood glucose, and GH secretion upon binding to its receptor GHSR (growth hormone secretagogue receptor; ghrelin receptor). GHSR is distributed in several brain regions and some peripheral cell-types including pituitary somatotrophs. The objective of the current study was to determine the functional significance of acyl-ghrelin's action on GHSR-expressing somatotrophs in mediating GH secretion and several of acyl-ghrelin's metabolic actions. METHODS: GH-IRES-Cre mice and loxP-flanked (floxed) GHSR mice were newly developed and then crossed to one another to generate mice that lacked GHSR selectively from somatotrophs. Following validation of mice with somatotroph-selective GHSR deletion, metabolic responses of these mice and control littermates were assessed following both acute and chronic acyl-ghrelin administration, a 24-h fast, and a prolonged 60% chronic caloric restriction protocol modeling starvation. RESULTS: In mice with somatotroph-selective GHSR deletion, a single peripheral injection of acyl-ghrelin failed to induce GH secretion or increase food intake, unlike wild-type and other littermate control groups. However, the usual acute blood glucose increase in response to the acyl-ghrelin bolus was preserved. Similarly, chronic s.c. acyl-ghrelin administration to mice with somatotroph-selective GHSR deletion failed to increase plasma GH, food intake, or body weight. Physiologically elevating plasma acyl-ghrelin via a 24-h fast also failed to raise plasma GH and resulted in a limited hyperphagic response upon food reintroduction in mice with somatotroph-selective GHSR deletion, although those mice nonetheless did not exhibit an exaggerated reduction in blood glucose. Physiologically elevating plasma acyl-ghrelin via a 15-day caloric restriction protocol which provided only 40% of usual daily calories failed to raise plasma GH in mice with somatotroph-selective GHSR deletion, although those mice did not exhibit life-threatening hypoglycemia. CONCLUSIONS: These results reveal that direct engagement of GHSR-expressing somatotrophs is required for a peripheral ghrelin bolus to acutely stimulate GH secretion and the actions of chronic acyl-ghrelin delivery and physiological plasma acyl-ghrelin elevations to increase plasma GH. These results also suggest that actions of acyl-ghrelin to increase food intake and body weight are reliant on direct activation of GHSRs expressed on somatotrophs. Furthermore, these results suggest that the glucoregulatory actions of acyl-ghrelin - in particular, its actions to raise blood glucose when acutely administered, prevent small blood glucose drops following a 24-h fast, and avert life-threatening hypoglycemia during an acute-on-chronic caloric restriction protocol - do not depend on GHSR expression by somatotrophs.


Assuntos
Grelina/metabolismo , Hormônio do Crescimento/metabolismo , Animais , Glicemia/metabolismo , Grelina/análogos & derivados , Camundongos , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Receptores de Grelina/metabolismo
7.
PLoS One ; 16(4): e0249420, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33793646

RESUMO

In response to cold or diet, fatty acids are dissipated into heat through uncoupling protein 1 (UCP1) in brown adipose tissue (BAT). This process is termed non-shivering thermogenesis, which is important for body temperature maintenance and contributes to obesity pathogenesis. Thermogenic enhancement has been considered a promising anti-obesity strategy. Ghrelin and its receptor Growth Hormone Secretagogue Receptor (GHS-R) have critical roles in energy intake, nutrient sensing, and lipid metabolism. We previously reported that global Ghsr-knockout mice have increased energy expenditure due to enhanced thermogenesis. To determine the site of action for GHS-R mediated thermogenesis, we generated brown adipocyte-specific Ghsr knockout mice (UCP1-CreER/Ghsrf/f) and assessed thermogenic responses under regular diet (RD) fed homeostatic metabolic state or high-fat diet (HFD) fed metabolically-impaired obese state, under normal or cold housing environment. Under a RD-feeding, UCP1-CreER/Ghsrf/f mice showed increased body fat and a slightly elevated core body temperature under cold but not under normal temperature. Consistently, the expression of thermogenic genes in BAT of RD-fed UCP1-CreER/Ghsrf/f mice was increased in reposes to cold. Under HFD feeding, HFD-fed UCP1-CreER/Ghsrf/f mice showed no difference in body fat or body temperature under either normal or cold exposure. Interestingly, the expression of thermogenic genes in BAT of HFD-fed UCP1-CreER/Ghsrf/f mice was upregulated under normal temperature but downregulated under cold exposure. Overall, our data show that GHS-R has cell-autonomous effect in brown adipocytes, and GHS-R regulates BAT thermogenic activity in a temperature- and metabolic state-dependent manner. The thermogenic effect of GHS-R in BAT is more pronounced in cold environment and differentially variable based on metabolic state; under cold exposure, GHS-R inhibition in BAT activates thermogenesis under homeostatic state but suppresses thermogenesis under obese state. Our finding collectively suggests that GHS-R in BAT, acting as a "metabolic thermostat", differentially regulates thermogenesis in response to different metabolic and thermal stimuli.


