Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Endocrinology ; 164(11)2023 09 23.
Article in English | MEDLINE | ID: mdl-37823477

ABSTRACT

The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.


Subject(s)
Corticotropin-Releasing Hormone , Ghrelin , Mice , Male , Animals , Corticotropin-Releasing Hormone/metabolism , Ghrelin/pharmacology , Ghrelin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Neurons/metabolism
2.
BMC Complement Med Ther ; 23(1): 123, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37069587

ABSTRACT

OBJECTIVE: This double-blind, placebo-controlled, clinical trial was conducted to define the effects of Nigella sativa (N. Sativa) powder plus conventional medical treatment of Helicobacter pylori (H. pylori) on serum ghrelin level and appetite in H. pylori-infected patients. METHODS: In the present study, 51 H. pylori-positive patients were randomly allocated to treatment (n = 26) or placebo (n = 25) groups. They received 2 g/day N. Sativa with quadruple therapy or 2 g/day placebo plus quadruple therapy for 8 weeks. The serum level of ghrelin was assessed before and after the intervention. Appetite was evaluated at the onset and at the end of the intervention. RESULTS: At the end of the study, the appetite of the treatment group improved significantly compared with the placebo group (P = 0.02). Statistically, the difference in serum ghrelin levels between the study's groups was insignificant (P > 0.05). CONCLUSION: Supplementation with N. Sativa powder may be a beneficial adjunctive therapy in H. pylori-infected patients. TRIAL REGISTRATION: This study was registered in the Iranian Registry of Clinical Trials (IRCT20170916036204N7) on 08/08/2018.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Nigella sativa , Humans , Ghrelin/pharmacology , Ghrelin/therapeutic use , Powders/pharmacology , Powders/therapeutic use , Helicobacter Infections/drug therapy , Iran , Double-Blind Method
3.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2637-2650, 2023 10.
Article in English | MEDLINE | ID: mdl-37097336

ABSTRACT

This study is to observe the upregulation effect of astragaloside IV on ghrelin in diabetic cognitive impairment (DCI) rats and to investigate the pathway in prevention and treatment by reducing oxidative stress. The DCI model was induced with streptozotocin (STZ) in conjunction with a high-fat and high-sugar diet and divided into three groups: model, low-dose (40 mg/kg), and high-dose (80 mg/kg) astragaloside IV. After 30 days of gavage, the learning and memory abilities of rats, as well as their body weight and blood glucose levels, were tested using the Morris water maze and then detection of insulin resistance, SOD activity, and serum MDA levels. The whole brain of rats was sampled for hematoxylin-eosin and Nissl staining to observe pathological changes in the hippocampal CA1 region. Immunohistochemistry was used to detect ghrelin expression in the hippocampal CA1 region. A Western blot was used to determine changes in GHS-R1α/AMPK/PGC-1α/UCP2. RT-qPCR was used to determine the levels of ghrelin mRNA. Astragaloside IV reduced nerve damage, increased superoxide dismutase (SOD) activity, decreased MDA levels, and improved insulin resistance. Ghrelin levels and expression increased in serum and hippocampal tissues, and ghrelin mRNA levels increased in rat stomach tissues. According to Western blot, it increased the expression of the ghrelin receptor GHS-R1α and upregulated the mitochondrial function associated-protein AMPK-PGC-1α-UCP2. Astragaloside IV increases ghrelin expression in the brain to reduce oxidative stress and delay diabetes-induced cognitive impairment. It may be related to the promotion of ghrelin mRNA levels.


Subject(s)
Cognitive Dysfunction , Diabetes Mellitus, Type 2 , Insulin Resistance , Rats , Animals , Up-Regulation , AMP-Activated Protein Kinases , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Ghrelin/pharmacology , Oxidative Stress , Cognitive Dysfunction/drug therapy , Superoxide Dismutase-1
4.
Neuroendocrinology ; 113(1): 64-79, 2023.
Article in English | MEDLINE | ID: mdl-35908540

ABSTRACT

INTRODUCTION: Ghrelin regulates a variety of functions by acting in the brain. The targets of ghrelin in the mouse brain have been mainly mapped using immunolabeling against c-Fos, a transcription factor used as a marker of cellular activation, but such analysis has several limitations. Here, we used positron emission tomography in mice to investigate the brain areas responsive to ghrelin. METHODS: We analyzed in male mice the brain areas responsive to systemically injected ghrelin using positron emission tomography imaging of 18F-fluoro-2-deoxyglucose (18F-FDG) uptake, an indicator of metabolic rate. Additionally, we studied if systemic administration of fluorescent ghrelin or native ghrelin displays symmetric accessibility or induction of c-Fos, respectively, in the brain of male mice. RESULTS: Ghrelin increased 18F-FDG uptake in few specific areas of the isocortex, striatum, pallidum, thalamus, and midbrain at 0-10-min posttreatment. At the 10-20 and 20-30 min posttreatment, ghrelin induced mixed changes in 18F-FDG uptake in specific areas of the isocortex, striatum, pallidum, thalamus, and midbrain, as well as in areas of the olfactory areas, hippocampal and retrohippocampal regions, hypothalamus, pons, medulla, and even the cerebellum. Ghrelin-induced changes in 18F-FDG uptake were transient and asymmetric. Systemically administrated fluorescent-ghrelin-labeled midline brain areas known to contain fenestrated capillaries and the hypothalamic arcuate nucleus, where a symmetric labeling was observed. Ghrelin treatment also induced a symmetric increased c-Fos labeling in the arcuate nucleus. DISCUSSION/CONCLUSION: Systemically injected ghrelin transiently and asymmetrically affects the metabolic activity of the brain of male mice in a wide range of areas, in a food intake-independent manner. The neurobiological bases of such asymmetry seem to be independent of the accessibility of ghrelin into the brain.


Subject(s)
Fluorodeoxyglucose F18 , Ghrelin , Mice , Male , Animals , Ghrelin/pharmacology , Ghrelin/metabolism , Brain/metabolism , Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/metabolism
5.
Chin J Integr Med ; 29(6): 490-499, 2023 Jun.
Article in English | MEDLINE | ID: mdl-35881212

ABSTRACT

OBJECTIVE: To investigate whether meranzin hydrate (MH) can alleviate depression-like behavior and hypomotility similar to Chaihu Shugan Powder (CSP), and further explore the potential common mechanisms. METHODS: Totally 120 Spraque-Dawley rats were randomly divided into 5-8 groups including sham, vehicle, fluoxetine (20 mg/kg), mosapride (10 mg/kg), CSP (30 g/kg), MH (9.18 mg/kg), [D-Lys3]-GHRP-6 (Dlys, 0.5 mg/kg), and MH+Dlys groups by a random number table, 8 rats in each group. And 32 mice were randomly divided into wild-type, MH (18 mg/kg), growth hormone secretagogue receptor-knockout (GHSR-KO), and GHSR+MH groups, 8 mice in each group. The forced swimming test (FST), open field test (OFT), tail suspension test (TST), gastric emptying (GE) test, and intestinal transit (IT) test were used to assess antidepressant and prokinetic (AP) effects after drug single administration for 30 min with absorbable identification in rats and mice, respectively. The protein expression levels of brain-derived neurotrophic factor (BDNF) and phosphorylated mammalian target of rapamycin (p-mTOR) in the hippocampus of rats were evaluated by Western blot. The differences in functional brain changes were determined via 7.0 T functional magnetic resonance imaging-blood oxygen level-dependent (fMRI-BOLD). RESULTS: MH treatment improved depression-like behavior (FST, OFT) and hypomotility (GE, IT) in the acute forced swimming (FS) rats (all P<0.05), and the effects are similar to the parent formula CSP. The ghrelin antagonist [D-Lys3]-GHRP-6 inhibited the effect of MH on FST and GE (P<0.05). Similarly, MH treatment also alleviated depression-like behavior (FST, TST) in the wild-type mice, however, no effects were found in the GHSR KO mice. Additionally, administration of MH significantly stimulated BDNF and p-mTOR protein expressions in the hippocampus (both P<0.01), which were also prevented by [D-Lys3]-GHRP-6 (P<0.01). Besides, 3 main BOLD foci following acute FS rats implicated activity in hippocampus-thalamus-basal ganglia (HTB) circuits. The [D-Lys3]-GHRP-6 synchronously inhibited BOLD HTB foci. As expected, prokinetic mosapride only had effects on the thalamus and basal ganglia, but not on the hippocampus. Within the HTB, the hippocampus is implicated in depression and FD. CONCLUSIONS: MH accounts for part of AP effects of parent formula CSP in acute FS rats, mainly via ghrelin-related shared regulation coupled to BOLD signals in brain areas. This novel functionally connection of HTB following acute stress, treatment, and regulation highlights anti-depression unified theory.


Subject(s)
Brain-Derived Neurotrophic Factor , Ghrelin , Rats , Mice , Animals , Brain-Derived Neurotrophic Factor/metabolism , Ghrelin/pharmacology , Ghrelin/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Hippocampus , Stress, Psychological , Mammals/metabolism
6.
Reprod Fertil Dev ; 34(18): 1128-1134, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36309990

ABSTRACT

CONTEXT: The role of metabolic hormones, medicinal plants and their interrelationships in the control of human reproductive processes are poorly understood. AIMS: To examine how leptin, obestatin and ginkgo (Ginkgo biloba L.) affect human ovarian hormone release. METHODS: We analysed the influence of leptin and obestatin alone and in combination with ginkgo extract on cultured human ovarian granulosa cells. The release of progesterone (P), insulin-like growth factor I (IGF-I), oxytocin (OT) and prostaglandin F (PGF) were analysed by enzyme immunoassay and enzyme-linked immunosorbent assay. KEY RESULTS: Leptin addition promoted the release of all the measured hormones. Obestatin stimulated the release of P, IGF-I and OT and inhibited PGF output. Ginkgo suppressed P, IGF-I and OT and promoted PGF release. Furthermore, ginkgo changed the stimulatory action of leptin on PGF to an inhibitory one. CONCLUSIONS: Leptin and obestatin are involved in the control of human ovarian hormone release and ginkgo influences their function. IMPLICATIONS: Leptin and obestatin could be useful as stimulators of human ovarian cell functions. The suppressive influence of ginkgo on ovarian function should lead to the development of ginkgo-containing drugs.


Subject(s)
Ghrelin , Ginkgo biloba , Granulosa Cells , Leptin , Plant Preparations , Female , Humans , Cells, Cultured , Ghrelin/pharmacology , Ginkgo biloba/chemistry , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/pharmacology , Progesterone/metabolism , Prostaglandins F/metabolism , Plant Preparations/pharmacology
7.
Psychoneuroendocrinology ; 144: 105862, 2022 10.
Article in English | MEDLINE | ID: mdl-35835020

ABSTRACT

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.


Subject(s)
Ghrelin , Receptors, Ghrelin , Adiponectin/metabolism , Animals , Ghrelin/metabolism , Ghrelin/pharmacology , Hypothalamus/metabolism , Interleukin-6/metabolism , Leptin/metabolism , Olanzapine/pharmacology , Rats , Receptors, Ghrelin/metabolism , Resistin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Valproic Acid/pharmacology , Weight Gain
8.
J Integr Med ; 20(5): 432-441, 2022 09.
Article in English | MEDLINE | ID: mdl-35850968

ABSTRACT

OBJECTIVE: To investigate the influence of electroacupuncture (EA) on ghrelin and the phosphoinositide 3-kinase/protein kinase B/endothelial nitric oxide synthase (PI3K/Akt/eNOS) signaling pathway in spontaneously hypertensive rats (SHRs). METHODS: Eight Wistar-Kyoto rats were used as the healthy blood pressure (BP) control (normal group), and 32 SHRs were randomized into model group, EA group, EA plus ghrelin group (EA + G group), and EA plus PF04628935 group (a potent ghrelin receptor blocker; EA + P group) using a random number table. Rats in the normal group and model group did not receive treatment, but were immobilized for 20 min per day, 5 times a week, for 4 continuous weeks. SHRs in the EA group, EA + G group and EA + P group were immobilized and given EA treatment in 20 min sessions, 5 times per week, for 4 weeks. Additionally, 1 h before EA, SHRs in the EA + G group and EA + P group were intraperitoneally injected with ghrelin or PF04628935, respectively, for 4 weeks. The tail-cuff method was used to measure BP. After the 4-week intervention, the rats were sacrificed by cervical dislocation, and pathological morphology of the abdominal aorta was observed using hematoxylin-eosin (HE) staining. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of ghrelin, nitric oxide (NO), endothelin-1 (ET-1) and thromboxane A2 (TXA2) in the serum. Isolated thoracic aortic ring experiment was performed to evaluate vasorelaxation. Western blot was used to measure the expression of PI3K, Akt, phosphorylated Akt (p-Akt) and eNOS proteins in the abdominal aorta. Further, quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to measure the relative levels of mRNA expression for PI3K, Akt and eNOS in the abdominal aorta. RESULTS: EA significantly reduced the systolic BP (SBP) and diastolic BP (DBP) (P < 0.05). HE staining showed that EA improved the morphology of the vascular endothelium to some extent. Results of ELISA indicated that higher concentrations of ghrelin and NO, and lower concentrations of ET-1 and TXA2 were presented in the EA group (P < 0.05). The isolated thoracic aortic ring experiment demonstrated that the vasodilation capacity of the thoracic aorta increased in the EA group. Results of Western blot and qRT-PCR showed that EA increased the abundance of PI3K, p-Akt/Akt and eNOS proteins, as well as expression levels of PI3K, Akt and eNOS mRNAs (P < 0.05). In the EA + G group, SBP and DBP decreased (P < 0.05), ghrelin concentrations increased (P < 0.05), and the concentrations of ET-1 and TXA2 decreased (P < 0.05), relative to the EA group. In addition, the levels of PI3K and eNOS proteins, the p-Akt/Akt ratio, and the expression of PI3K, Akt and eNOS mRNAs increased significantly in the EA + G group (P < 0.05), while PF04628935 reversed these effects. CONCLUSION: EA effectively reduced BP and protected the vascular endothelium, and these effects may be linked to promoting the release of ghrelin and activation of the PI3K/Akt/eNOS signaling pathway.


Subject(s)
Electroacupuncture , Nitric Oxide Synthase Type III , Animals , Ghrelin/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type III/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinase/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction
9.
Life Sci ; 301: 120636, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35568227

ABSTRACT

AIMS: The present study aims to compare the responses between male and female C57BL/6 mice to multiple metabolic challenges to understand the importance of sex in the control of energy homeostasis. MAIN METHODS: Male and female C57BL/6 mice were subjected to nutritional and hormonal challenges, such as food restriction and refeeding, diet-induced obesity, feeding response to ghrelin and leptin, ghrelin-induced growth hormone secretion, and central responsiveness to ghrelin and leptin. The hypothalamic expression of transcripts that control energy homeostasis was also evaluated. KEY FINDINGS: Male mice lost more weight and lean body mass in response to food restriction, compared to females. During refeeding, males accumulated more body fat and exhibited lower energy expenditure and glycemia, as compared to females. Additionally, female mice exhibited a higher protection against diet-induced obesity and related metabolic imbalances in comparison to males. Low dose ghrelin injection elicited higher food intake and growth hormone secretion in male mice, whereas the acute anorexigenic effect of leptin was more robust in females. However, the sex differences in the feeding responses to ghrelin and leptin were not explained by variations in the central responsiveness to these hormones nor by differences in the fiber density from arcuate nucleus neurons. Female, but not male, mice exhibited compensatory increases in hypothalamic Pomc mRNA levels in response to diet-induced obesity. SIGNIFICANCE: Our findings revealed several sexually differentiated responses to metabolic challenges in C57BL/6 mice, highlighting the importance of taking into account sex differences in metabolic studies.


Subject(s)
Ghrelin , Leptin , Animals , Female , Ghrelin/pharmacology , Growth Hormone/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism
10.
Journal of Integrative Medicine ; (12): 432-441, 2022.
Article in English | WPRIM | ID: wpr-939903

ABSTRACT

OBJECTIVE@#To investigate the influence of electroacupuncture (EA) on ghrelin and the phosphoinositide 3-kinase/protein kinase B/endothelial nitric oxide synthase (PI3K/Akt/eNOS) signaling pathway in spontaneously hypertensive rats (SHRs).@*METHODS@#Eight Wistar-Kyoto rats were used as the healthy blood pressure (BP) control (normal group), and 32 SHRs were randomized into model group, EA group, EA plus ghrelin group (EA + G group), and EA plus PF04628935 group (a potent ghrelin receptor blocker; EA + P group) using a random number table. Rats in the normal group and model group did not receive treatment, but were immobilized for 20 min per day, 5 times a week, for 4 continuous weeks. SHRs in the EA group, EA + G group and EA + P group were immobilized and given EA treatment in 20 min sessions, 5 times per week, for 4 weeks. Additionally, 1 h before EA, SHRs in the EA + G group and EA + P group were intraperitoneally injected with ghrelin or PF04628935, respectively, for 4 weeks. The tail-cuff method was used to measure BP. After the 4-week intervention, the rats were sacrificed by cervical dislocation, and pathological morphology of the abdominal aorta was observed using hematoxylin-eosin (HE) staining. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of ghrelin, nitric oxide (NO), endothelin-1 (ET-1) and thromboxane A2 (TXA2) in the serum. Isolated thoracic aortic ring experiment was performed to evaluate vasorelaxation. Western blot was used to measure the expression of PI3K, Akt, phosphorylated Akt (p-Akt) and eNOS proteins in the abdominal aorta. Further, quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to measure the relative levels of mRNA expression for PI3K, Akt and eNOS in the abdominal aorta.@*RESULTS@#EA significantly reduced the systolic BP (SBP) and diastolic BP (DBP) (P < 0.05). HE staining showed that EA improved the morphology of the vascular endothelium to some extent. Results of ELISA indicated that higher concentrations of ghrelin and NO, and lower concentrations of ET-1 and TXA2 were presented in the EA group (P < 0.05). The isolated thoracic aortic ring experiment demonstrated that the vasodilation capacity of the thoracic aorta increased in the EA group. Results of Western blot and qRT-PCR showed that EA increased the abundance of PI3K, p-Akt/Akt and eNOS proteins, as well as expression levels of PI3K, Akt and eNOS mRNAs (P < 0.05). In the EA + G group, SBP and DBP decreased (P < 0.05), ghrelin concentrations increased (P < 0.05), and the concentrations of ET-1 and TXA2 decreased (P < 0.05), relative to the EA group. In addition, the levels of PI3K and eNOS proteins, the p-Akt/Akt ratio, and the expression of PI3K, Akt and eNOS mRNAs increased significantly in the EA + G group (P < 0.05), while PF04628935 reversed these effects.@*CONCLUSION@#EA effectively reduced BP and protected the vascular endothelium, and these effects may be linked to promoting the release of ghrelin and activation of the PI3K/Akt/eNOS signaling pathway.


Subject(s)
Animals , Rats , Electroacupuncture , Ghrelin/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/pharmacology , Phosphatidylinositol 3-Kinase/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction
11.
Neuropeptides ; 89: 102180, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34293597

ABSTRACT

Appetite loss or anorexia substantially decreases the quality of life in patients with cancer, depression and gastrointestinal disorders, and can lead to sarcopenia and frailty. Foods that restore appetite have been sought-for but are not currently available. Historically, onion intake was adopted to treat a variety of diseases with reduced appetite including cancer and gastrointestinal disturbances. While isoalliin is a core component of onion, the effects of isoalliin on feeding behavior and feeding centers remain unknown. Neuropeptide Y (NPY) and ghrelin are the most potent central and peripheral inducers of appetite. A Japanese kampo medicine Ninjin'yoeito activates ghrelin-responsive NPY neurons in the hypothalamic arcuate nucleus (ARC) and counteracts anorexia induced by an anti-cancer drug cisplatin. This study explored the effects of isoalliin on feeding behavior and activities of ARC neurons in mice. Isoalliin, injected intraperitoneally, dose-dependently increased food intake during dark phase (DP) and daily without altering light phase (LP) food intake. We measured cytosolic Ca2+ concentration ([Ca2+]i) in single ARC neurons including NPY neurons identified by GFP fluorescence. Isoalliin increased [Ca2+]i in 10 of 18 (55.6%) NPY neurons, a majority of which also responded to ghrelin with [Ca2+]i increases, indicating that the ARC ghrelin-responsive NPY neuron is the major target of isoalliin. Isoalliin also increased [Ca2+]i in the ARC neurons that responded to Ninjin'yoeito. These results indicate that isoalliin enhances feeding at the active period and activates ARC ghrelin-responsive NPY neurons and Ninjin'yoeito-responsive neurons. These abilities of isoalliin to stimulate DP feeding and activate ARC orexigenic neurons provide scientific evidence for the health beneficial effects of onion experienced historically and globally.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Cysteine/analogs & derivatives , Drugs, Chinese Herbal/pharmacology , Eating/drug effects , Ghrelin/pharmacology , Neurons/drug effects , Neuropeptide Y/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Calcium/metabolism , Cysteine/pharmacology , Dose-Response Relationship, Drug , Mice
12.
Hum Exp Toxicol ; 40(12): 2178-2187, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34151639

ABSTRACT

Despite the various and newly developed chemotherapeutic agents in recent years, cisplatin is still used very frequently as a chemotherapeutic agent, even though cisplatin has toxic effects on many organs. The aim of our study is to show whether ghrelin reduces the liver toxicity of cisplatin in the rat model. Twenty-eight male Sprague Dawley albino mature rats were chosen to be utilized in the study. Group 1 rats (n = 7) were taken as the control group, and no medication was given to them. Group 2 rats (n = 7) received 5 mg/kg/day cisplatin and 1 ml/kg/day of 0.9% NaCl, Group 3 rats (n = 7) received 5 mg/kg/day cisplatin and 10 ng/kg/day ghrelin, Group 4 rats (n = 7) received 5 mg/kg/day cisplatin and 20 ng/kg/day ghrelin for 3 days. Glutathione, malondialdehyde (MDA), superoxide dismutase (SOD), plasma alanine aminotransferase (ALT) levels, and liver biopsy results were measured in rats. It was determined that, especially in the high-dose group, the MDA, plasma ALT, and SOD levels increased less in the ghrelin group as compared to the cisplatin group, and the glutathione level decreased slightly with a low dose of ghrelin, while it increased with a higher dose. In histopathological examination, it was determined that the toxic effect of cisplatin on the liver was reduced with a low dose of ghrelin, and its histopathological appearance was similar to normal liver tissue when given a high dose of ghrelin. These findings show that ghrelin, especially in high doses, can be used to reduce the toxic effect of cisplatin.


Subject(s)
Antineoplastic Agents/toxicity , Chemical and Drug Induced Liver Injury/drug therapy , Cisplatin/toxicity , Ghrelin/therapeutic use , Protective Agents/therapeutic use , Alanine Transaminase/blood , Animals , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Ghrelin/pharmacology , Glutathione/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/metabolism , Protective Agents/pharmacology , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
13.
Physiol Res ; 70(2): 237-243, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33992047

ABSTRACT

The objective of this study was to examine the direct effects of the medicinal plant fennel (Foeniculumvulgare Mill.) on basic functions of ovarian cells, including proliferation, apoptosis, and response to the physiological hormonal stimulator, ghrelin. In the first series of experiments, porcine ovarian granulosa cells were cultured with (1, 10, 100 µg/ml) or without fennel extract. In the second series of experiments, cells were cultured with (1, 10, 100 ng/ml) or without ghrelin, alone or in combination with fennel extract (10 µg/ml). Expression of the proliferation marker, PCNA, and the apoptosis marker, bax, were analyzed via quantitative immunocytochemical methods. Fennel stimulated the accumulation of the proliferation marker, and suppressed the expression of the apoptosis marker. Ghrelin alone promoted proliferation and apoptosis of ovarian cells. The presence of fennel inhibited these ghrelin effects. These observations provide the first demonstration of (1) effects of fennel on farm animal reproduction, (2) direct effects of fennel on ovarian cells, (3) the ability of fennel to promote ovarian cell proliferation, to inhibit ovarian cell apoptosis, and to enhance the ovarian cell proliferation:apoptosis ratio. Furthermore, our results (4) confirm the involvement of ghrelin in the control of ovarian cell apoptosis and proliferation, and (5) demonstrate the ability of fennel to affect not only ovarian cell proliferation and apoptosis, but also to suppress the responses of ovarian cells to the upstream hormonal regulator ghrelin. Our results indicate the potential applicability of fennel as a bio-stimulator of farm animal reproduction.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Foeniculum , Ghrelin/pharmacology , Granulosa Cells/drug effects , Plant Extracts/pharmacology , Animals , Cells, Cultured , Female , Foeniculum/chemistry , Granulosa Cells/metabolism , Granulosa Cells/pathology , Plant Extracts/isolation & purification , Proliferating Cell Nuclear Antigen/metabolism , Sus scrofa , bcl-2-Associated X Protein/metabolism
14.
Neurobiol Learn Mem ; 179: 107409, 2021 03.
Article in English | MEDLINE | ID: mdl-33609738

ABSTRACT

Ghrelin (Gr) is an orexigenic peptide that acts via its specific receptor, GHSR-1a distributed throughout the brain, being mainly enriched in pituitary, cortex and hippocampus (Hp) modulating a variety of brain functions. Behavioral, electrophysiological and biochemical evidence indicated that Gr modulates the excitability and the synaptic plasticity in Hp. The present experiments were designed in order to extend the knowledge about the Gr effect upon structural synaptic plasticity since morphological and quantitative changes in spine density after Gr administration were analyzed "in vitro" and "in vivo". The results show that Gr administered to hippocampal cultures or stereotactically injected in vivo to Thy-1 mice increases the density of dendritic spines (DS) being the mushroom type highly increased in secondary and tertiary extensions. Spines classified as thin type were increased particularly in primary extensions. Furthermore, we show that Gr enhances selectively the expression of BDNF-mRNA species.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Ghrelin/pharmacology , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Pyramidal Cells/drug effects , RNA, Messenger/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Dendritic Spines/drug effects , Dendritic Spines/pathology , Hippocampus/cytology , Hippocampus/metabolism , Microscopy, Confocal , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , RNA, Messenger/metabolism , Rats
15.
Neuropeptides ; 87: 102134, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33639357

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by the loss of dopaminergic neurons in the substantia nigra and the deposition of Lewy bodies. Mitochondrial dysfunction, oxidative stress, and autophagy dysfunction are involved in the pathogenesis of PD. Ghrelin is a brain-gut peptide that has been reported that protected against 1-methyl-4-phenyl-1,2,3,6- tetrahydropyran (MPTP)/MPP+-induced toxic effects. In the present work, human neuroblastoma SH-SY5Y cells were exposed to rotenone as a PD model to explore the underlying mechanism of ghrelin. We found that ghrelin inhibited rotenone-induced cytotoxicity, mitochondrial dysfunction, and apoptosis by improving cell viability, increasing the ratio of red/green of JC-1, inhibiting the production of reactive oxidative species (ROS), and regulating Bcl-2, Bax, Cytochrome c, caspase-9, and caspase-3 expression. Besides, ghrelin promoted mitophagy accompanied by up-regulating microtubule-associated protein 1 Light Chain 3B-II/I(LC3B-II/I) and Beclin1 but decreasing the expression of p62. Moreover, ghrelin promoted PINK1/Parkin mitochondrial translocation. Additionally, we investigated that ghrelin activated the AMPK/SIRT1/PGC1α pathway and pharmacological inhibition of AMPK and SIRT1 abolished the cytoprotection of ghrelin, decreased the level of mitophagy, and PINK1/Parkin mitochondrial translocation. Taken together, our findings suggested that mitophagy and AMPK/SIRT1/PGC1α pathways were related to the cytoprotection of ghrelin. These findings provided novel insights into the underlying mechanisms of ghrelin, further mechanistic studies on preclinical and clinical levels are required to be conducted with ghrelin to avail and foresee it as a potential agent in the treatment and management of PD.


Subject(s)
Ghrelin/physiology , Mitochondria/drug effects , Mitophagy/physiology , Nerve Tissue Proteins/physiology , Rotenone/toxicity , Signal Transduction/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/physiology , Apoptosis/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Ghrelin/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neuroblastoma , Oxidative Stress/drug effects , Parkinson Disease , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Protein Kinases/metabolism , Protein Transport/drug effects , Reactive Oxygen Species , Rotenone/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/physiology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics
16.
Nat Prod Res ; 35(1): 57-62, 2021 Jan.
Article in English | MEDLINE | ID: mdl-31135190

ABSTRACT

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].


Subject(s)
Camellia sinensis/chemistry , Flavonoids/chemistry , Acylation , Flavonoids/metabolism , Ghrelin/chemistry , Ghrelin/pharmacology , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Molecular Structure , Plant Extracts/chemistry , Receptors, Ghrelin/metabolism , Spectrometry, Mass, Electrospray Ionization , Tea/chemistry
17.
Diabetes ; 70(1): 62-75, 2021 01.
Article in English | MEDLINE | ID: mdl-33115827

ABSTRACT

Circulating branched-chain amino acids (BCAAs) are elevated in obesity and diabetes, and recent studies support a causal role for BCAAs in insulin resistance and defective glycemic control. The physiological mechanisms underlying BCAA regulation are poorly understood. Here we show that insulin signaling in the mediobasal hypothalamus (MBH) of rats is mandatory for lowering plasma BCAAs, most probably by inducing hepatic BCAA catabolism. Insulin receptor deletion only in agouti-related protein (AgRP)-expressing neurons (AgRP neurons) in the MBH impaired hepatic BCAA breakdown and suppression of plasma BCAAs during hyperinsulinemic clamps in mice. In support of this, chemogenetic stimulation of AgRP neurons in the absence of food significantly raised plasma BCAAs and impaired hepatic BCAA degradation. A prolonged fasting or ghrelin treatment recapitulated designer receptors exclusively activated by designer drugs-induced activation of AgRP neurons and increased plasma BCAAs. Acute stimulation of vagal motor neurons in the dorsal motor nucleus was sufficient to decrease plasma BCAAs. Notably, elevated plasma BCAAs were associated with impaired glucose homeostasis. These findings suggest a critical role of insulin signaling in AgRP neurons for BCAA regulation and raise the possibility that this control may be mediated primarily via vagal outflow. Furthermore, our results provide an opportunity to closely examine the potential mechanistic link between central nervous system-driven BCAA control and glucose homeostasis.


Subject(s)
Agouti-Related Protein/metabolism , Amino Acids, Branched-Chain/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Neurons/metabolism , Amino Acids, Branched-Chain/blood , Animals , Blood Glucose/metabolism , Ghrelin/pharmacology , Glucose Clamp Technique , Hypothalamus/drug effects , Insulin Resistance/physiology , Male , Mice , Motor Neurons/metabolism , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Vagus Nerve/metabolism
18.
Phytomedicine ; 81: 153424, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33278782

ABSTRACT

BACKGROUD: Exposure to high-dose radiation, such as after a nuclear accident or radiotherapy, elicits severe intestinal damage and is associated with a high mortality rate. In treating patients exhibiting radiation-induced intestinal dysfunction, countermeasures to radiation are required. In principle, the cellular event underlying radiation-induced gastrointestinal syndrome is intestinal stem cell (ISC) apoptosis in the crypts. High-dose irradiation induces the loss of ISCs and impairs intestinal barrier function, including epithelial regeneration and integrity. Notch signaling plays a critical role in the maintenance of the intestinal epithelium and regulates ISC self-renewal. Ghrelin, a hormone produced mainly by enteroendocrine cells in the gastrointestinal tract, has diverse physiological and biological functions. PURPOSE: We investigate whether ghrelin mitigates radiation-induced enteropathy, focusing on its role in maintaining epithelial function. METHODS: To investigate the effect of ghrelin in radiation-induced epithelial damage, we analyzed proliferation and Notch signaling in human intestinal epithelial cell. And we performed histological analysis, inflammatory response, barrier functional assays, and expression of notch related gene and epithelial stem cell using a mouse model of radiation-induced enteritis. RESULTS: In this study, we found that ghrelin treatment accelerated the reversal of radiation-induced epithelial damage including barrier dysfunction and defective self-renewing property of ISCs by activating Notch signaling. Exogenous injection of ghrelin also attenuated the severity of radiation-induced intestinal injury in a mouse model. CONCLUSION: These data suggest that ghrelin may be used as a potential therapeutic agent for radiation-induced enteropathy.


Subject(s)
Ghrelin/pharmacology , Intestinal Diseases/drug therapy , Intestinal Mucosa/cytology , Receptors, Notch/metabolism , Stem Cells/radiation effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Humans , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Male , Mice, Inbred C57BL , Radiation Injuries , Radiation-Protective Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Stem Cells/drug effects , Stem Cells/pathology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/radiation effects
19.
Endocrinology ; 161(11)2020 11 01.
Article in English | MEDLINE | ID: mdl-32961558

ABSTRACT

Organophosphate flame retardants (OPFRs) are a class of chemicals that have become near ubiquitous in the modern environment. While OPFRs provide valuable protection against flammability of household items, they are increasingly implicated as an endocrine disrupting chemical (EDC). We previously reported that exposure to a mixture of OPFRs causes sex-dependent disruptions of energy homeostasis through alterations in ingestive behavior and activity in adult mice. Because feeding behavior and energy expenditure are largely coordinated by the hypothalamus, we hypothesized that OPFR disruption of energy homeostasis may occur through EDC action on melanocortin circuitry within the arcuate nucleus. To this end, we exposed male and female transgenic mice expressing green fluorescent protein in either neuropeptide Y (NPY) or proopiomelanocortin (POMC) neurons to a common mixture of OPFRs (triphenyl phosphate, tricresyl phosphate, and tris(1,3-dichloro-2-propyl)phosphate; each 1 mg/kg bodyweight/day) for 4 weeks. We then electrophysiologically examined neuronal properties using whole-cell patch clamp technique. OPFR exposure depolarized the resting membrane of NPY neurons and dampened a hyperpolarizing K+ current known as the M-current within the same neurons from female mice. These neurons were further demonstrated to have increased sensitivity to ghrelin excitation, which more potently reduced the M-current in OPFR-exposed females. POMC neurons from female mice exhibited elevated baseline excitability and are indicated in receiving greater excitatory synaptic input when exposed to OPFRs. Together, these data support a sex-selective effect of OPFRs to increase neuronal output from the melanocortin circuitry governing feeding behavior and energy expenditure, and give reason for further examination of OPFR impact on human health.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Flame Retardants/pharmacology , Ghrelin/pharmacology , Melanocortins/metabolism , Nerve Net/drug effects , Neurons/drug effects , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Drug Resistance/drug effects , Endocrine Disruptors/pharmacology , Female , Hypothalamus/metabolism , Male , Mice , Mice, Transgenic , Nerve Net/physiology , Neurons/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Organophosphates/pharmacology , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
20.
Neurosci Lett ; 737: 135306, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32822766

ABSTRACT

Fasting increases ghrelin that is a peptide hormone with two circulating isoforms, acyl and des-acyl ghrelin. We reported that fasting or des-acyl ghrelin facilitates behavioral thermoregulation in the cold in rats assessed by tail-hiding behavior that was the indicator of rats' thermoregulatory behavior in the cold; however, the effect of acyl-ghrelin on the same process remains to be elucidated. We investigated the effect of acyl-ghrelin on thermoregulatory behavior in the cold in rats. The animals received an intraperitoneal saline or 24 µg acyl-ghrelin injection and were exposed to 27 °C or 15 °C for 2 h, while their body temperature, tail skin temperature, and tail-hiding behavior were constantly monitored. cFos immunoreactive (cFos-IR) cells in the median preoptic area, medial preoptic area, paraventricular nucleus (PVN), and arcuate nucleus were counted. Body temperature and the duration of thermoregulatory behavior did not show a significant difference between the acyl-ghrelin-treated and control groups at 15 °C; however, tail skin temperature in the acyl-ghrelin-treated group was higher than that in the control group. The number of cFos-IR cells in the PVN was greater in the control group than that in the acyl-ghrelin-treated group at 27 °C. These results indicate that acyl-ghrelin did not affect behavioral thermoregulation but might affect tail skin temperature in rats in the cold.


Subject(s)
Body Temperature Regulation/drug effects , Ghrelin/analogs & derivatives , Hypothalamus/drug effects , Skin Temperature/drug effects , Animals , Behavior, Animal/drug effects , Cold Temperature , Ghrelin/pharmacology , Hypothalamus/metabolism , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Tail
SELECTION OF CITATIONS
SEARCH DETAIL