Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Exp Biol Med (Maywood) ; 247(17): 1558-1569, 2022 09.
Article in English | MEDLINE | ID: mdl-35833540

ABSTRACT

A major component of aging is chronic, low-grade inflammation, attributable in part by impaired gut barrier function. We previously reported that deletion of ghrelin, a peptidergic hormone released mainly from the gut, exacerbates experimental muscle atrophy in aged mice. In addition, ghrelin has been shown to ameliorate colitis in experimental models of inflammatory bowel disease (IBD), although the role of endogenous ghrelin in host-microbe interactions is less clear. Here, we showed that 22-month-old global ghrelin knockout (Ghrl-/-) mice exhibited significantly increased depressive-like behaviors, while anxiety levels and working memory were similar to littermate wild-type (WT) mice. Furthermore, old Ghrl-/- mice showed significantly increased intestinal permeability to fluorescein isothiocyanate (FITC)-dextran, significantly higher colonic interleukin (IL-1ß) levels, and trends for higher colonic IL-6 and tumor necrosis factor-α (TNF-α) compared to WT mice. Interestingly, young Ghrl-/- and WT mice showed comparable depressive-like behavior and gut permeability, suggesting age-dependent exacerbation in gut barrier dysfunction in Ghrl-/- mice. While fecal short-chain fatty acids levels were comparable between old Ghrl-/- and WT mice, serum metabolome revealed alterations in metabolic cascades including tryptophan metabolism. Specifically, tryptophan and its microbial derivatives indole-3-acetic acid and indole-3-lactic acid were significantly reduced in old Ghrl-/-mice. Furthermore, in an experimental model of dextran sulfate sodium (DSS)-induced colitis, Ghrl-/- mice showed exacerbated disease symptoms, and higher levels of chemoattractant and pro-inflammatory cytokines in the colon. Overall, these data demonstrated that ghrelin deficiency is associated with gut barrier dysfunction, alterations in microbially derived tryptophan metabolites, and increased susceptibility to colitis. These data suggested that endogenous ghrelin contributes to maintaining a healthy host-microbe environment, ultimately impacting on brain function.


Subject(s)
Colitis, Ulcerative , Colitis , Ghrelin , Tryptophan , Animals , Chemotactic Factors/adverse effects , Colitis/chemically induced , Colitis/pathology , Colitis, Ulcerative/chemically induced , Cytokines/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Fatty Acids, Volatile , Fluorescein-5-isothiocyanate/analogs & derivatives , Gene Deletion , Ghrelin/deficiency , Ghrelin/genetics , Inflammation , Interleukin-6/metabolism , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Tryptophan/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Biochem Biophys Res Commun ; 529(4): 1131-1136, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819576

ABSTRACT

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.


Subject(s)
Anxiety/metabolism , Behavior, Animal , Caloric Restriction , Ghrelin/metabolism , Receptors, Ghrelin/metabolism , Signal Transduction , Animals , Anti-Anxiety Agents/metabolism , Anxiety/blood , Ghrelin/deficiency , Interleukin-6/blood , Male , Mice, Inbred C57BL , Receptors, Ghrelin/deficiency
3.
J Gerontol A Biol Sci Med Sci ; 75(4): 621-630, 2020 03 09.
Article in English | MEDLINE | ID: mdl-30407483

ABSTRACT

Sarcopenia is the aging-associated progressive loss of skeletal muscle; however, the pathogenic mechanism of sarcopenia is not clear. The orexigenic hormone ghrelin stimulates growth hormone secretion, increases food intake, and promotes adiposity. Here we showed that fasting-induced muscle loss was exacerbated in old ghrelin-null (Ghrl-/-) mice, exhibiting decreased expression of myogenic regulator MyoD and increased expression of protein degradation marker MuRF1, as well as altered mitochondrial function. Moreover, acylated ghrelin and unacylated ghrelin treatments significantly increased mitochondrial respiration capacity in muscle C2C12 cells. Consistently, acylated ghrelin and unacylated ghrelin treatments effectively increased myogenic genes and decreased degradation genes in the muscle in fasted old Ghrl-/- mice, possibly by stimulating insulin and adenosine monophosphate-activated protein kinase pathways. Furthermore, Ghrl-/- mice showed a profile of pro-inflammatory gut microbiota, exhibiting reduced butyrate-producing bacteria Roseburia and ClostridiumXIVb. Collectively, our results showed that ghrelin has a major role in the maintenance of aging muscle via both muscle-intrinsic and -extrinsic mechanisms. Acylated ghrelin and unacylated ghrelin enhanced muscle anabolism and exerted protective effects for muscle atrophy. Because unacylated ghrelin is devoid of the obesogenic side effect seen with acylated ghrelin, it represents an attractive therapeutic option for sarcopenia.