Assuntos
Tecido Adiposo Marrom/metabolismo , Receptores de Grelina/genética , Termogênese/fisiologia , Animais , Peso Corporal , Temperatura Baixa , Dieta Hiperlipídica , Camundongos , Camundongos Knockout , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo , Receptores de Grelina/deficiência , Transcriptoma
8.
Mol Metab ; 51: 101223, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33798772

RESUMO

OBJECTIVE: The orexigenic hormone ghrelin exerts its physiological effects by binding to and activating the growth hormone secretagogue receptor (GHSR). The recent development of a Ghsr-IRES-Cre knock-in mouse line has enabled to genetically access GHSR-expressing neurons. Inserting a Cre construct using a knock-in strategy, even when following an upstream internal ribosome entry site (IRES) can, however, interfere with expression of a targeted gene, with consequences for the phenotype emerging. This study aimed to phenotype, both physically and metabolically, heterozygous and homozygous Ghsr-IRES-Cre mice, with a view to discovering the extent to which the ghrelin signalling system remains functional in these mice. METHODS: We assessed feeding and arcuate nucleus (Arc) Fos activation in wild-type, heterozygous and homozygous Ghsr-IRES-Cre mice in response to peripherally-administered ghrelin. We also characterised their developmental and growth phenotypes, as well as their metabolic responses upon an overnight fast. RESULTS: Insertion of the IRES-Cre cassette into the 3'-untranslated region of the Ghsr gene led to a gene-dosage GHSR depletion in the Arc. Whereas heterozygotes remained ghrelin-responsive and more closely resembled wild-types, ghrelin had reduced orexigenic efficacy and failed to induce Arc Fos expression in homozygous littermates. Homozygotes had a lower body weight accompanied by a shorter body length, less fat tissue content, altered bone parameters, and lower insulin-like growth factor-1 levels compared to wild-type and heterozygous littermates. Moreover, both heterozygous and homozygous Ghsr-IRES-Cre mice lacked the usual fasting-induced rise in growth hormone (GH) and displayed an exaggerated drop in blood glucose and insulin compared to wild-types. Unexpectedly, fasting acyl-ghrelin levels were allele-dependently increased. CONCLUSIONS: Our data suggest that (i) heterozygous but not homozygous Ghsr-IRES-Cre mice retain the usual responsiveness to administered ghrelin, (ii) the impact of fasting on GH release and glucose homeostasis is altered even when only one copy of the Ghsr gene is non-functional (as in heterozygous Ghsr-IRES-Cre mice) and (iii) homozygous Ghsr-IRES-Cre mice exhibit growth retardation. Of the many transgenic models of suppressed ghrelin signalling, Ghsr-IRES-Cre mice emerge as best representing the full breadth of the expected phenotype with respect to body weight, growth, and metabolic parameters.


Assuntos
Jejum/metabolismo , Transtornos do Crescimento/genética , Hormônio do Crescimento/metabolismo , Receptores de Grelina/deficiência , Animais , Modelos Animais de Doenças , Dosagem de Genes , Grelina/administração & dosagem , Transtornos do Crescimento/metabolismo , Heterozigoto , Humanos , Sítios Internos de Entrada Ribossomal/genética , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Receptores de Grelina/genética
9.
Biochem Biophys Res Commun ; 529(4): 1131-1136, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32819576

