Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Front Pharmacol ; 13: 824745, 2022.
Article in English | MEDLINE | ID: mdl-35401184

ABSTRACT

Purpose: To investigate the association between long-term use of aspirin and age-related macular degeneration (AMD). Methods: An updated systematic literature search was conducted in PubMed, Medline, Cochrane Library, and embase from conception to February 26, 2021, without any language restriction. All studies that evaluated the relationship between long-term aspirin use and AMD were included. Results: In the current study, 16 articles were pooled. Overall, no significant association was observed (estimate ratio = 1.108, 95% confidence interval (CI): 0.886-1.385). When the subgroups were evaluated according to various standards, aspirin use was significantly correlated with AMD in studies with volunteer participants (estimate ratio = 0.899, 95% CI: 0.830-0.974, p < 0.01), studies followed up for >10 years (estimate ratio = 2.206, 95% CI: 2.124-2.292, p < 0.01), duration of aspirin use >10 years (estimate ratio = 2.323, 95% CI: 2.234-2.416, p < 0.01), and cohort studies (estimate ratio = 1.961, 95% CI: 1.893-2.032, p < 0.01). Conclusion: Therefore, the association of aspirin and AMD can be demonstrated with a long-term follow-up or aspirin use, appropriate study design and participant source. The findings in our study might provide practical information on intervention strategies.

2.
Endocr Connect ; 9(12): 1168-1177, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33112816

ABSTRACT

Ghrelin plays a pivotal role in the regulation of food intake, body weight and energy metabolism. However, these effects of ghrelin in the lateral parabrachial nucleus (LPBN) are unexplored. C57BL/6J mice and GHSR-/- mice were implanted with cannula above the right LPBN and ghrelin was microinjected via the cannula to investigate effect of ghrelin in the LPBN. In vivo electrophysiological technique was used to record LPBN glucose-sensitive neurons to explore potential udnderlying mechanisms. Microinjection of ghrelin in LPBN significantly increased food intake in the first 3 h, while such effect was blocked by [D-Lys3]-GHRP-6 and abolished in GHSR-/- mice. LPBN ghrelin microinjection also significantly increased the firing rate of glucose-excited (GE) neurons and decreased the firing rate of glucose-inhibited (GI) neurons. Additionally, LPBN ghrelin microinjection also significantly increased c-fos expression. Chronic ghrelin administration in the LPBN resulted in significantly increased body weight gain. Meanwhile, no significant changes were observed in both mRNA and protein expression levels of UCP-1 in BAT. These results demonstrated that microinjection of ghrelin in LPBN could increase food intake through the interaction with growth hormone secretagogue receptor (GHSR) in C57BL/6J mice, and its chronic administration could also increase body weight gain. These effects might be associated with altered firing rate in the GE and GI neurons.

3.
Mater Sci Eng C Mater Biol Appl ; 93: 846-852, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30274120

ABSTRACT

The combination of biologically active compounds and nanomaterials in biomedicine field is growing rapidly and provided excellent forecasts for the progress of facile non-invasive approaches for the diagnosis of cancer therapy. In this present study, we demonstrated that as-prepared gallic acid (GA) coated reduced graphene oxide (rGO) combined with radiofrequency (RF) ablation has facilitated for the treatment of human renal epithelial cancer (A-489) cells without disturbing the renal proximal epithelial (HK-2) cells. The successful biofabrication of GA onto the rGO nanostructure has been elaborated with the support of peak intensities of diffracted X-ray patterns and Raman spectral visualizations. The micrograph imageries are displayed that the warped and ribbed structure containing wrinkled paper-like arrangement with multilayer structure of rGO with assistance of GA molecules. The synthesized GA-rGO nanomaterials exhibited a very good apoptosis and toxic effect on A-489 renal cancer cells, which was found to exhibit enhanced tumor cytotoxicity in RF combination treatment compared to RF treated alone. Moreover, the results are highly cheering to go for functionalized nanomaterial and RF radiation, combined drug delivery system to overcome the limitation to treat kidney cancers.