Subject(s)
Aging/pathology , Aging/physiology , Fasting/adverse effects , Ghrelin/physiology , Muscular Atrophy/prevention & control , Acylation , Adiposity/genetics , Adiposity/physiology , Aging/genetics , Animals , Disease Models, Animal , Fasting/physiology , Gastrointestinal Microbiome , Ghrelin/deficiency , Ghrelin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/metabolism , Motor Activity/genetics , Motor Activity/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Sarcopenia/etiology , Sarcopenia/physiopathology , Sarcopenia/prevention & control
4.
Neuropharmacology ; 155: 31-43, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31103617

ABSTRACT

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.


Subject(s)
Ghrelin/deficiency , Hippocampus/metabolism , Neurogenesis/physiology , Neuroprotective Agents/metabolism , Receptors, Ghrelin/deficiency , Stress, Psychological/metabolism , Animals , Cells, Cultured , Chromones/pharmacology , Chronic Disease , Doublecortin Protein , Ghrelin/genetics , Hippocampus/cytology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Neurogenesis/drug effects , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/genetics , Stress, Psychological/prevention & control , Stress, Psychological/psychology
5.
Aging Cell ; 17(1)2018 02.
Article in English | MEDLINE | ID: mdl-29024407

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide. NAFLD usually begins as low-grade hepatic steatosis which further progresses in an age-dependent manner to nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma in some patients. Ghrelin is a hormone known to promote adiposity in rodents and humans, but its potential role in hepatic steatosis is unknown. We hypothesized that genetic ghrelin deletion will protect against the development of age-related hepatic steatosis. To examine this hypothesis, we utilized ghrelin knockout (KO) mice. Although no different in young animals (3 months old), we found that at 20 months of age, ghrelin KO mice have significantly reduced hepatic steatosis compared to aged-matched wild-type (WT) mice. Examination of molecular pathways by which deletion of ghrelin reduces steatosis showed that the increase in expression of diacylglycerol O-acyltransferase-1 (DGAT1), one of the key enzymes of triglyceride (TG) synthesis, seen with age in WT mice, is not present in KO mice. This was due to the lack of activation of CCAAT/enhancer binding protein-alpha (C/EBPα) protein and subsequent reduction of C/EBPα-p300 complexes. These complexes were abundant in livers of old WT mice and were bound to and activated the DGAT1 promoter. However, the C/EBPα-p300 complexes were not detected on the DGAT1 promoter in livers of old KO mice resulting in lower levels of the enzyme. In conclusion, these studies demonstrate the mechanism by which ghrelin deletion prevents age-associated hepatic steatosis and suggest that targeting this pathway may offer therapeutic benefit for NAFLD.


Subject(s)
Age Factors , Diacylglycerol O-Acyltransferase/pharmacology , Fatty Liver/metabolism , Ghrelin/deficiency , Animals , Diacylglycerol O-Acyltransferase/drug effects , Diacylglycerol O-Acyltransferase/genetics , Down-Regulation , Fatty Liver/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/metabolism
6.
Aging Cell ; 16(4): 859-869, 2017 08.
Article in English | MEDLINE | ID: mdl-28585250

ABSTRACT

During aging, decreases in energy expenditure and locomotor activity lead to body weight and fat gain. Aging is also associated with decreases in muscle strength and endurance leading to functional decline. Here, we show that lifelong deletion of ghrelin prevents development of obesity associated with aging by modulating food intake and energy expenditure. Ghrelin deletion also attenuated the decrease in phosphorylated adenosine monophosphate-activated protein kinase (pAMPK) and downstream mediators in muscle, and increased the number of type IIa (fatigue resistant, oxidative) muscle fibers, preventing the decline in muscle strength and endurance seen with aging. Longevity was not affected by ghrelin deletion. Treatment of old mice with pharmacologic doses of ghrelin increased food intake, body weight, and muscle strength in both ghrelin wild-type and knockout mice. These findings highlight the relevance of ghrelin during aging and identify a novel AMPK-dependent mechanism for ghrelin action in muscle.