RESUMO

The brain-gut hormone ghrelin and its receptor GHS-R1a, the growth hormone secretagogue receptor 1a, regulates diverse functions of central nervous system including stress response and mood. Both acute and chronic caloric restrictions (CR) were reported to increase endogenous ghrelin level meanwhile regulate anxiety-related behaviors; however, the causal relationship between CR-induced ghrelin elevation and anxiety are not fully established. Here, we introduced an acute (24 h) and a chronic (10wks) CR procedure to both GHS-R1a KO (Ghsr-/-) mice and WT (Ghsr+/+) littermates, and compared their anxiety-related behaviors. We found that acute CR induced anxiolytic and anti-despairing behaviors in Ghsr+/+ mice but not in Ghsr-/- mice. Ad-libitum refeeding abolished the effect of acute CR on anxiety-related behaviors. In contrast, chronic CR for 10wks facilitated despair-like behavior meanwhile inhibited anxiety-like behavior in Ghsr+/+ mice. GHS-R1a deficiency rescued despair-like behavior while did not affect anxiolytic response induced by chronic CR. In addition, we found elevated interleukin-6 (IL-6) in serum of Ghsr+/+ mice after chronic CR, but not in Ghsr-/- mice. Altogether, our findings indicated that acute CR and chronic CR have different impacts on anxiety-related behaviors, and the former is dependent on ghrelin/GHS-R1a signaling while the latter may not always be. In addition, our findings suggested that GHS-R1a-dependent elevation in serum IL-6 might contribute to increased despair-like behavior in chronic CR state.


Assuntos
Ansiedade/metabolismo , Comportamento Animal , Restrição Calórica , Grelina/metabolismo , Receptores de Grelina/metabolismo , Transdução de Sinais , Animais , Ansiolíticos/metabolismo , Ansiedade/sangue , Grelina/deficiência , Interleucina-6/sangue , Masculino , Camundongos Endogâmicos C57BL , Receptores de Grelina/deficiência
10.
Mol Metab ; 32: 69-84, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32029231

RESUMO

OBJECTIVE: Arcuate nucleus (ARC) neurons producing Agouti-related peptide (AgRP) and neuropeptide Y (NPY; ARCAgRP/NPY neurons) are activated under energy-deficit states. ARCAgRP/NPY neurons innervate the hypothalamic paraventricular nucleus (PVH), and ARC→PVH projections are recognized as key regulators of food intake. Plasma ghrelin levels increase under energy-deficit states and activate ARCAgRP/NPY neurons by acting on the growth hormone secretagogue receptor (GHSR). Here, we hypothesized that activation of ARCAgRP/NPY neurons in fasted mice would promote morphological remodeling of the ARCAgRP/NPY→PVH projections in a GHSR-dependent manner. METHODS: We performed 1) fluorescent immunohistochemistry, 2) imaging of green fluorescent protein (GFP) signal in NPY-GFP mice, and 3) DiI axonal labeling in brains of ad libitum fed or fasted mice with pharmacological or genetic blockage of the GHSR signaling and then estimated the density and strength of ARCAgRP/NPY→PVH fibers by assessing the mean fluorescence intensity, the absolute area with fluorescent signal, and the intensity of the fluorescent signal in the fluorescent area of the PVH. RESULTS: We found that 1) the density and strength of ARCAgRP/NPY fibers increase in the PVH of fasted mice, 2) the morphological remodeling of the ARCAgRP/NPY→PVH projections correlates with the activation of PVH neurons, and 3) PVH neurons are not activated in ARC-ablated mice. We also found that fasting-induced remodeling of ARCAgRP/NPY→PVH fibers and PVH activation are impaired in mice with pharmacological or genetic blockage of GHSR signaling. CONCLUSION: This evidence shows that the connectivity between hypothalamic circuits controlling food intake can be remodeled in the adult brain, depending on the energy balance conditions, and that GHSR activity is a key regulator of this phenomenon.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Jejum/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Grelina/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Grelina/deficiência
11.
Cardiovasc Res ; 116(13): 2091-2102, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31790138

RESUMO

AIMS: Sustained activation of ß-adrenergic signalling induces cardiac fibrosis, which marks progression to heart failure. GHSR (growth hormone secretagogue receptor) is the receptor for ghrelin, which is an orexigenic gastric hormone with newly defined cardiovascular effects. The present study determined the effects of GHSR deficiency in a mouse model of isoproterenol (ISO)-induced cardiac fibrosis and examined the underlying mechanism. METHODS AND RESULTS: Histochemical studies showed that GHSR deficiency exacerbated cardiac fibrosis. Quantitative RT-PCR, western blotting, and immunofluorescence staining demonstrated that cardiac fibroblasts isolated from GHSR-/- mice exhibited increased expression of marker genes for myofibroblast trans-differentiation (α-SMA, SM22, and calponin) upon transforming growth factor-ß treatment compared to wild-type mice. RNA-sequencing of heart transcriptomes revealed that differentially expressed genes in GHSR-/- hearts were enriched in such biological processes as extracellular matrix organization, inflammatory response, lipid metabolism, cell cycle, migration, and adhesion. Particularly, GHSR deficiency increased Wnt/ß-catenin pathway activation in ISO-induced myocardial fibrosis. In addition, loss of GHSR in macrophages instigated inflammasome activation with increased cleavage and release of interleukin-18. CONCLUSION: These results for the first time demonstrated that GHSR deficiency aggravated ISO-induced cardiac fibrosis, suggesting that GHSR was a potential target for the intervention of cardiac fibrosis.