Subject(s)
Carcinoma, Renal Cell/therapy , Gallic Acid/chemistry , Graphite/chemistry , Kidney Neoplasms/therapy , Nanostructures , Radiofrequency Therapy , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Nanostructures/chemistry , Nanostructures/therapeutic use
4.
Int J Mol Med ; 41(3): 1635-1642, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29328403

ABSTRACT

The aim of the present study was to determine whether arcuate nucleus (ARC) lesions affect the ghrelin level in the plasma and the stomach in monosodium glutamate (MSG)­treated mice. The aim of the present study was to investigate whether the ARC was destroyed in mice treated neonatally with MSG, and whether the ARC lesions affect the ghrelin level in the plasma and lipid mobilization in MSG­treated mice. The results revealed that MSG led to a marked reduction in ARC cresyl violet staining, tyrosine hydroxylase-immunoreactive (IR) neurons and neuropeptide Y­IR fibers, compared with saline controls. MSG­treated mice exhibited significantly increased body mass compared with saline controls, and MSG treatment did not prevent food deprivation­induced decrease in white adipose tissue mass compared with controls. Plasma ghrelin levels were significantly increased in MSG­treated mice that were fasted for 48 h, compared with the levels prior to fasting and re­feeding, and the preprandial peak of plasma ghrelin persisted in MSG­treated mice. In summary, the ARC was not found to be essential for food deprivation­induced lipid mobilization and preprandial peak in MSG­treated mice. However, this finding does not mean that ARC neurons do not contribute to food sensing and lipid mobilization under normal conditions, as compensatory mechanisms may have emerged after the ablation of ARC neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Ghrelin/blood , Sodium Glutamate/administration & dosage , Adipose Tissue, White/anatomy & histology , Adipose Tissue, White/drug effects , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/drug effects , Area Postrema/drug effects , Area Postrema/metabolism , Body Weight/drug effects , Cell Count , Feeding Behavior/drug effects , Gastric Mucosa/metabolism , Ghrelin/genetics , Ghrelin/metabolism , Mice , Organ Size/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium Glutamate/pharmacology , Stomach/drug effects
5.
Front Physiol ; 8: 235, 2017.
Article in English | MEDLINE | ID: mdl-28484396

ABSTRACT

Nesfatin-1, an 82-amino acid neuropeptide, has been shown to induce anorexia and energy expenditure. Food intake is decreased in ad libitum-fed rats following injections of nesfatin-1 into the lateral, third, or fourth ventricles of the brain. Although the lateral parabrachial nucleus (LPBN) is a key regulator of feeding behavior and thermogenesis, the role of nesfatin-1 in this structure has not yet been delineated. We found that intra-LPBN microinjections of nesfatin-1 significantly reduced nocturnal cumulative food intake and average meal sizes without affecting meal numbers in rats. Because glucose sensitive neurons are involved in glucoprivic feeding and glucose homeostasis, we examined the effect of nesfatin-1 on the excitability of LPBN glucosensing neurons. In vivo electrophysiological recordings from LPBN glucose sensitive neurons showed that nesfatin-1 (1.5 × 10-8 M) excited most of the glucose-inhibited neurons. Chronic administration of nesfatin-1 into the LPBN of rats reduced body weight gain and enhanced the expression of uncoupling protein 1 (UCP1) in brown adipose tissue (BAT) over a 10-day period. Furthermore, the effects of nesfatin-1 on food intake, body weight, and BAT were attenuated by treatment with the melanocortin antagonist SHU9119. These results demonstrate that nesfatin-1 in LPBN inhibited food intake, modulated excitability of glucosensing neurons and enhanced UCP1 expression in BAT via the melanocortin system.

6.
Biomed Res Int ; 2017: 2034691, 2017.
Article in English | MEDLINE | ID: mdl-29318141

ABSTRACT

Alpha-melanocyte-stimulating hormone (α-MSH) is processed from proopiomelanocortin (POMC) and acts on the melanocortin receptors, MC3 and MC4. α-MSH plays a key role in energy homeostasis. In the present study, to shed light on the mechanisms by which α-MSH exerts its anorectic effects, extracellular neuronal activity was recorded in the hypothalamus and the dorsal vagal complex (DVC) of anesthetized rats. We examined the impact of α-MSH on glucose-sensing neurons and gastric distension (GD) sensitive neurons. In the lateral hypothalamus (LHA), α-MSH inhibited 75.0% of the glucose-inhibited (GI) neurons. In the ventromedial nucleus (VMN), most glucose-sensitive neurons were glucose-excited (GE) neurons, which were mainly activated by α-MSH. In the paraventricular nucleus (PVN), α-MSH suppressed the majority of GI neurons and excited most GE neurons. In the DVC, among the 20 GI neurons examined for a response to α-MSH, 1 was activated, 16 were depressed, and 3 failed to respond. Nineteen of 24 GE neurons were activated by α-MSH administration. Additionally, among the 42 DVC neurons examined for responses to GD, 23 were excited (GD-EXC) and 19 were inhibited (GD-INH). Fifteen of 20 GD-EXC neurons were excited, whereas 11 out of 14 GD-INH neurons were suppressed by α-MSH. All these responses were abolished by pretreatment with the MC3/4R antagonist, SHU9119. In conclusion, the activity of glucose-sensitive neurons and GD-sensitive neurons in the hypothalamus and DVC can be modulated by α-MSH.