Subject(s)
AMP-Activated Protein Kinases/genetics , Energy Metabolism/genetics , Ghrelin/genetics , Longevity/genetics , Obesity/prevention & control , Sarcopenia/prevention & control , AMP-Activated Protein Kinases/metabolism , Animals , Body Weight , Eating/genetics , Gene Expression Regulation , Ghrelin/deficiency , Growth Hormone/genetics , Growth Hormone/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Sarcopenia/genetics , Sarcopenia/metabolism , Sarcopenia/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
PLoS One ; 12(5): e0177995, 2017.
Article in English | MEDLINE | ID: mdl-28542403

ABSTRACT

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.


Subject(s)
Cryptorchidism/pathology , Ghrelin/pharmacology , Testis/drug effects , Animals , Apoptosis/drug effects , Cryptorchidism/drug therapy , Cryptorchidism/etiology , Disease Models, Animal , Female , Germ Cells/cytology , Germ Cells/drug effects , Germ Cells/metabolism , Ghrelin/deficiency , Ghrelin/genetics , Ghrelin/therapeutic use , Glutathione/analysis , Infertility, Male/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics , Spermatogenesis/drug effects , Testis/metabolism , Testis/pathology
8.
Neurochem Res ; 42(9): 2658-2665, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28462451

ABSTRACT

Molecular hydrogen (H2), as a new medical gas, has protective effects in neurological disorders including Parkinson's disease (PD). In our previous report, the neuroprotective effect of drinking water with saturated H2 (H2 water) in PD mice might be due to stomach-brain interaction via release of gastric hormone, ghrelin. In the present study, we assessed the effect of H2-induced ghrelin more precisely. To confirm the contribution of ghrelin in H2 water-drinking PD model mice, ghrelin-knock out (KO) mice were used. Despite the speculation, the effect of H2 water was still observed in ghrelin-KO PD model mice. To further check the involvement of ghrelin, possible contribution of ghrelin-induced vagal afferent effect was tested by performing subdiaphragmatic vagotomy before treating with H2 water and administration of MPTP (1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine). The protective effect of H2 water was still observed in the vagotomized mice in substantia nigra, suggesting that stimulation of vagal afferent nerves is not involved in H2-induced neuroprotection. Other neuroprotective substitutes in ghrelin-KO mice were speculated because H2-induced neuroprotection was not cancelled by ghrelin receptor antagonist, D-Lys3 GHRP-6, in ghrelin-KO PD model mice, unlike in wild-type PD model mice. Our results indicate that ghrelin may not be the only factor for H2-induced neuroprotection and other factors can substitute the role of ghrelin when ghrelin is absent, raising intriguing options of research for H2-responsive factors.


Subject(s)
Brain/metabolism , Deuterium/administration & dosage , Gastric Mucosa/metabolism , Ghrelin/deficiency , Parkinsonian Disorders/metabolism , Animals , Brain/drug effects , Brain/pathology , Ghrelin/antagonists & inhibitors , Ghrelin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Stomach/drug effects , Vagotomy/methods , Vagus Nerve/metabolism , Vagus Nerve/surgery
9.
Proc Natl Acad Sci U S A ; 112(4): 1226-31, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25583513

ABSTRACT

Plasma growth hormone (GH) and hepatic autophagy each have been reported to protect against hypoglycemia in the fasted state, but previous data have not linked the two. Here we demonstrate a connection using a mouse model of fasting in a fat-depleted state. Mice were subjected to 1 wk of 60% calorie restriction, causing them to lose nearly all body fat. They were then fasted for 23 h. During fasting, WT mice developed massive increases in plasma GH and a concomitant increase in hepatic autophagy, allowing them to maintain viable levels of blood glucose. In contrast, lethal hypoglycemia occurred in mice deficient in the GH secretagogue ghrelin as a result of knockout of the gene encoding ghrelin O-acyltransferase (GOAT), which catalyzes a required acylation of the peptide. Fasting fat-depleted Goat(-/-) mice showed a blunted increase in GH and a marked decrease in hepatic autophagy. Restoration of GH by infusion during the week of calorie restriction maintained autophagy in the Goat(-/-) mice and prevented lethal hypoglycemia. Acute injections of GH after 7 d of calorie restriction also restored hepatic autophagy, but failed to increase blood glucose, perhaps owing to ATP deficiency in the liver. These data indicate that GH stimulation of autophagy is necessary over the long term, but not sufficient over the short term to maintain blood glucose levels in fasted, fat-depleted mice.