Assuntos
Cardiomiopatias/metabolismo , Transdiferenciação Celular , Inflamassomos/metabolismo , Macrófagos/metabolismo , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Receptores de Grelina/deficiência , Animais , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/genética , Cardiomiopatias/patologia , Células Cultivadas , Modelos Animais de Doenças , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibrose , Interleucina-18/metabolismo , Isoproterenol , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/patologia , Miofibroblastos/patologia , Receptores de Grelina/genética , Via de Sinalização Wnt
12.
Neuropharmacology ; 155: 31-43, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31103617

RESUMO

Ghrelin is an orexigenic hormone that also plays an important role in mood disorders. Our previous studies demonstrated that ghrelin administration could protect against depression-like behaviors of chronic unpredictable mild stress (CUMS) in rodents. However, the mechanism related to the effect of ghrelin on CUMS mice has yet to be revealed. This article shows that ghrelin (5 nmol/kg/day for 2 weeks, i.p.) decreased depression-like behaviors induced by CUMS and increased hippocampal integrity (neurogenesis and spine density) measured via Ki67, 5-bromo-2-deoxyuridine (BrdU), doublecortin (DCX) labeling and Golgi-cox staining, which were decreased under CUMS. The behavioral phenotypes of Growth hormone secretagogue receptor (Ghsr)-null and wild type (WT) mice were evaluated under no stress condition and after CUMS exposure to determine the effect of Ghsr knockout on the behavioral phenotypes and stress susceptibility of mice. Ghsr-null mice exhibited depression-like behaviors under no stress condition. CUMS induced similar depression- and anxiety-like behavioral manifestations in both Ghsr-null and WT mice. A similar pattern of behavioral changes was observed after hippocampal GHSR knockdown. Additionally, both Ghsr knockout as well as CUMS exhibited deleterious effects on neurogenesis and spine density in the dentate gyrus (DG). Besides, CCK8 assay and 5-Ethynyl-2'-deoxyuridine (EdU) incorporation assay showed that ghrelin has a proliferative effect on primary cultured hippocampal neural stem cells (NSCs) and this proliferation was blocked by D-Lys3-GHRP-6 (DLS, the antagonist of GHSR, 100 µM) pretreatment. Ghrelin-induced proliferation is associated with the inhibition of G1 arrest, and this inhibition was blocked by LY294002 (specific inhibitor of PI3K, 20 µM). Furthermore, the in vivo data displayed that LY294002 (50 nmol, i.c.v.) can significantly block the antidepressant-like action of exogenous ghrelin treatment. All these results suggest that ghrelin/GHSR signaling maintains the integrity of hippocampus and has an inherent neuroprotective effect whether facing stress or not.


Assuntos
Grelina/deficiência , Hipocampo/metabolismo , Neurogênese/fisiologia , Fármacos Neuroprotetores/metabolismo , Receptores de Grelina/deficiência , Estresse Psicológico/metabolismo , Animais , Células Cultivadas , Cromonas/farmacologia , Doença Crônica , Proteína Duplacortina , Grelina/genética , Hipocampo/citologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfolinas/farmacologia , Neurogênese/efeitos dos fármacos , Receptores de Grelina/antagonistas & inibidores , Receptores de Grelina/genética , Estresse Psicológico/prevenção & controle , Estresse Psicológico/psicologia
13.
Cell Physiol Biochem ; 50(5): 1698-1710, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30384380