Subject(s)
Hypothalamus/physiology , Neurons/physiology , Synaptic Transmission/drug effects , Vagus Nerve/physiology , alpha-MSH/pharmacology , Animals , Female , Male , Rats , Rats, Wistar , Synaptic Transmission/physiology
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 36(3): 419-22, 2016 Mar.
Article in Chinese | MEDLINE | ID: mdl-27063175

ABSTRACT

OBJECTIVE: To compare the serum miR-663 levels in renal transplant patients with and without acute rejection (AR) and explore the role of miR-663 acute renal graft rejection. METHODS: Real time-PCR was used to determine serum miR-663 levels in renal transplant recipients with and without AR. MTT assay and Annexin V-FITC assay were employed to examine the viability and apoptosis of human renal glomerular endothelial cells (HRGEC) treated with a miR-663 mimic or a miR-663 inhibitor, and ELISA was performed to detect the expression of inflammation-related cytokines including IL-6, IFN-γ, CCL-2 and TNF-α in the cells. Transwell assay was used to examine the effect of miR-663 mimic and miR-663 inhibitor on the chemotactic capability of macrophages. RESULTS: Serum miR-663 level was significantly higher in renal transplant recipients with AR than in those without AR. The miR-663 mimic significantly inhibited the viability of HRGECs and increase the cell apoptosis rate, while miR-663 inhibitor suppressed the cell apoptosis. The miR-663 mimic increased the expression levels of inflammation-related cytokines and enhanced the chemotactic capability of macrophages. CONCLUSION: miR-663 might play important roles in acute renal graft rejection and may become a therapeutic target for treating AR.


Subject(s)
Graft Rejection/blood , Kidney Transplantation , MicroRNAs/blood , Apoptosis , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/cytology , Humans , Kidney Glomerulus/cytology , Macrophages/cytology , Macrophages/drug effects
8.
Sheng Li Xue Bao ; 68(1): 19-26, 2016 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-26915318

ABSTRACT

The present study was aimed to explore the effects of intraperitoneal injection of growth hormone releasing peptide-6 (GHRP-6), a ghrelin receptor agonist, on food intake and neuronal activity of feeding-related nuclei in the hypothalamus of NMRI mice. Accumulated amount of food intake was measured, and total number of c-fos immunoreactive neurons in arcuate nucleus (ARC), paraventricular nucleus (PVN) and supraoptic nucleus (SON) was counted by immunohistochemistry at 1, 3 and 6 h after the GHRP-6 injection. The results showed that GHRP-6 significantly increased the amount of food intake with a peak at 3 h after the GHRP-6 injection. Meanwhile, GHRP-6 could promote c-fos expression in the ARC and PVN independent of food intake, and the total number of c-fos immunoreactive neurons was peaked at 1 h after injection and then decreased gradually. These results suggest that GHRP-6 may increase food intake in time-dependent manner, which is associated with up-regulations of c-fos protein expression in the ARC and PVN.


Subject(s)
Arcuate Nucleus of Hypothalamus , Neurons , Animals , Eating , Immunohistochemistry , Male , Mice , Oligopeptides , Paraventricular Hypothalamic Nucleus , Proto-Oncogene Proteins c-fos , Receptors, Ghrelin , Supraoptic Nucleus
9.
Neuropeptides ; 53: 45-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26299312

ABSTRACT

Nesfatin-1 is a novel 82-amino acid anorectic peptide. Previous studies of nesfatin-1 have focused on hypothalamic and brainstem circuits implicated in feeding regulation. Recently, nesfatin-1 expression was also reported in the nucleus accumbens (NAc), amygdaloid nucleus and insular cortex of mice, areas that are related to the control of reward behavior. Therefore, it is possible that nesfatin-1 might also inhibit food intake via central reward circuits. Using electrophysiology and electrochemical and behavioral tests, we investigated the effect of nesfatin-1 on the dopaminergic reward pathway between the ventral tegmental area (VTA) and the NAc. Our results showed that injection of nesfatin-1 into the VTA significantly inhibited dark-phase cumulative food intake in mice. The excitability of VTA dopaminergic neurons was inhibited by nesfatin-1. In addition, nesfatin-1 decreased dopamine release in the NAc. Therefore, we concluded that nesfatin-1 acts on dopaminergic neurons, and these effects might contribute to the decrease of food intake that results from the injection of nesfatin-1 into the VTA.