Subject(s)
Autophagy , Blood Glucose/metabolism , Caloric Restriction , Fasting/blood , Ghrelin , Hypoglycemia , Liver/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Blood Glucose/genetics , Ghrelin/deficiency , Ghrelin/pharmacology , Hypoglycemia/blood , Hypoglycemia/drug therapy , Hypoglycemia/genetics , Hypoglycemia/metabolism , Membrane Proteins , Mice , Mice, Knockout
10.
Biol Reprod ; 92(3): 76, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25631345

ABSTRACT

The chemotherapeutic drug cisplatin causes a number of dose-dependent side effects, including cachexia and testicular damage. Patients receiving a high cumulative dose of cisplatin may develop permanent azoospermia and subsequent infertility. Thus, the development of chemotherapeutic regimens with the optimal postsurvival quality of life (fertility) is of high importance. This study tested the hypothesis that ghrelin administration can prevent or minimize cisplatin-induced testicular damage and cachexia. Ghrelin and its receptor, the growth hormone secretagogue receptor (GHSR-1a), are expressed and function in the testis. Targeted deletion of ghrelin, or its receptor, significantly increases the rate of cell death in the testis, suggesting a protective role. Intraperitoneal administration of vehicle, ghrelin, or cisplatin alone or in combination with ghrelin, in cycles of 9 or 18 days, to adult male C57Bl/6 mice was performed. Body weight was measured daily and testicular and epididymal weight, sperm density and motility, testicular histology, and testicular cell death were analyzed at the time of euthanization. Ghrelin coadministration decreased the severity of cisplatin-induced cachexia and gonadal toxicity. Body, testicular, and epididymal weights significantly increased as testicular cell death decreased with ghrelin coadministration. The widespread damage to the seminiferous epithelium induced by cisplatin administration was less severe in mice simultaneously treated with ghrelin. Furthermore, ghrelin diminished the deleterious effects of cisplatin on testis and body weight homeostasis in wild-type but not Ghsr(-/-) mice, showing that ghrelin's actions are mediated via GHSR. Ghrelin or more stable GHSR agonists potentially offer a novel therapeutic approach to minimize the testicular damage that occurs after gonadotoxin exposure.


Subject(s)
Apoptosis/drug effects , Cachexia/chemically induced , Cachexia/prevention & control , Cisplatin/adverse effects , Cisplatin/pharmacology , Ghrelin/physiology , Receptors, Ghrelin/physiology , Testis/physiopathology , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Body Weight/drug effects , Cachexia/physiopathology , Ghrelin/deficiency , Ghrelin/pharmacology , Homeostasis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Organ Size/drug effects , Signal Transduction/drug effects , Spermatogenesis/drug effects , Spermatozoa/drug effects , Spermatozoa/pathology , Testis/drug effects
11.
Endocrinology ; 155(9): 3561-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24949662

ABSTRACT

Ghrelin is a gut hormone processed from the proghrelin peptide acting as the endogenous ligand of the GH secretagogue receptor 1a. The regulatory role of endogenous ghrelin on pulsatile GH secretion and linear growth had to be established. The aim of the present study was to delineate the endogenous actions of preproghrelin on peripheral and central components of the GH axis. Accordingly, the ultradian pattern of GH secretion was measured in young and old preproghrelin-deficient males. Blood samples were collected by tail bleeding every 10 minutes over a period of 6 hours. Analysis of the GH pulsatile pattern by deconvolution showed that GH was secreted in an ultradian manner in all genotypes, with major secretory peaks occurring at about 3-hour intervals. In older mice, the peak number was reduced and secretion was less irregular compared with younger animals. Remarkably, in young Ghrl(-/-) mice, the amplitude of GH secretory bursts was significantly reduced. In older mice, however, genotype differences were less significant. Changes in GH pulsatility in young Ghrl(-/-) mice were associated with a tendency for reduced GH pituitary contents and plasma IGF-I concentrations, but with only a minor impact on linear growth. In Ghrl(+/-) mice, despite reduced Acyl ghrelin to des-acyl ghrelin ratio, GH secretion was not impaired. Ghrelin deficiency was not associated with a reduction in hypothalamic GHRH content or altered response to GHRH stimulation. Therefore, reduction in GHRH production and/or sensitivity do not primarily account for the altered GH pulsatile secretion of young Ghrl(-/-) mice. Instead, GHRH expression was elevated in young but not old Ghrl(-/-) mice, suggesting that differential compensatory responses resulting from the absence of endogenous ghrelin is occurring according to age. These results show that endogenous ghrelin is a regulator of GH pulse amplitude in growing mice but does not significantly modulate linear growth.


Subject(s)
Ghrelin/deficiency , Growth Hormone/metabolism , Mice/growth & development , Mice/metabolism , Protein Precursors/metabolism , Animals , Ghrelin/genetics , Male , Mice/genetics , Mice, Inbred C57BL , Mice, Knockout , Protein Precursors/genetics
12.
Am J Physiol Lung Cell Mol Physiol ; 306(3): L233-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285267

ABSTRACT

Acute lung injury (ALI) is a critical syndrome consisting of acute respiratory failure associated with extensive pulmonary infiltrates. The pathological characterization of ALI includes injuries of alveolar epithelial cells (AECs), alveolar neutrophilic infiltration, and increases in proinflammatory cytokines, which cause destruction of the alveolar capillary barrier and subsequent devastating lung fibrosis. Rikkunshito (RKT), a traditional Japanese herbal medicine, is widely used for the treatment of patients with gastrointestinal symptoms and is known to stimulate ghrelin secretion. The therapeutic effects of RKT on organ inflammation and fibrosis remain unknown. We investigated the pharmacological potential of RKT in the treatment of ALI by using a bleomycin-induced ALI model in mice. RKT or distilled water (DW) was given to mice daily starting 12 h after bleomycin administration. The RKT-treated mice showed a definitively higher survival rate than the DW-treated mice after injury. They also had smaller reductions in body weight and food intake. The amelioration of neutrophil alveolar infiltration, pulmonary vascular permeability, induction of proinflammatory cytokines, activation of the NF-κB pathway, apoptosis of AECs, and subsequent lung fibrosis were notable in the RKT-treated mice. RKT administration increased the plasma ghrelin levels in wild-type mice, and it also mitigated the ALI response in both ghrelin-deficient mice and growth hormone secretagogue receptor-deficient mice after lung injury. Our results indicate that RKT administration exerts protective effects against ALI by protecting the AECs and regulating lung inflammation independently of the ghrelin system, and they highlight RKT as a promising therapeutic agent for the management of this intractable disease.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Ghrelin/deficiency , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Apoptosis/drug effects , Bleomycin , Disease Models, Animal , Eating/drug effects , Epithelial Cells/drug effects , Epithelial Cells/physiology , Ghrelin/blood , Ghrelin/metabolism , Glycyrrhizic Acid/pharmacology , Hesperidin/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Neutrophil Infiltration/drug effects , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Fibrosis/drug therapy , Signal Transduction/drug effects
13.
Exp Neurol ; 252: 75-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24295570

ABSTRACT

Ghrelin has been shown to regulate neurogenesis in the hippocampus. The aim of this study was to investigate the possible influence of ghrelin on cell proliferation and neuroblast formation in the subventricular zone (SVZ) and rostral migratory system (RMS) and generation of interneurons in the olfactory bulb (OB). We found that ghrelin receptors were expressed in the SVZ-RMS-OB system. Ghrelin knockout (GKO) mice have fewer proliferating neural progenitor cells and neuroblasts in the SVZ, while ghrelin administration attenuated these changes. We also found that not only the number of BrdU-labeled cells but also the fraction of migratory neuroblasts in the RMS was decreased in the GKO mice compared with controls. Treatment of GKO mice with ghrelin restored these numbers to the wild-type control values. Far fewer BrdU/NeuN double-labeled cells were found in the OB of GKO mice than in wild-type mice 4 weeks after labeling, which were increased by ghrelin replacement. GKO mice showed less numbers of BrdU/calbindin, BrdU/calretinin and BrdU/tyrosine hydroxylase double-labeled cells in the periglomerular layer of the OB. However, these numbers were increased to wild-type values after ghrelin administration. Finally, in the GH-deficient spontaneous dwarf rats, ghrelin increased the number of progenitor cells and neuroblasts in the SVZ, without significant effect on the differentiation in the OB. These findings suggest that ghrelin is involved in the regulation of proliferation of progenitor cells in the SVZ, the number of migratory neuroblasts in the SVZ, and the differentiation of interneurons in the OB.


Subject(s)
Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Ghrelin/pharmacology , Lateral Ventricles/cytology , Neural Stem Cells/drug effects , Animals , Bromodeoxyuridine/metabolism , Calbindin 2/metabolism , Calbindins/metabolism , Cell Movement/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Ghrelin/deficiency , Ghrelin/genetics , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurons/drug effects , Olfactory Bulb/cytology , Receptors, Ghrelin/metabolism , Sialic Acids/metabolism , Tyrosine 3-Monooxygenase/metabolism
14.
J Am Heart Assoc ; 2(6): e000376, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24308935

ABSTRACT

BACKGROUND: Surgical treatment of peripheral artery disease, even if successful, does not prevent reoccurrence. Under these conditions, increased oxidative stress is a crucial determinant of tissue damage. Given its reported antioxidant effects, we investigated the potential of unacylated-ghrelin (UnAG) to reduce ischemia-induced tissue damage in a mouse model of peripheral artery disease. METHODS AND RESULTS: We show that UnAG but not acylated ghrelin (AG) induces skeletal muscle regeneration in response to ischemia via canonical p38/mitogen-actived protein kinase signaling UnAG protected against reactive oxygen species-induced cell injuries by inducing the expression of superoxide dismutase-2 (SOD-2) in satellite cells. This led to a reduced number of infiltrating CD68(+) cells and was followed by induction of the myogenic process and a reduction in functional impairment. Moreover, we found that miR-221/222, previously linked to muscle regeneration processes, was up-regulated and negatively correlated with p57(Kip2) expression in UnAG-treated mice. UnAG, unlike AG, promoted cell-cycle entry in satellite cells of mice lacking the genes for ghrelin and its receptor (GHSR1a). UnAG-induced p38/mitogen-actived protein kinase phosphorylation, leading to activation of the myogenic process, was prevented in SOD-2-depleted SCs. By siRNA technology, we also demonstrated that SOD-2 is the antioxidant enzyme involved in the control of miR-221/222-driven posttranscriptional p57(Kip2) regulation. Loss-of-function experiments targeting miR-221/222 and local pre-miR-221/222 injection in vivo confirmed a role for miR-221/222 in driving skeletal muscle regeneration after ischemia. CONCLUSIONS: These results indicate that UnAG-induced skeletal muscle regeneration after ischemia depends on SOD-2-induced miR-221/222 expression and highlight its clinical potential for the treatment of reactive oxygen species-mediated skeletal muscle damage.


Subject(s)
Antioxidants/pharmacology , Ghrelin/pharmacology , Ischemia/drug therapy , MicroRNAs/metabolism , Muscle, Skeletal/drug effects , Regeneration/drug effects , Superoxide Dismutase/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Disease Models, Animal , Ghrelin/analogs & derivatives , Ghrelin/deficiency , Ghrelin/genetics , Hindlimb , Ischemia/enzymology , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , MyoD Protein/metabolism , Oxidative Stress/drug effects , PAX7 Transcription Factor/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/enzymology , Signal Transduction/drug effects , Superoxide Dismutase/genetics , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
15.
PLoS One ; 8(10): e76553, 2013.
Article in English | MEDLINE | ID: mdl-24124572

ABSTRACT

Taste perception plays an important role in regulating food preference, eating behavior and energy homeostasis. Taste perception is modulated by a variety of factors, including gastric hormones such as ghrelin. Ghrelin can regulate growth hormone release, food intake, adiposity, and energy metabolism. Octanoylation of ghrelin by ghrelin O-acyltransferase (GOAT) is a specific post-translational modification which is essential for many biological activities of ghrelin. Ghrelin and GOAT are both widely expressed in many organs including the gustatory system. In the current study, overall metabolic profiles were assessed in wild-type (WT), ghrelin knockout (ghrelin(-/-)), and GOAT knockout (GOAT(-/-)) mice. Ghrelin(-/-) mice exhibited decreased food intake, increased plasma triglycerides and increased ketone bodies compared to WT mice while demonstrating WT-like body weight, fat composition and glucose control. In contrast GOAT(-/-) mice exhibited reduced body weight, adiposity, resting glucose and insulin levels compared to WT mice. Brief access taste behavioral tests were performed to determine taste responsivity in WT, ghrelin(-/-) and GOAT(-/-) mice. Ghrelin and GOAT null mice possessed reduced lipid taste responsivity. Furthermore, we found that salty taste responsivity was attenuated in ghrelin(-/-) mice, yet potentiated in GOAT(-/-) mice compared to WT mice. Expression of the potential lipid taste regulators Cd36 and Gpr120 were reduced in the taste buds of ghrelin and GOAT null mice, while the salt-sensitive ENaC subunit was increased in GOAT(-/-) mice compared with WT mice. The altered expression of Cd36, Gpr120 and ENaC may be responsible for the altered lipid and salt taste perception in ghrelin(-/-) and GOAT(-/-) mice. The data presented in the current study potentially implicates ghrelin signaling activity in the modulation of both lipid and salt taste modalities.


Subject(s)
Acyltransferases/genetics , Ghrelin/genetics , Lipids , Salts , Taste Perception/genetics , Acyltransferases/deficiency , Animals , Body Composition , Feeding Behavior , Gene Expression Regulation , Ghrelin/deficiency , Hormones/metabolism , Ketone Bodies/blood , Male , Membrane Proteins , Mice , Mice, Knockout , Phenotype , Taste Buds/anatomy & histology , Taste Buds/metabolism , Triglycerides/blood
16.
Gastroenterology ; 144(1): 50-52.e5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22995675

ABSTRACT

Reductions in levels of the hunger-stimulating hormone ghrelin have been proposed to mediate part of the effects of vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass surgeries for obesity. We studied circulating levels of acyl and desacyl ghrelin in rats after these surgeries. We found that blood levels of ghrelin were reduced after VSG, but not after Roux-en-Y gastric bypass, based on enzyme-linked immunosorbent assay and mass-spectrometry analyses. We compared the effects of VSG in ghrelin-deficient mice and wild-type mice on food intake, body weight, dietary fat preference, and glucose tolerance. We found that VSG produced comparable outcomes in each strain. Reduced ghrelin signaling therefore does not appear to be required for these effects of VSG.


Subject(s)
Eating , Feeding Behavior , Gastrectomy , Ghrelin/blood , Animals , Body Weight , Dietary Fats , Genotype , Ghrelin/deficiency , Ghrelin/genetics , Glucose Tolerance Test , Male , Mice , Mice, Knockout , Rats , Rats, Long-Evans , Signal Transduction
17.
Am J Physiol Endocrinol Metab ; 303(3): E422-31, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22669248

ABSTRACT

The orexigenic hormone ghrelin is important in diabetes because it has an inhibitory effect on insulin secretion. Ghrelin ablation in leptin-deficient ob/ob (Ghrelin(-/-):ob/ob) mice increases insulin secretion and improves hyperglycemia. The physiologically relevant ghrelin receptor is the growth hormone secretagogue receptor (GHS-R), and GHS-R antagonists are thought to be an effective strategy for treating diabetes. However, since some of ghrelin's effects are independent of GHS-R, we have utilized genetic approaches to determine whether ghrelin's effect on insulin secretion is mediated through GHS-R and whether GHS-R antagonism indeed inhibits insulin secretion. We investigated the effects of GHS-R on glucose homeostasis in Ghsr-ablated ob/ob mice (Ghsr(-/-):ob/ob). Ghsr ablation did not rescue the hyperphagia, obesity, or insulin resistance of ob/ob mice. Surprisingly, Ghsr ablation worsened the hyperglycemia, decreased insulin, and impaired glucose tolerance. Consistently, Ghsr ablation in ob/ob mice upregulated negative ß-cell regulators (such as UCP-2, SREBP-1c, ChREBP, and MIF-1) and downregulated positive ß-cell regulators (such as HIF-1α, FGF-21, and PDX-1) in whole pancreas; this suggests that Ghsr ablation impairs pancreatic ß-cell function in leptin deficiency. Of note, Ghsr ablation in ob/ob mice did not affect the islet size; the average islet size of Ghsr(-/-):ob/ob mice is similar to that of ob/ob mice. In summary, because Ghsr ablation in leptin deficiency impairs insulin secretion and worsens hyperglycemia, this suggests that GHS-R antagonists may actually aggravate diabetes under certain conditions. The paradoxical effects of ghrelin ablation and Ghsr ablation in ob/ob mice highlight the complexity of the ghrelin-signaling pathway.


Subject(s)
Ghrelin/genetics , Glucose/metabolism , Leptin/genetics , Obesity/genetics , Obesity/metabolism , Receptors, Ghrelin/genetics , Animals , Gene Deletion , Ghrelin/deficiency , Ghrelin/physiology , Glucose Tolerance Test , Homeostasis/genetics , Homeostasis/physiology , Hyperglycemia/etiology , Hyperglycemia/genetics , Leptin/deficiency , Leptin/physiology , Male , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/complications , Receptors, Ghrelin/physiology
18.
Neuroscience ; 218: 12-9, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22641088

ABSTRACT

Ghrelin is an orexigenic stomach peptide previously found to be important for the full display of anticipatory locomotor activity and hypothalamic neuronal activation that precedes a daily scheduled meal in mice. Ghrelin is also important for food-related motivation and seems to have direct effects in the mesocorticolimbic dopamine reward system. Here we hypothesized that neuronal activation in reward-related areas in anticipation of a scheduled meal could be mediated by elevated ghrelin induced by scheduled feeding, and therefore this would be attenuated in ghrelin receptor knock-out (GHSR KO) animals. We found that this was indeed the case for the ventral tegmental area and the shell, but not the core, of the nucleus accumbens. In addition, our results show a reduction in the proportion of activated orexin-immunoreactive (IR) neurons in GHSR KO animals in anticipation of the scheduled meal in comparison to the proportion of activated orexin neurons in wild type (WT) mice. Interestingly we observed that both GHSR and ghrelin KO mice had fewer orexin-IR cells than their WT littermates suggesting that lack of ghrelin or sensitivity to ghrelin may play a role in the development of the orexin system. Our data also suggest that ghrelin may mediate food anticipation, in part, by stimulating both the orexin system and the mesolimbic reward system.


Subject(s)
Feeding Behavior/physiology , Ghrelin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Dopamine/metabolism , Food Deprivation , Ghrelin/deficiency , Hypothalamic Area, Lateral/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Neural Pathways/metabolism , Orexins , Reward , Transcriptome
19.
Endocrinol Nutr ; 58(8): 422-32, 2011 Oct.
Article in Spanish | MEDLINE | ID: mdl-21824829

ABSTRACT

Obesity is associated to significant disturbances in endocrine function. Hyper insulinemia and insulin resistance are the best known changes in obesity, but their mechanisms and clinical significance are not clearly established. Adipose tissue is considered to be a hormone-secreting endocrine organ; and increased leptin secretion from the adipocyte, a satiety signal, is a well-established endocrine change in obesity. In obesity there is a decreased GH secretion. Impairment of somatotropic function in obesity is functional and may be reversed in certain circumstances. The pathophysiological mechanism responsible for low GH secretion in obesity is probably multifactorial. There are many data suggesting that a chronic state of somatostatin hypersecretion results in inhibition of GH release. Increased FFA levels, as well as a deficient ghrelin secretion, probably contribute to the impaired GH secretion. In women, abdominal obesity is associated to hyperandrogenism and low sex hormone-binding globulin levels. Obese men, particularly those with morbid obesity, have decreased testosterone and gonadotropin levels. Obesity is associated to an increased cortisol production rate, which is compensated for by a higher cortisol clearance, resulting in plasma free cortisol levels that do not change when body weight increases. Ghrelin is the only known circulating orexigenic factor, and has been found to be decreased in obese people. In obesity there is also a trend to increased TSH and free T3 levels.


Subject(s)
Endocrine System/physiopathology , Obesity/physiopathology , Adiponectin/metabolism , Adipose Tissue/metabolism , Female , Ghrelin/deficiency , Gonadal Steroid Hormones/blood , Human Growth Hormone/metabolism , Humans , Hydrocortisone/metabolism , Hyperinsulinism/etiology , Leptin/metabolism , Male , Sex Hormone-Binding Globulin/analysis , Somatostatin/metabolism , Thyroid Hormones/blood
20.
Peptides ; 32(11): 2229-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21781995

ABSTRACT

Ghrelin is a hormone made in the stomach and known primarily for its growth hormone releasing and orexigenic properties. Nevertheless, ghrelin through its receptor, the GHS-R1a, has been shown to exert many roles including regulation of glucose homeostasis, memory & learning, food addiction and neuroprotection. Furthermore, ghrelin could promote overall health and longevity by acting directly in the immune system and promoting an extended antigen repertoire. The development of mice lacking either ghrelin (ghrelin-/-) or its receptor (ghsr-/-) have provided a valuable tool for determining the relevance of ghrelin and its receptor in these multiple and diverse roles. In this review, we summarize the most important findings and lessons learned from the ghrelin-/- and ghsr-/- mice.


Subject(s)
Central Nervous System/physiology , Energy Metabolism/physiology , Gastric Mucosa/metabolism , Ghrelin/deficiency , Neuroprotective Agents/metabolism , Receptors, Ghrelin/deficiency , Signal Transduction/physiology , Animals , Dopaminergic Neurons/metabolism , Female , Ghrelin/genetics , Glucose/metabolism , Humans , Learning , Leptin/deficiency , Leptin/genetics , Longevity , Male , Memory , Mice , Mice, Knockout , Models, Animal , Receptors, Ghrelin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...