RESUMO

BACKGROUND/AIMS: Obesity is associated with a reduction in ghrelin, a 28 aa gastric hormone. Whether reduced ghrelin contributes to the impaired proliferation of hepatocytes associated with obesity-related steatosis remains largely unknown. Here we examined the effects of ghrelin on the proliferation of hepatocytes derived from lean and obese mice. METHODS: AML 12 cells or hepatocytes isolated from mice fed normal chow diet (NCD) or high fat diet (HFD) were used. Effects of ghrelin on hepatocyte proliferation were detected with CCK8 assay and EdU staining. Cell cycle was analyzed by flow cytometry. Levels of proliferation markers was examined by Western blot. RESULTS: Growth hormone secretagogue receptor 1a (GHS-R1a) mRNA and protein were present in hepatocytes. Levels of GHS-R1a were increased upon ghrelin treatment. Ghrelin significantly increased hepatocyte proliferation measured by Cell Counting Kit-8(CCK8) assay and EdU staining in a dose- and time-dependent manner. Proportion of cells in S phase was markedly increased upon treatment with ghrelin. Ghrelin significantly increased levels of proliferating cell nuclear antigen (PCNA) and cyclin D1, while reducing p27 in hepatocytes from mice fed NCD or HFD. Deletion of GHS-R1a completely abolished the effects of ghrelin in cultured hepatocytes. Ghrelin stimulated the phosphorylation of glycogen synthase kinase 3 beta (GSK3ß), leading to subsequent increase of nuclear ß-catenin in hepatocytes derived from lean and obese mice. This effect was dependent on the GHS-R1a. CONCLUSION: Ghrelin activates GHS-R1a to stimulate hepatocyte proliferation via GSK3/ß-catenin signaling pathway.


Assuntos
Proliferação de Células/efeitos dos fármacos , Grelina/farmacologia , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas , Ciclina D1/metabolismo , Dieta Hiperlipídica , Relação Dose-Resposta a Droga , Glicogênio Sintase Quinase 3 beta/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Regulação para Cima/efeitos dos fármacos , beta Catenina/metabolismo
14.
Neurosci Lett ; 681: 50-55, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-29802915

RESUMO

The vagus nerve connects peripheral organs to the central nervous system (CNS), and gastrointestinal hormones transmit their signals to the CNS via the vagal afferent nerve. Ghrelin, a gastric-derived orexigenic peptide, stimulates food intake by transmitting starvation signals via the vagus nerve. To understand peripheral ghrelin signaling via the vagus nerve, we investigated the ghrelin receptor (GHSR)-null mouse. For this purpose, we tried to produce mice in which GHSR was selectively expressed in the hindbrain and vagus nerve. GHSR was expressed in some nodose ganglion neurons in these mice, but GHSR-expressing neurons were less abundant than in wild-type mice. Intraperitoneal administration of ghrelin did not induce food intake or growth hormone release, but did increase blood glucose levels. Our findings suggest that the abundance of GHSR-expressing neurons in the nodose ganglion is critical for peripheral administration of ghrelin-induced food intake and growth hormone release via the vagus nerve.


Assuntos
Grelina/metabolismo , Gânglio Nodoso/metabolismo , Receptores de Grelina/deficiência , Transdução de Sinais/fisiologia , Nervo Vago/metabolismo , Animais , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Grelina/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Gânglio Nodoso/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
15.
J Neurochem ; 144(1): 58-67, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29063591

RESUMO

Ghrelin is an appetite-stimulating peptide. Serine 3 on ghrelin must be acylated by octanoate via the enzyme ghrelin-O-acyltransferase (GOAT) for the peptide to bind and activate the cognate receptor, growth hormone secretagogue receptor type 1a (GHSR1a). Interest in GHSR1a increased dramatically when GHSR1a mRNA was demonstrated to be widespread in the brain, including the cortex and hippocampus, indicating that it has multifaceted functions beyond the regulation of metabolism. However, the source of octanoylated ghrelin for GHSR1a in the brain, outside of the hypothalamus, is not well understood. Here, we report the presence of GOAT and its ability to acylate non-octanoylated ghrelin in the hippocampus. GOAT immunoreactivity is aggregated at the base of the dentate granule cell layer in the rat and wild-type mouse. This immunoreactivity was not affected by the pharmacological inhibition of GHSR1a or the metabolic state-dependent fluctuation of systemic ghrelin levels. However, it was absent in the GHSR1a knockout mouse hippocampus, pointing the possibility that the expression of GHSR1a may be a prerequisite for the production of GOAT. Application of fluorescein isothiocyanate (FITC)-conjugated non-octanoylated ghrelin in live hippocampal slice culture (but not in fixed culture or in the presence of GOAT inhibitors) mimicked the binding profile of FITC-conjugated octanoylated ghrelin, suggesting that extracellularly applied non-octanoylated ghrelin was acylated by endogenous GOAT in the live hippocampus while GOAT being mobilized out of neurons. Our results will advance the understanding for the role of endogenous GOAT in the hippocampus and facilitate the search for the source of ghrelin that is intrinsic to the brain.