Subject(s)
Appetite Depressants/pharmacology , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Dopaminergic Neurons/drug effects , Eating/drug effects , Nerve Tissue Proteins/pharmacology , Neural Pathways/drug effects , Reward , Animals , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Nucleobindins , Nucleus Accumbens/drug effects , Patch-Clamp Techniques , Ventral Tegmental Area/drug effects
10.
Exp Cell Res ; 333(1): 155-63, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25659925

ABSTRACT

Kidney transplantation is the major therapeutic option for end-stage kidney diseases. However, acute rejection could cause allograft loss in some of these patients. Emerging evidence supports that microRNA (miRNA) dysregulation is implicated in acute allograft rejection. In this study, we used next-generation sequencing to profile miRNA expression in normal and acutely rejected kidney allografts. Among 75 identified dysregulated miRNAs, miR-10b was the most significantly downregulated miRNAs in rejected allografts. Transfecting miR-10b inhibitor into human renal glomerular endothelial cells recapitulated key features of acute allograft rejection, including endothelial cell apoptosis, release of pro-inflammatory cytokines (interleukin-6, tumor necrosis factor α, interferon-γ, and chemokine (C-C motif) ligand 2) and chemotaxis of macrophages whereas transfection of miR-10b mimics had opposite effects. Downregulation of miR-10b directly derepressed the expression of BCL2L11 (an apoptosis inducer) as revealed by luciferase reporter assay. Taken together, miR-10b downregulation mediates many aspects of disease pathogenicity of acute kidney allograft rejection. Restoring miR-10b expression in glomerular endothelial cells could be a novel therapeutic approach to reduce acute renal allograft loss.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Base Sequence , Graft Rejection/metabolism , Membrane Proteins/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Allografts , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Binding Sites , Case-Control Studies , Cells, Cultured , Chemokines/metabolism , Chemotaxis , Down-Regulation , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Kidney Transplantation , Macrophages/physiology , Membrane Proteins/metabolism , MicroRNAs/metabolism , Proto-Oncogene Proteins/metabolism , RNA Interference
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 34(12): 1763-7, 2014 Dec.
Article in Chinese | MEDLINE | ID: mdl-25537898

ABSTRACT

OBJECTIVE: To explore the impact of killer cell immunoglobulin-like receptor (KIR) gene mismatch on the outcomes of renal transplantation. METHODS: We collected the data from 111 donor-recipient pairs of kidney transplant and analyzed the status of KIR gene matching, acute rejection (AR), and 1-year and 3-year survival of the recipients who were followed continuously for over 37 months. RESULTS: Seventeen KIR genes were expressed in both recipient and donor groups, and the frequency of KIR3DS1 was significantly higher in the recipients than in the donors (38.75% vs 24.66%, OR=2.17, χ² = 3.94, P<0.05). The average rate of donor-recipient KIR matching was 82.53%. The donor-recipient KIR2DS1 matching rate was significantly higher in AR group than in no-AR group (85.00% vs 54.95%, χ² = 6.19, P<0.05). The rate of donor-recipient KIR AB-AB genotype was significantly higher in AR group than in no-AR group (33.33% vs 8.00%, P<0.05). The 1- and 3-year survival rates was 94.59% and 82.88% in these recipients, respectively. The frequency of donor KIR-AB genotpye was significantly higher in recipients with poor outcomes (57.89% vs 29.63%, χ² = 8.19, P<0.05); the frequency of both donor and recipient KIR-AB genotype was also significantly higher in recipients with poor prognoses (36.84% vs 9.78%, χ² = 14.87, P<0.05). CONCLUSIONS: KIR3DS1 may be the susceptible gene associated with uremia. A KIR-AB genotype of either the donor or the recipient can increase the risk of AR and reduce the 1- and 3-year survival rate. This finding can be of ethically importance in choosing a living related donor.


Subject(s)
Kidney Transplantation , Receptors, KIR/genetics , Genotype , Humans , Survival Rate , Tissue Donors , Treatment Outcome
12.
PLoS One ; 9(6): e98967, 2014.
Article in English | MEDLINE | ID: mdl-24906120

ABSTRACT

Nesfatin-1 is a recently discovered metabolic peptide hormone that decreases food intake after lateral, third, or fourth brain ventricle; cisterna magna; or paraventricular nucleus (PVN) injection in ad libitum fed rats. Additional micro-injection studies will improve the understanding of how nesfatin-1 acts on the brain and define specific nuclei responsive to nesfatin-1, which will provide insight on its effects on food intake. We evaluated how nesfatin-1 injection into the dorsal vagal complex (DVC) modulates food intake response in rats during the dark phase. Consistent with previous observations, nesfatin-1-injected rats significantly reduced cumulative food intake over a 5-h period in rats. Chronic administration of nesfatin-1 into the DVC reduced body weight gain over a 10-day period. Because glucosensing neurons in the DVC are involved in glucoprivic feeding and homeostatic control of blood glucose, we examined the effect of nesfatin-1 on the excitability of DVC glucosensing neurons. Nesfatin-1 inhibited most of the glucose-inhibitory (GI) neurons and excited most of the glucose-excitatory (GE) neurons in the DVC. Current-clamp electrophysiology recordings from DVC glucosensing neurons in slice preparation showed that bath applied nesfatin-1(10 nM) increased the firing frequency of GE neurons and inhibited the firing rate of GI-neurons. Nesfatin-1 inhibited 88.9% (16/18) of gastric distension inhibitory (GD-INH) neurons and excited 76.2% (32/42) of gastric distension excitatory (GD-EXC) neurons. Thus, nesfatin-1 may control food intake by modulating the excitability of glucosensing neurons in the DVC.


Subject(s)
Brain/cytology , Eating/drug effects , Glucose/metabolism , Nerve Tissue Proteins/pharmacology , Neurons/cytology , Neurons/drug effects , Animals , Brain/physiology , Eating/physiology , Injections , Male , Nerve Tissue Proteins/administration & dosage , Rats , Rats, Wistar , Stomach/physiology
13.
PLoS One ; 8(12): e83397, 2013.
Article in English | MEDLINE | ID: mdl-24391760

ABSTRACT

Nesfatin-1 is an anorexigenic peptide involved in energy homeostasis. Recently, nesfatin-1 was reported to decrease blood glucose level and improve insulin sensitivity in high-fat diet-fed rats. However, little information is known about the influence of nesfatin-1 on lipid metabolism either in physiological or diabetic condition. This study undertook whether nesfatin-1 was involved in the pathophysiology in Streptozotocin-induced type 2 diabetic mice (T2DM), which was induced by a combination of high-calorie diet and two low-doses Streptozotocin. We observed that plasma nesfatin-1 was significantly increased while expression of nesfatin-1 neurons were decreased in hypothalamus in diabetes group compared to only high-calorie diet control group; intravenous injection of nesfatin-1 decreased 0-1h, 0-2h, 0-3h cumulative food intake in T2DM, but 0-24h total food intake had no difference between groups. Body weight and plasma FFA were normalized after nesfatin-1(10 µg/Kg) administration for 6 days. These results suggested that nesfatin-1 improved lipid disorder in T2DM. It was found that blood glucose and insulin resistance coefficient decreased with treatment of nesfatin-1 (both in 1 µg/Kg and 10 µg/Kg doses) in diabetes mice. For further understanding the role of nesfatin-1 on lipid metabolism, we detected p-AMPK and p-ACC of skeletal muscle in T2DM using western blotting. The expression of p-AMPK and p-ACC increased when nesfatin-1 was given with doses 1 µg/Kg but not in doses 10 µg/Kg. Taken together, nesfatin-1 participated in the development of T2DM and stimulated free fatty acid utilization via AMPK-ACC pathway in skeletal muscle in T2DM.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Fatty Acids, Nonesterified/metabolism , Nerve Tissue Proteins/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Blood Glucose/metabolism , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/blood , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Diet, High-Fat/adverse effects , Enzyme Activation , Fatty Acids, Nonesterified/blood , Gastric Mucosa/metabolism , Hypothalamus/metabolism , Insulin/blood , Insulin Resistance , Male , Mice , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/blood , Nucleobindins , Oxidation-Reduction , Phosphorylation , Streptozocin/toxicity
14.
Regul Pept ; 177(1-3): 21-6, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22561448

ABSTRACT

Nesfatin-1 is a recently discovered neuropeptide that has been shown to decrease food intake after lateral, third, or fourth brain ventricle, cisterna magna administration, or PVN injection in ad libitum fed rats. With regards to the understanding of nesfatin-1 brain sites of action, additional microinjection studies will be necessary to define specific nuclei, in addition to the PVN, responsive to nesfatin-1 to get insight into the differential effects on food intake. In the present study, we evaluated nesfatin-1 action to modulate food intake response upon injection into the specific hypothalamic nuclei (PVN, LHA and VMN) in freely fed rats during the dark phase. We extend previous observations by showing that the nesfatin-1 (50 pmol) injected before the onset of the dark period significantly reduced the 1 to 5 h cumulative food intake in rats cannulated into the PVN, LHA, but not in rats cannulated into the VMN. Glucosensing neurons located in the hypothalamus are involved in glucoprivic feeding and homeostatic control of blood glucose. In order to shed light on the mechanisms by which nesfatin-1 exerts its satiety-promoting actions, we examined the effect of nesfatin-1 on the excitability of hypothalamic glucosensing neurons. Nesfatin-1 excited most of the glucose-inhibited (GI) neurons and inhibited most of the glucose-excited (GE) neurons in the PVN. Of 34 GI neurons in the LHA tested, inhibitory effects were seen in 70.6% (24/34) of GI neurons. The main effects were excitatory after intra-VMN administration of nesfatin-1 in GE neurons (27/35, 77.1%). Thus, our data clearly demonstrate that nesfatin-1 may exert at least a part of its physiological actions on the control of food intake as a direct result of its role in modulating the excitability of glucosensing neurons in the PVN, LHA and VMN.


Subject(s)
Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Eating/drug effects , Nerve Tissue Proteins/pharmacology , Neural Pathways/drug effects , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacology , Appetite Regulation/drug effects , Calcium-Binding Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Glucose/pharmacology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Male , Membrane Potentials , Microinjections/methods , Nerve Tissue Proteins/administration & dosage , Neurons/metabolism , Nucleobindins , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Wistar , Time Factors , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/metabolism
15.
Sheng Li Xue Bao ; 62(4): 357-64, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20717637

ABSTRACT

Ghrelin, an endogenous ligand for the growth hormone secretagogue (GHS) receptor, stimulates feeding and increases body weight. The primary action site of ghrelin has been reported to be the neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons in the hypothalamic arcuate nucleus (ARC). In addition to the hypothalamus, the caudal brainstem also appears to be an important mediator for the orexigenic activity of ghrelin. However, it is not clear whether ghrelin applied directly to the caudal brainstem activates forebrain structures. The aim of this study was to determine whether recruitment of forebrain structures was required for hyperphagic responses stimulated by ghrelin delivery within the caudal brainstem. In our experiment, all rats were surgically implanted with indwelling cannulas in the dorsal vagal complex (DVC), and ghrelin (20 pmol in 0.5 µL) was delivered to the DVC. After the injection, the orexigenic response to ghrelin was recorded by Feeding and Activity Analyser, and NPY/AgRP mRNA expressions in rat hypothalamus were detected by real-time PCR. In addition, the NPY immunoreactive neurons in the ARC were assayed by immunohistochemistry. The results showed that ghrelin significantly increased cumulative food intake at 1, 2 and 3 h after ghrelin injection, maximal response occurring at 2 h after injection. NPY/AgRP mRNA levels in ARC treated with ghrelin increased significantly compared with those in control group (injected with saline). The highest levels of NPY and AgRP mRNA were detected at 2 h after injection. The total number and mean optical density of NPY-positive neurons increased in ghrelin treated rats compared with those in control group. Consistently, ghrelin's effect was most pronounced at 2 h after injection. Taken together, we conclude that the activation of NPY/AgRP neurons in the ARC is involved in the mediation of the hyperphagic response to brainstem ghrelin administration in neurologically intact rats.


Subject(s)
Agouti-Related Protein/metabolism , Ghrelin/pharmacology , Hyperphagia/physiopathology , Hypothalamus/physiology , Neuropeptide Y/metabolism , Peptide Fragments/metabolism , Agouti-Related Protein/genetics , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Brain Stem/metabolism , Brain Stem/physiology , Feeding Behavior/drug effects , Hypothalamus/metabolism , Male , Neurons/metabolism , Neurons/physiology , Neuropeptide Y/genetics , Peptide Fragments/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
Regul Pept ; 159(1-3): 72-7, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19896982

ABSTRACT

The novel satiety factor nesfatin-1 has been shown to decrease food intake and body weight in rodents after i.c.v. injection. However, no further developments regarding the true patho-physiological relevance of nesfatin-1 in obesity and type 1 diabetes mellitus (T1 DM) and type 2 diabetes mellitus (T2 DM) have been reported. A recent study by Stengel et al. demonstrated that a down-regulation of NUCB2 mRNA in gastric endocrine cells was observed after 24-h fasting. They raised the possibility that nesfatin/NUCB2 gene expression may be regulated by nutritional status, suggesting that nesfatin-1 in the stomach might play a role in satiety. In the present study, fasting levels in plasma nesfatin-1, insulin and glucose were measured and analyzed in healthy subjects and in patients with T1 DM and T2 DM. Plasma nesfatin-1 levels were measured 6 times before and after oral glucose ingestion in healthy subjects. No sex differences in plasma nesfatin-1 were found. The mean fasting plasma nesfatin-1 levels were slightly but not significantly higher in T1 DM patients compared to healthy subjects. However, fasting plasma nesfatin-1 levels were significantly lower in T2 DM patients compared to healthy subjects and T1 DM patients. Plasma nesfatin-1 did not change acutely, although a small rise in circulating nesfatin-1 occurred within 30 min after the beginning of an oral glucose ingestion (from a mean basal value of 0.99+/-0.23 ng/ml to a maximum of 1.08+/-0.24 ng/ml). No significant difference in plasma nesfatin-1 before and after an oral glucose was observed. In conclusion, we showed that fasting nesfatin-1 was significantly lower in T2 DM patients compared to healthy subjects and T1 DM patients. The significance of this result is unclear but the reduction in fasting nesfatin-1 may be one of the appetite-related hormones involved in diabetic hyperphagia. In addition, neither glucose nor saline ingestions affected plasma nesfatin-1, suggesting that gastric chemosensation is not sufficient for the nesfatin-1 response under the present conditions.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Fasting/blood , Glucose/administration & dosage , Peptide Hormones/blood , Sweetening Agents/administration & dosage , Administration, Oral , Adult , Appetite/drug effects , Calcium-Binding Proteins , DNA-Binding Proteins , Female , Food , Humans , Hyperphagia/blood , Male , Middle Aged , Nerve Tissue Proteins , Nucleobindins
17.
Sheng Li Xue Bao ; 60(1): 149-55, 2008 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-18288371

ABSTRACT

In this study, the expressions of growth hormone secretagogue receptor type 1a (GHS-R1a) in the rat dorsal root ganglion (DRG) and nodose ganglion (NG) were investigated by using immunohistochemistry and in situ hybridization. The results clearly showed the presence of GHS-R1a mRNA and GHS-R1a-positive neurons in the rat DRG and NG. GHS-R1a was also co-localized with calcitonin gene-related peptide (CGRP) in some DRG and NG neurons, indicating the existence of subpopulations of the visceral afferents. The extrinsic primary afferent visceroceptive DRG and NG neurons from the stomach were identified by retrograde tracing fluorogold and stained for GHS-R1a and CGRP. Some neurons both positive for CGRP and GHS-Rla were labled by fluorogold. Our results not only demonstrate the expression of GHS-R1a in the vagal afferents but also provide the first and direct morphological evidence for its presence in the spinal visceral afferents, and gherin might have a modulatory role in the visceral afferent signaling.


Subject(s)
Afferent Pathways , Ganglia, Spinal/cytology , Nodose Ganglion/cytology , Receptors, Ghrelin/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Immunohistochemistry , Neurons, Afferent/cytology , Rats , Stomach/innervation
18.
Regul Pept ; 146(1-3): 169-75, 2008 Feb 07.
Article in English | MEDLINE | ID: mdl-17913259

ABSTRACT

Ghrelin has been identified as the endogenous ligand of the growth hormone secretagogue receptor (GHS-R). Recent studies have shown that site-specific injection of ghrelin directly into the dorsal vagal complex (DVC) of rats is equally as sensitive in its orexigenic response to ghrelin as the arcuate nucleus of the hypothalamus (ARC). It is as yet unclear how circulating ghrelin would gain access to and influence the activity of the neurons in the DVC in which GHS receptors are expressed. In the present study, neuronal activity was recorded extracellularly in the DVC of anesthetized rats in order to examine the effects of ghrelin on the glucosensing neurons and the gastric distension (GD) sensitive neurons. The 82 neurons were tested with glucose, of which 26 were depressed by glucose and identified as glucose-inhibited (glucose-INH) neurons; 11 were activated and identified as glucose-excited (glucose-EXC) neurons. Of 26 glucose-inhibited neurons examined for response to ghrelin, 23 were depressed, 1 was activated, and 2 failed to respond to ghrelin. Nine of 11 glucose-excited neurons were suppressed by ghrelin application, and the responses are abolished by the pretreatment with the GHS-R antagonist, [D-Lys-3]-GHRP-6. In addition, of 47 DVC neurons examined for responses to gastric distension (GD), 25 were excited (GD-EXC), 18 were inhibited (GD-INH). 18 out of the 25 GD-EXC neurons were excited, whereas 15 out of 18 GD-INH neurons were suppressed by ghrelin. In conclusion, the activity of the glucosensing neurons in the DVC can be modulated by ghrelin, the primary effect of ghrelin on the glucose-INH and glucose-EXC neurons was inhibitory. Two distinct population of GD-sensitive neurons exist in the rat DVC: GD-EXC neurons are activated by ghrelin; the GD-INH neurons are suppressed by ghrelin. There is a diversity of effects of ghrelin on neuronal activity within the DVC, it is as yet unclear how this diversity in ghrelin's effects on cellular excitability contributes to ghrelin biological actions to influence food intake and gastric motility.


Subject(s)
Ghrelin/pharmacology , Glucose , Neurons/drug effects , Stomach/drug effects , Vagus Nerve/drug effects , Animals , Electrophysiology , Glucose/physiology , Neurons/physiology , Rats , Rats, Wistar , Stomach/physiology
19.
Neurosci Bull ; 23(2): 75-82, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17592529

ABSTRACT

OBJECTIVE: To investigate the expression of motilin-immunoreactive neurons in the hypothalamus and the effect of central administration of erythromycin (EM) on the regulation of gastric motility in diabetic rats. METHODS: The motilin immunoreactive neurons in the hypothalamus and the hippocampus were detected by immunohistochemistry with rabbit anti-motilin polyclonal antibody. To measure the gastric motility, force transducers were surgically affixed to the gastric serosa. A microinjection syringe was connected via a plastic tube to an injection cannula, which was connected with a stainless steel guide cannula. The syringe was inserted into the right lateral cerebral ventricle for microinjecting the chemicals. RESULTS: Diabetic mellitus was successfully induced in cohorts of rats. Motilin-immunoreactive neurons significantly increased in the paraventricular (PVN) and supraoptic nuclei (SON) of the hypothalamus in the diabetic rats. Intracerebroventricular (i.c.v.) administration of EM, a motilin receptor agonist, stimulated the gastric motility of diabetic rats. EM (91.56 nmol, i.c.v.) dose-dependently increased the amplitude by (174.82 +/- 48.62)% (P<0.05), and increased the frequency by (70.43 +/- 27.11)% (P < 0.05) in 5 min. The stimulatory effect lasted more than 15 min to the end of the measurement, and can be blocked partially by the prior treatment of motilin receptor antagonist GM-109. CONCLUSION: Motilin-immunoreactive neurons are increased in the PVN and SON of the hypothalamus in diabetic rats. Centrally administered EM may regulate gastric motility by binding to the central motilin receptors, and central motilin might be involved in regulation of gastric motility in diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Erythromycin/pharmacology , Gastrointestinal Agents/pharmacology , Gastrointestinal Motility/physiology , Motilin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Supraoptic Nucleus/metabolism , Animals , Dose-Response Relationship, Drug , Erythromycin/administration & dosage , Gastrointestinal Agents/administration & dosage , Gastrointestinal Motility/drug effects , Hippocampus/cytology , Hippocampus/metabolism , Injections, Intraventricular , Male , Microinjections , Motilin/agonists , Neurons/cytology , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Rats , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/agonists , Receptors, Neuropeptide/agonists , Statistics, Nonparametric , Supraoptic Nucleus/cytology
20.
Brain Res ; 1055(1-2): 131-6, 2005 Sep 07.
Article in English | MEDLINE | ID: mdl-16084498

ABSTRACT

Ghrelin is an endogenous ligand of the growth hormone secretagogue receptor (GHS-R) with potent stimulatory effects on food intake. The aim of the present study was to investigate the effects of ghrelin on neuronal activity of hypothalamic glucose responding neurons. Single unit discharges in the lateral hypothalamic area (LHA), the ventromedial hypothalamic nucleus (VMH), and the parvocellular part of the paraventricular nucleus(pPVN) were recorded extracellularly by means of four-barrel glass micropipettes in anesthetized rats. The activity of glucose-sensitive neurons (GSNs) in the LHA, pPVN, and of glucoreceptor neurons (GRNs) in the VMH modulated by administration of ghrelin was analyzed. In the LHA, the majority of GSNs (17/25) increased in frequency due to ghrelin. Whereas the majority of VMH-GRNs (27/33) and pPVN-GSNs (9/13) was inhibited. The responses to ghrelin were abolished by pretreatment of [D-Lys-3]-GHRP-6, ghrelin receptor antagonist. These data indicate that the glucose responding neurons in the LHA, VMH, and pPVN are also involved in the orexigenic actions of ghrelin in the hypothalamic circuits, although AgRP/NPY neurons in the arcuate nucleus (ARC) are the primary targets of ghrelin.


Subject(s)
Glucose/pharmacology , Hypothalamus/cytology , Neurons/drug effects , Peptide Hormones/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cell Count/methods , Chi-Square Distribution , Drug Interactions , Female , Ghrelin , Male , Neurons/classification , Neurons/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...