Assuntos
Aciltransferases/metabolismo , Giro Denteado/enzimologia , Grelina/metabolismo , Acilação , Animais , Caprilatos/metabolismo , Feminino , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Grelina/farmacologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Receptores de Grelina/antagonistas & inibidores , Receptores de Grelina/deficiência , Receptores de Grelina/fisiologia
16.
PLoS One ; 12(5): e0177995, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542403

RESUMO

Cryptorchidism or undescended testis (UDT) is a common congenital abnormality associated with increased risk for developing male infertility and testicular cancer. This study elucidated the effects of endogenous ghrelin or growth hormone secretagogue receptor (GHSR) deletion on mouse reproductive performance and evaluated the ability of ghrelin to prevent testicular damage in a surgical cryptorchid mouse model. Reciprocal matings with heterozygous/homozygous ghrelin and GHSR knockout mice were performed. Litter size and germ cell apoptosis were recorded and testicular histological evaluations were performed. Wild type and GHSR knockout adult mice were subjected to creation of unilateral surgical cryptorchidism that is a model of heat-induced germ cell death. All mice were randomly separated into two groups: treatment with ghrelin or with saline. To assess testicular damage, the following endpoints were evaluated: testis weight, seminiferous tubule diameter, percentage of seminiferous tubules with spermatids and with multinucleated giant cells. Our findings indicated that endogenous ghrelin deletion altered male fertility. Moreover, ghrelin treatment ameliorated the testicular weight changes caused by surgically induced cryptorchidism. Testicular histopathology revealed a significant preservation of spermatogenesis and seminiferous tubule diameter in the ghrelin-treated cryptorchid testes of GHSR KO mice, suggesting that this protective effect of ghrelin was mediated by an unknown mechanism. In conclusion, ghrelin therapy could be useful to suppress testicular damage induced by hyperthermia, and future investigations will focus on the underlying mechanisms by which ghrelin mitigates testicular damage.


Assuntos
Criptorquidismo/patologia , Grelina/farmacologia , Testículo/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Criptorquidismo/tratamento farmacológico , Criptorquidismo/etiologia , Modelos Animais de Doenças , Feminino , Células Germinativas/citologia , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Grelina/deficiência , Grelina/genética , Grelina/uso terapêutico , Glutationa/análise , Infertilidade Masculina/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Espermatogênese/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia
17.
Int J Mol Sci ; 18(5)2017 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-28445429

RESUMO

Ghrelin receptor (Ghr-R) signaling in neurons of the ventral tegmental area (VTA) can modulate dopaminergic function and the reward-related effects of both palatable foods and drugs of abuse. In this study, we re-introduced the Ghr-R in VTA neurons in Ghr-R knockout mice (Ghr-RVTA mice) to specifically study the importance of the constitutively active Ghr-R for VTA neuronal signaling. Our results showed that re-introduction of the Ghr-R in the VTA had no impact on body weight or food intake under basal conditions. However, during novel environment stress Ghr-RVTA mice showed increased food intake and energy expenditure compared to Ghr-R knockout mice, demonstrating the significance of Ghr-R signaling in the response to stress. Ghr-RVTA mice also showed increased cocaine-induced locomotor activity compared to Ghr-R knockout mice, highlighting the importance of ghrelin signaling for the reward-related effects of activation of VTA neurons. Overall, our data suggest that re-introduction of the Ghr-R in the mesolimbic reward system of Ghr-R knockout mice increases the level of activation induced by both cocaine and novelty stress.


Assuntos
Comportamento Animal/fisiologia , Receptores de Grelina/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Peso Corporal , Dependovirus/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Ingestão de Alimentos , Metabolismo Energético , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Consumo de Oxigênio , Receptores de Dopamina D2/metabolismo , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Sci Rep ; 7: 45541, 2017 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-28361877

RESUMO

Despite being unable to activate the cognate ghrelin receptor (GHS-R), unacylated ghrelin (UAG) possesses a unique activity spectrum that includes promoting bone marrow adipogenesis. Since a receptor mediating this action has not been identified, we re-appraised the potential interaction of UAG with GHS-R in the regulation of bone marrow adiposity. Surprisingly, the adipogenic effects of intra-bone marrow (ibm)-infused acylated ghrelin (AG) and UAG were abolished in male GHS-R-null mice. Gas chromatography showed that isolated tibial marrow adipocytes contain the medium-chain fatty acids utilised in the acylation of UAG, including octanoic acid. Additionally, immunohistochemistry and immunogold electron microscopy revealed that tibial marrow adipocytes show prominent expression of the UAG-activating enzyme ghrelin O-acyl transferase (GOAT), which is located in the membranes of lipid trafficking vesicles and in the plasma membrane. Finally, the adipogenic effect of ibm-infused UAG was completely abolished in GOAT-KO mice. Thus, the adipogenic action of exogenous UAG in tibial marrow is dependent upon acylation by GOAT and activation of GHS-R. This suggests that UAG is subject to target cell-mediated activation - a novel mechanism for manipulating hormone activity.


Assuntos
Aciltransferases/metabolismo , Adipogenia , Medula Óssea/metabolismo , Grelina/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Grelina/metabolismo , Acilação , Animais , Cromatografia Gasosa , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Imunoeletrônica , Receptores de Grelina/deficiência
19.
Epilepsia ; 57(9): e195-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27378373

RESUMO

Ghrelin has anticonvulsant and neuroprotective effects in models of chemoconvulsant-induced seizures and status epilepticus. In this study we investigated whether deletion of the ghrelin receptor could alter the kindling process in the 6 Hz corneal kindling model and whether ghrelin receptor ligands possess anticonvulsant effects in fully kindled mice. Ghrelin receptor wild-type and knockout mice were electrically stimulated at a subconvulsive current twice daily via corneal electrodes until they reached the fully kindled state. Mice lacking the ghrelin receptor showed similar seizure severity during kindling acquisition as well as in the maintenance phase when compared to their wild-type littermates. Subsequently we proceeded by investigating possible anticonvulsant effects of the ghrelin receptor ligands in the acute 6 Hz seizure model and the fully 6 Hz kindled mice. The ghrelin receptor agonist JMV-1843 decreased the seizure severity score both in acutely 6 Hz stimulated mice and in fully kindled ghrelin receptor wild-type mice, but not in fully kindled ghrelin receptor knockout mice. No effect on seizure severity was observed following the ghrelin receptor antagonist JMV-2959 in both models. This finding indicates that JMV-1843 exerts an anticonvulsant effect in kindled mice via the ghrelin receptor.


Assuntos
Anticonvulsivantes/uso terapêutico , Córnea/inervação , Excitação Neurológica/efeitos dos fármacos , Receptores de Grelina/agonistas , Estado Epiléptico/tratamento farmacológico , Animais , Modelos Animais de Doenças , Estimulação Elétrica/efeitos adversos , Glicina/análogos & derivados , Glicina/uso terapêutico , Indóis , Excitação Neurológica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligopeptídeos/uso terapêutico , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Estado Epiléptico/genética , Triazóis/uso terapêutico , Triptofano/análogos & derivados
20.
Biochem Biophys Res Commun ; 479(2): 125-131, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27404127

RESUMO

Growth hormone secretagogue receptor (GHSR) is involved in appetite regulation and energy homeostasis. In the present study, we examined the role of GHSR in neointimal formation following vascular injury. In the mouse model of femoral artery wire injury, we found that vessel intima-to-media ratio was significantly reduced in GHSR deficiency (GHSR-/-) mice compared with that in wild-type mice. Immunohistochemical staining showed that the smooth muscle cell (SMCs) in the neointima were significantly decreased in the injured arteries of GHSR-/- mice which was associated with decreased SMC proliferation and migration. Furthermore, immunoblotting demonstrated that, in cultured rat aortic SMCs, small interfering RNA-mediated GHSR knockdown suppressed the activation of Akt and ERK1/2 signaling pathway. These findings suggested a novel role of GHSR in neointimal formation likely via promoting the proliferation and migration of SMCs involving Akt and ERK1/2 signaling. Therefore, GHSR may be a potential therapeutic target in restenosis and vascular remodeling.


Assuntos
Artéria Femoral/lesões , Artéria Femoral/fisiopatologia , Neointima/fisiopatologia , Receptores de Grelina/deficiência , Animais , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Artéria Femoral/metabolismo , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Neointima/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Receptores de Grelina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA