Ghrelin Suppresses Glucose-stimulated Insulin Secretion and Deteriorates

Ghrelin Suppresses Glucose-stimulated Insulin Secretion and Deteriorates
Ghrelin Suppresses Glucose-stimulated Insulin Secretion and Deteriorates

Diabetes Publish Ahead of Print, published online June 28, 2010

Ghrelin Suppresses Glucose-stimulated Insulin Secretion and Deteriorates

Glucose Tolerance in Healthy Humans

1Jenny Tong, 2Ronald L Prigeon, 1Harold W Davis, 3Martin Bidlingmaier, 4Steven E Kahn, 4David E Cummings, 1Matthias H Tsch?p, 1,5David D’Alessio

1Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of

Cincinnati, Cincinnati, OH

2School of Medicine, University of Maryland, Veterans Affairs Medical Center, Baltimore, MA 3Medizinische Klinik–Innenstadt, Ludwig-Maximilians-Universit?t, Munich, Germany

4Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA

5Cincinnati VA Medical Center, OH

Running title: ghrelin and insulin secretion in healthy humans

Corresponding author:

Jenny Tong, MD, MPH

Email: jenny.tong@https://www.360docs.net/doc/6c2539524.html,

Additional information for this article can be found in an online appendix at

https://www.360docs.net/doc/6c2539524.html,

Submitted 15 April 2010 and accepted 15 June 2010.

This is an uncopyedited electronic version of an article accepted for publication in Diabetes. The American Diabetes Association, publisher of Diabetes, is not responsible for any errors or omissions in this version of the manuscript or any version derived from it by third parties. The definitive publisher-authenticated version will be available in a future issue of Diabetes in print and online at https://www.360docs.net/doc/6c2539524.html,.

Background: The orexigenic gut hormone ghrelin and its receptor are present in pancreatic islets. While ghrelin reduces insulin secretion in rodents, its effect on insulin secretion in humans has not been established.

Objective: To test the hypothesis that circulating ghrelin suppresses glucose-stimulated insulin secretion in healthy subjects.

Research Design and Methods:Ghrelin (0.3, 0.9 and 1.5 nmol/kg/hr) or saline was infused over 65 min in 12 healthy subjects (8M/4F) on 4 separate occasions in a counterbalanced fashion. An intravenous (IV) glucose tolerance test was performed during steady state plasma ghrelin levels. The acute insulin response to IV glucose (AIRg) was calculated from plasma insulin concentrations between 2 and 10 min after the glucose bolus. IV glucose tolerance was measured as the glucose disappearance constant (Kg) from 10 to 30 min.

Results:The three ghrelin infusions raised plasma total ghrelin concentrations to 4-, 11-, and 23-fold above the fasting level, respectively. Ghrelin infusion did not alter fasting plasma insulin or glucose, but compared to saline the 0.3, 0.9 and 1.5 nmol/kg/hr doses decreased AIRg (2152 ±448 vs. 1478 ± 2889, 1419 ± 275, and 1120 ± 174 pM) and Kg (0.3 and 1.5 nmol/kg/hr doses only) significantly (p < 0.05 for all). Ghrelin infusion raised plasma growth hormone and serum cortisol concentrations significantly (p < 0.001 for both) but had no effect on glucagon, epinephrine or norepinephrine levels (p = 0.44, 0.74 and 0.48, respectively).

Conclusions:This is a robust proof-of-concept study showing that exogenous ghrelin reduces glucose stimulated insulin secretion and glucose disappearance in healthy humans. Our findings raise the possibility that endogenous ghrelin has a role in physiologic insulin secretion, and that ghrelin antagonists could improve β-cell function.

hrelin has gained considerable

attention over the last decade for its

unique role in regulating mealtime hunger and lipid metabolism as well as short- and long-term energy homeostasis (1-3). It is the only known circulating factor that promotes food intake and increases fat mass. Ghrelin is secreted mainly from the stomach and proximal small bowel, and stimulates growth hormone (GH) secretion (4-6) in addition to its effect on energy balance. In healthy subjects, plasma ghrelin levels rise progressively before meals and fall to a nadir within one hour after eating, with changes in plasma levels during meals varying two- to three-fold (7-8). Under pathological conditions associated with severe malnutrition and weight loss, such as anorexia nervosa (9), cancer or cardiac cachexia (10-11), plasma total ghrelin levels are increased up to three-fold compared to healthy persons. Besides its well known effects on feeding behavior, fat mass, and GH secretion, ghrelin has more recently been implicated in the regulation of glucose homeostasis (12-13).

The GH secretagogue receptor (GHSR) 1a, also known as the ghrelin receptor, is widely distributed and has been localized to the hypothalamus, pituitary, liver, adipocyte and pancreas (14-15). Both ghrelin and GHSR are

G

2

3

expressed in human and rat pancreatic islets on both α- (16-17) and β-cells (18-19), and ghrelin is produced in a novel endocrine islet cell type that shares lineage with glucagon-

secreting cells (20-21). Pancreatic ghrelin cells exist as the predominant cell type in fetal

human islets and expression in the pancreas during development significantly precedes its occurrence in the stomach (20). In animal mutant models, an early block in the differentiation of insulin-producing β cells leads to an enormous increase in ghrelin-

producing ε cells, suggesting a developmental

link between ghrelin and insulin (22). In

vitro , ghrelin inhibits glucose-stimulated insulin secretion in a dose-dependent manner from cultured pancreata (23), isolated pancreatic islets (19; 24), and immortalized β-

cell lines (19; 21), suggesting that it acts directly on β cells to achieve this effect. In experimental animals, both ghrelin released from pancreatic islets and exogenous ghrelin inhibit glucose-stimulated insulin secretion (16; 24-26). Targeted gene deletion of ghrelin improves glucose tolerance and augments insulin secretion in ob/ob mice, suggesting a possible physiologic role which could be mediated by effects on islet function (27). Consistent with these findings, ghrelin gene deletion was shown to prevent glucose intolerance induced by high-fat diet, an environmentally-induced model of

hyperglycemia (26). Together, these findings indicate the potential of ghrelin blockade to prevent both genetically (ob gene)- and environmentally (high-fat diet) -induced glucose intolerance.

The effect of ghrelin on insulin secretion in humans is controversial. Intravenous (IV) injection of ghrelin decreases plasma insulin and increases blood glucose in some studies, suggesting inhibition of insulin secretion (12; 28). However, this finding has not been universally observed (29), and it is unclear whether such effects occur at physiologic or only pharmacologic doses of ghrelin. Prior studies performed in humans primarily assessed the impact of ghrelin on β-cell

function in the fasting state and there is little information on the effect of the peptide on

stimulated insulin release. Therefore, the role of ghrelin in the regulation of glucose homeostasis in humans remains poorly understood.

In this study, we determined the effect of ghrelin on glucose-stimulated insulin

secretion and glucose tolerance. We infused acyl-ghrelin, the bioactive endogenous ligand of the GHSR 1a, at variable doses with the aim of raising plasma total ghrelin level to physiologic (≤ 2 fold), supra-physiologic (2-3 fold) and pharmacologic (> 3 fold) levels. An

IV glucose tolerance test (IVGTT) was performed at steady state plasma ghrelin levels to determine the effect on glucose-stimulated insulin secretion and glucose tolerance in healthy, non-obese subjects.

PATIENTS AND METHODS

Subjects : Healthy volunteers between the ages of 18 and 55 years with a BMI between 18 and 29 kg/m 2 were recruited from the greater Cincinnati area. Subjects with a history or

clinical evidence of impaired fasting glucose or diabetes mellitus, recent myocardial infarction, congestive heart failure, active

liver or kidney disease, growth hormone deficiency or excess, neuroendocrine tumor, anemia or who were on medications known to alter insulin sensitivity were excluded. All study procedures were conducted at the

Cincinnati Veteran Affairs Medical Center General Clinical Research Center (CRC). All study participants gave informed consent for the study by signing a form approved by University of Cincinnati Institutional Review Board.

Experimental protocol : Subjects arrived at the CRC between 0700 and 0730 after a 10-12 hour fast for four separate experiments. IV

4

catheters were placed in veins of both forearms for blood sampling and infusion of test substances. The arm with the sampling catheter was heated to 55° to arterialize venous blood.

Synthetic human acylated ghrelin was obtained from Bachem AG (Rubendorf, Switzerland). The authenticity of the peptide was verified by mass spectrometry, the purity was > 95%, and reconstituted material was sterile and free of detectable pyrogens. On the morning of the four study days, either saline (as a control) or synthetic ghrelin dissolved in sterile saline solution was infused at doses of 0.3, 0.9, or 1.5 nmol/kg/hr (equivalent to 1, 3, or 5 μg/kg/hr) for a total of 65 minutes. The order of infusions was randomized, and study visits separated by at least five days. The use of synthetic human ghrelin was approved under the U.S. Food and Drug Administration Investigational New Drug 79,009. Following 55 minutes of ghrelin infusion, ~ 6 plasma half-lives of acyl-ghrelin (28), subjects received an IV bolus of glucose (11.4 g/m 2 body surface area) over 60 seconds as the initiation of an IVGTT (time 0). Blood samples were removed at 2, 3, 4, 5, 6, 8 and 10 minutes following IV glucose bolus for the estimation of acute insulin response to glucose (AIRg) and acute C-peptide response to glucose (ACRg). Another seven blood samples were taken at 12, 14, 16, 20, 22, 25 and 30 minutes for the calculation of glucose disappearance and ghrelin pharmacokinetics. Blood was placed on ice and plasma separated

by centrifugation within one hour, with the plasma being stored at -80° until used for assay. Blood pressure and heart rate were monitored every 15 minutes during the study procedure. A complete blood count, liver and kidney function tests, and an electrocardiogram were obtained as part of the safety monitoring of ghrelin use at the end of the last visit. Assays : Blood glucose concentrations were determined by the glucose oxidase method using a glucose analyzer (YSI 2300 STAT Plus; Yellow Springs Instruments, Yellow

Springs, OH). Plasma immunoreactive insulin levels were measured using a double-antibody radioimmunoassay (RIA) as described previously (30). C-peptide levels were measured using a commercial RIA kit (Millipore). Total immunoreactive ghrelin was measured by RIA (Millipore, Billarica, MA). The lower and upper limits of detection were 27 and 1765 pM (93 and 6000 pg/ml) respectively, and the intra-assay and inter-assay coefficients of variation (CV) were 6.4 and 16.3%, respectively. The ghrelin antibody used in the assay was directed toward the C-terminus of the molecule and binds both acyl- and desacyl-ghrelin, as well as truncated ghrelin species. Serum concentrations of human GH (hGH) were measured using the automated Immulite 2000 chemiluminescent assay system (Siemens, Bad Nauheim, Germany). This sandwich immunoassay utilizes a monoclonal mouse-anti-hGH capture- and a polyclonal rabbit-anti-hGH detection-antibody. The intra-assay CV was 3% and inter-assay variability ranged from 7%. Samples for glucagon were collected with benzamidine and heparin and were measured by RIA (Millipore, Billarica, MA). Cortisol levels were measured using the Corti-Cote RIA kit (MP Biomedicals, Orangeburg, NY). Plasma epinephrine and

norepinephrine were measured using the CatCombi ELISA kit (IBL International; Hamburg, Germany). All samples were run in duplicate, and all specimens from a given participant were run in the same assay. Calculations : AIRg and ACRg were calculated as the average plasma insulin and C-peptide increment above baseline from 2-10 minutes following IV glucose administration, respectively. The glucose

disappearance constant (31) was computed for

each IVGTT as the slope of the natural logarithm of glucose from 10 to 30 minutes. The rate of ghrelin disappearance was calculated as the slope of the natural logarithm of ghrelin after cessation of the ghrelin infusion at 65 minutes (10 minutes after the glucose bolus was given).

Statistical analysis: The data were analyzed using analysis of variance (ANOVA) with four treatment levels (control, and ghrelin infusion rates of 0.3, 0.9, and 1.5 nmol/kg/hr) and time of sampling being the repeated measure. Dependent variables included insulin, glucose, GH, cortisol, and glucagon concentrations. AIRg and ACRg for the four treatment levels were compared using a single-factor ANOVA. Posthoc analysis to compare control to each of the ghrelin infusion levels was performed using Dunnett's test. Data were analyzed using GraphPad Prism version 5.0 (GraphPad Software). All results are expressed as mean ± SEM unless otherwise noted.

RESULTS

Subject characteristics: Twelve healthy subjects (8 male and 4 females) aged 26.0 ±3.8 years with a BMI of 24.1 ± 1.4 kg/m2 were enrolled in the study. No subject had a fasting blood glucose of > 5.5 mM, mean fasting blood glucose for the group was 4.9 ±0.2 mM, and mean fasting plasma insulin was 37.8 ± 6.2 pM.

Ghrelin pharmacokinetics: Steady-state levels were reached after approximately 45 minutes for all three doses of acyl-ghrelin infusion. The average total ghrelin concentration during the time period between 45 and 54 minutes (10, 5 and 1 minute prior to IV glucose administration) for saline and the 3 acyl-ghrelin infusions were 304 ± 18, 1,429 ± 49, 4,629 ± 194, and 7,045 ± 295 pM. The 0.3, 0.9 and 1.5 nmol/kg/hr infusions raised the total ghrelin immunoreactivity 4.5 -, 15.4 -, and 22.6 - fold above an average basal level of 308 ± 30 pM for the three infusions (Figure

1; Supplemental Material Table 1 available in

the online appendix at https://www.360docs.net/doc/6c2539524.html,). The intra-subject CV% for the saline, 0.3, 0.9 and

1.5 nmol/kg/hr ghrelin infusions were 13.8,

7.7, 6.8, and 7.0%, respectively. The inter-subject CV% for the steady-state total ghrelin measurement with different ghrelin infusion

rates were 23.7, 20.1, 14.6, and 24.1%, respectively. After cessation of the ghrelin infusion at 65 min, total ghrelin levels declined following a first-order (exponential) decrease with an overall elimination rate constant (K el) of 0.023 min-1, corresponding

to an elimination half life of 30 minutes.

Effects exogenous ghrelin on plasma insulin

and glucose:The average fasting plasma glucose and insulin values at baseline and at

times when ghrelin concentration reached steady state (45 to 54 minutes) were shown in

Table 1. Infusion of exogenous ghrelin did

not alter fasting plasma concentrations of insulin and glucose from baseline (p > 0.05

for all comparisons).

Compared to saline, the doses of 0.3, 0.9, and

1.5 nmol/kg/hr ghrelin each resulted in a significant reduction of AIRg (2152 ± 448 to

1478 ± 288, 1419 ± 2751 and 1210 ± 188 pM,

p < 0.05, < 0.05, and < 0.01, respectively) during an IVGTT (Figures 2A and 2B). The magnitude of suppression in AIRg increased

with higher doses of ghrelin administration suggesting a dose-dependent relationship between circulating ghrelin concentration and insulin secretion. Similar to AIRg, a significant suppression of C-peptide release in response to IV glucose was also seen with all

three doses of ghrelin infusions (5.8 ± 0.9 to

4.1 ± 0.4, 4.2 ± 0.5, and 3.6 ± 0.6 nM, p <

0.05, < 0.05, and <0.01, respectively) (Figure

2C). In addition, ghrelin infusion at the 0.3

and 1.5 nmol/kg/hr doses also significantly decreased the rate of glucose disappearance (p

< 0.05 for both comparisons; Figure 3).

5

6

Effects of exogenous ghrelin on counterregulatory hormones : The three doses of ghrelin raised peak plasma GH levels by 12-, 114-, and 75-fold above baseline, respectively (Figure 4). The 0.9 and 1.5 nmol/kg/hr rates of ghrelin infusion also raised plasma cortisol levels significantly as compared to baseline at 30, 54, and 65 minutes (p < 0.01) (Figure 5). Ghrelin infusion, regardless of dose, had no effect on glucagon secretion (p = 0.44) (Supplemental Material Figure 1). Plasma epinephrine and norepinephrine levels did not differ between baseline and 54 minutes when ghrelin in the circulation reached a steady state regardless of the type of infusion the subjects received (Supplemental Material Figure 2).

Side effects: Ghrelin infusion was generally well tolerated. The most common complaints during infusion of ghrelin were hunger and “warm sensation”. These symptoms were transient and resolved spontaneous after cessation of the infusion. One subject while receiving the 1.5 nmol/kg/hr ghrelin infusion experienced a 23 mmHg decrease in mean arterial blood pressure without a significant change in heart rate. The subject was asymptomatic except for feeling “warm and hungry” during the event. The blood pressure returned to baseline within minutes after the ghrelin infusion was discontinued prematurely. This blood pressure change was

not observed in any other subject or with any other dose.

DISCUSSION

Preclinical studies support a role for ghrelin to

regulate glucose metabolism as well as energy

balance and GH secretion. However, the effect of ghrelin on insulin secretion and glucose tolerance in humans has not been clearly established in the limited number of studies reported previously. In the present study, we examined the effect of a range of ghrelin doses on dynamic insulin secretion and glucose metabolism and demonstrated that acyl-ghrelin suppresses glucose-stimulated insulin secretion and worsens IV glucose tolerance in healthy humans. These effects appear to be present at concentrations of ghrelin above the usual physiologic range, in a pattern consistent with dose-dependence. Our findings extend to humans the effects previously best demonstrated in mice, and suggest that ghrelin has a role in systemic glucose homeostasis. Moreover our results raise possibilities for targeting the human ghrelin system as a means to improve disorders of glucose metabolism.

Several studies have examined the effect of ghrelin on insulin secretion in humans. In a

study of healthy young males by Broglio et al (12), an IV bolus injection of ghrelin (0.3 nmol/kg or 1.0 μg/kg) significantly increased fasting plasma glucose levels followed by a reduction in serum insulin levels beginning at 15 and 30 minutes after ghrelin administration, respectively, suggesting inhibition of insulin secretion. When the same dose of ghrelin was given as a continuous IV infusion to subjects who had undergone total gastrectomy, by necessity reducing the production of most endogenous ghrelin, C-peptide levels were suppressed when compared to saline infusion (32). In contrast, Lucidi et al infused acyl ghrelin at a

rate of 7.5 or 15 pmol/kg/min for two hours in 8 healthy subjects and failed to observe a

significant change in fasting plasma glucose and insulin levels (29). However, all previous studies in humans used fasting insulin as the marker of ghrelin effects on the β-cell, with no examination of stimulated insulin secretion. In the present study, we examined the effect of continuous infusions of low-, medium- and high-doses of acyl ghrelin on AIRg, a well-established measure of insulin secretion that we think provides a more sensitive measure of β-cell function. The measure of C-peptide levels during the

IVGTT confirms the changes in AIRg, and support an effect of ghrelin on insulin secretion rather than insulin clearance. Moreover, the continuous infusion of ghrelin during the IVGTT also eliminated any potential bias in the β-cell response introduced by rapid changes in plasma ghrelin levels as occur with a bolus injection of the peptide. Based on these design advantages we believe that ours is the most robust proof-

of-concept study yet of the effect of ghrelin on insulin secretion in humans.

Similar to Lucidi et al (29), we did not observe a significant change in fasting insulin or glucose levels with any of the three doses of ghrelin. On the other hand, we did find a clear suppressive effect of ghrelin on the first-phase insulin response in an apparent dose-dependent fashion, with the greatest effect seen with the highest dose ghrelin (Figure 2). In addition, the decrease in IV glucose tolerance is consistent with a reduction of insulin secretion. Our observations are in keeping with several in vitro studies that have provided evidence that ghrelin has an inhibitory effect on stimulated insulin secretion from pancreatic β-cells (16; 19; 21; 24-26) and in vivo studies that have shown a deteriorating effect on glucose tolerance (25;

27). The mechanism(s) by which ghrelin could inhibit insulin secretion is unknown. Ghrelin may exert a direct effect on the β-cell or act indirectly by stimulating the secretion of counter-regulatory hormones that affect insulin secretion, or activating neural pathways that regulate islet function (33-38). The signaling mechanisms for insulinostatic ghrelin action in islet β-cells have been explored. Both endogenous- and exogenous-ghrelin has been shown to attenuate glucose-induced insulin release via Gαi2-mediated activation of Kv channels and suppression of action potential firing and [Ca2+]i increases in β-cells (39). Furthermore, both ghrelin and its receptor are expressed in human and rat pancreatic islets (on α-, β-, and ε-cells) (16; 18; 20; 40) and normal mouse pancreas contains a small population of ghrelin producing ε-cells which appear to be distinct from α- and β-cells (22; 41). Ghrelin-immunoreactive cells are abundant in human islets during development, outnumbering those in the stomach, but few are present in adults (20). It is interesting to note that mice lacking the homeodomain protein Nkx2.2, which is essential for the differentiation of insulin producing β-cells, have islets in which

the β-cells are almost completely replaced by

ε-cells (22). These findings raise the possibility of a shared common progenitor for both β- and ε-cells and suggest a role of ghrelin in the pancreatic islet, perhaps as a regulator of glucose homeostasis. Lastly, gut-brain crosstalk has been well described and it

is possible that ghrelin achieves its metabolic actions in the pancreas, muscle, adipose tissue and liver via central ghrelin and insulin signaling (42-44).

As for possible indirect mechanisms of ghrelin action on the islet, previous studies in animals and humans have shown that both epinephrine and cortisol exhibit inhibitory effects on insulin secretion (33-36). We have shown here that cortisol levels were significantly elevated when higher doses of acyl ghrelin were given (0.9 and 1.5 nmol/kg/hr). However, since steroid hormone action is thought to be mediated primarily by changes in gene transcription (45), it seems unlikely that the acute effect of ghrelin on AIRg can be explained by glucocorticoid activity. In contrast to a previous observation (46), we did not observe an increase in epinephrine levels with ghrelin administration. This could be due to the difference in assay reproducibility (or perhaps sensitivity) or the method of ghrelin administration. As expected, GH levels were significantly elevated during ghrelin infusion (Figure 5). Acutely, infusion of GH to levels

7

within the physiological range (27 ± 2 ng/ml) decrease insulin-mediated glucose uptake in the periphery within 2 to 12 hours (37). In this study the plasma insulin response to hyperglycemia was not altered by GH, and other investigators have noted increased plasma insulin concentration after 12-hour infusion of GH to healthy volunteers (38). Therefore we do not think the effects of ghrelin to reduce AIRg can be explained by changes in plasma GH.

Theoretically, the decrease in insulin secretion with ghrelin administration could be an adaptation to an increase in peripheral insulin sensitivity. However, previous studies

in humans and animals seem to suggest that ghrelin consistently reduces, rather than improves, peripheral insulin sensitivity (12; 28; 32; 47). The length of the IVGTT was limited by the total dose of ghrelin we could administer to each individual based on FDA requirements. For this reason, we do not have insulin sensitivity measures from IVGTT in this study. Overall, our data do not support indirect actions of counter-regulatory hormones or systemic insulin sensitivity to mediate the effects of ghrelin on insulin secretion.

Although in this study the effects of ghrelin on β cell function occurred at supraphysiological concentrations, it is important to consider that since ghrelin is produced in the islet ε cells (20-21), intra-islet ghrelin concentrations may reach very high levels, raising the possibility that ghrelin could act locally on β cells via paracrine mechanisms (48-49) similarly to what has been demonstrated in adult rat islets (16). It is generally accepted that the level of hormone working in a paracrine/autocrine manner is higher than that working in an endocrine manner. Therefore, our observation of a suppressive effect of ghrelin on insulin secretion while the circulating level is in the supraphysiologic range does not exclude the possibility of a physiologic function of this hormone. Further studies will be necessary to delineate mechanisms by which endogenous ghrelin may affect islet function. Based on our results endocrine, paracrine and neural mechanisms are all plausible possibilities.

The effect of ghrelin on α-cell function in humans has not been previously studied. Glucagon secretion is enhanced by ghrelin in vitro (25), but the effect of ghrelin on its release is less impressive in vivo, with levels largely unchanged or mildly increased following ghrelin administration (23; 25; 29; 32). In our hands, no relevant change in glucagon level was seen with pharmacologic level ghrelin administration during fasting or IVGTT. Future studies that employ measurement of dynamic changes of glucagon level using more sensitivity methods should be done to confirm this finding. CONCLUSION

Our study demonstrates that exogenous ghrelin markedly reduces the first-phase insulin and C-peptide responses to IV glucose

in healthy humans. These findings raise the possibility that endogenous ghrelin has a role

in physiologic insulin secretion, and that ghrelin antagonists could improve β-cell function and serve as a novel drug target for the treatment of type 2 diabetes.

Author Contributions:J.T. researched data, wrote manuscript. R.L.P. researched data, contributed to discussion, reviewed/edited manuscript. H.W.D. researched data, contributed to discussion, reviewed/edited manuscript. M.B. researched data, reviewed/edited manuscript. S.E.K. contributed to discussion, reviewed/edited manuscript. D.E.C. reviewed/edited manuscript. M.H.T. contributed to discussion, reviewed/edited manuscript. D.D. contributed to discussion, reviewed/edited manuscript.

8

ACKNOWLEDGMENTS

Funding for this research is provided by NIH/NIDDK (5K23DK80081 to J.T. and R0157900 to D.D.) and the Department of Veterans Affairs. We want to thank the GCRC nursing staff, Kay Ellis, and Brianne Paxton for their excellent support for the study. M.H.T. is a scientific advisory board member and stockholder of Marcadia Biotech, Acylin Pharmaceutical, and Ambrx Inc.

9

Figure legends:

Figure 1: Plasma total ghrelin levels during continuous IV infusions (-15 to 65 minutes) of saline, 0.3, 0.9, or 1.5 nmol/kg/hr of acyl ghrelin in healthy men and women. A bolus IV dose of glucose (11.4 g/m2 body surface area) was infused over 1 minute after plasma ghrelin had reached a steady state (55 minutes). The acyl ghrelin infusions resulted in a dose-dependent increase in plasma ghrelin.

Figure 2A: Plasma insulin concentrations during an IVGTT following 55 minute infusions of acyl ghrelin at 0.3, 0.9, or 1.5 nmol/kg/hr, or saline.

Figure 2B: The acute insulin response to IV glucose (AIRg) determined during infusions of acyl ghrelin at 0.3, 0.9, or 1.5 nmol/kg/hr dose, or saline. *p < 0.05, **p < 0.01.

Figure 2C: The acute C-peptide response to IV glucose (ACRg) determined during infusions of acyl ghrelin at 0.3, 0.9, or 1.5 nmol/kg/hr dose, or saline. *p < 0.05, **p < 0.01.

Figure 3: Glucose disappearance constant (Kg) determined during infusions of acyl ghrelin at 0.3, 0.9, or 1.5 nmol/kg/hr, or saline. *p < 0.05.

Figure 4: Plasma growth hormone concentrations during a 65 minute infusion of acyl ghrelin at 0.3, 0.9, or 1.5 nmol/kg/hr, or saline. Glucose was administered as an IV bolus following 55 minutes of the infusion. a, b and c are saline vs. 0.3, 0.9, or 1.5 nmol/kg/hr of ghrelin, respectively; *p < 0.05, ***p < 0.001.

Figure 5: Plasma cortisol concentrations during a 65 minute infusion of acyl ghrelin at 0.3, 0.9, or 1.5 nmol/kg/hr, or saline. Glucose was administered as an IV bolus following 55 minutes of the infusion. b and c are saline vs. 0.9 and 1.5 nmol/kg/hr ghrelin, respectively; *p < 0.05, ***p < 0.001.

REFERENCES

1. Tschop M, Smiley DL, Heiman ML: Ghrelin induces adiposity in rodents. Nature 407:908-913, 2000

2. Cummings DE: Ghrelin and the short- and long-term regulation of appetite and body weight. Physiol Behav 89:71-84, 2006

3. Castaneda TR, Tong J, Datta R, Culler M, Tschop MH: Ghrelin in the regulation of body weight and metabolism. Front Neuroendocrinol 31:44-60, 2010

4. Bowers CY, Momany FA, Reynolds GA, Hong A: On the in vitro and in vivo activity of a new synthetic hexapeptide that acts on the pituitary to specifically release growth hormone. Endocrinology 114:1537-1545, 1984

5. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K: Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402:656-660, 1999

6. Arvat E, Di Vito L, Broglio F, Papotti M, Muccioli G, Dieguez C, Casanueva FF, Deghenghi R, Camanni F, Ghigo E: Preliminary evidence that Ghrelin, the natural GH secretagogue (GHS)-receptor ligand, strongly stimulates GH secretion in humans. J Endocrinol Invest 23:493-495, 2000

7. Tschop M, Wawarta R, Riepl RL, Friedrich S, Bidlingmaier M, Landgraf R, Folwaczny C: Post-prandial decrease of circulating human ghrelin levels. J Endocrinol Invest 24:RC19-21, 2001

10

8. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS: A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50:1714-1719, 2001

9. Otto B, Cuntz U, Fruehauf E, Wawarta R, Folwaczny C, Riepl RL, Heiman ML, Lehnert P, Fichter M, Tschop M: Weight gain decreases elevated plasma ghrelin concentrations of patients with anorexia nervosa. Eur J Endocrinol 145:669-673, 2001

10. Shimizu Y, Nagaya N, Isobe T, Imazu M, Okumura H, Hosoda H, Kojima M, Kangawa K, Kohno N: Increased plasma ghrelin level in lung cancer cachexia. Clin Cancer Res 9:774-778, 2003

11. Nagaya N, Uematsu M, Kojima M, Date Y, Nakazato M, Okumura H, Hosoda H, Shimizu W, Yamagishi M, Oya H, Koh H, Yutani C, Kangawa K: Elevated circulating level of ghrelin in cachexia associated with chronic heart failure: relationships between ghrelin and anabolic/catabolic factors. Circulation 104:2034-2038, 2001

12. Broglio F, Arvat E, Benso A, Gottero C, Muccioli G, Papotti M, van der Lely AJ, Deghenghi R, Ghigo E: Ghrelin, a natural GH secretagogue produced by the stomach, induces hyperglycemia and reduces insulin secretion in humans. J Clin Endocrinol Metab 86:5083-5086, 2001

13. Sun Y, Asnicar M, Smith RG: Central and peripheral roles of ghrelin on glucose homeostasis. Neuroendocrinology 86:215-228, 2007

14. Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, Rosenblum CI, Hamelin M, Hreniuk DL, Palyha OC, Anderson J, Paress PS, Diaz C, Chou M, Liu KK, McKee KK, Pong SS, Chaung LY, Elbrecht A, Dashkevicz M, Heavens R, Rigby M, Sirinathsinghji DJ, Dean DC, Melillo DG, Patchett AA, Nargund R, Griffin PR, DeMartino JA, Gupta SK, Schaeffer JM, Smith RG, Van der Ploeg LH: A receptor in pituitary and hypothalamus that functions in growth hormone release. Science 273:974-977, 1996

15. Guan XM, Yu H, Palyha OC, McKee KK, Feighner SD, Sirinathsinghji DJ, Smith RG, Van der Ploeg LH, Howard AD: Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res Mol Brain Res 48:23-29, 1997

16. Dezaki K, Hosoda H, Kakei M, Hashiguchi S, Watanabe M, Kangawa K, Yada T: Endogenous ghrelin in pancreatic islets restricts insulin release by attenuating Ca2+ signaling in beta-cells: implication in the glycemic control in rodents. Diabetes 53:3142-3151, 2004

17. Date Y, Nakazato M, Hashiguchi S, Dezaki K, Mondal MS, Hosoda H, Kojima M, Kangawa K, Arima T, Matsuo H, Yada T, Matsukura S: Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes 51:124-129, 2002

18. Volante M, Allia E, Gugliotta P, Funaro A, Broglio F, Deghenghi R, Muccioli G, Ghigo E, Papotti M: Expression of ghrelin and of the GH secretagogue receptor by pancreatic islet cells and related endocrine tumors. J Clin Endocrinol Metab 87:1300-1308, 2002

19. Colombo M, Gregersen S, Xiao J, Hermansen K: Effects of ghrelin and other neuropeptides (CART, MCH, orexin A and B, and GLP-1) on the release of insulin from isolated rat islets. Pancreas 27:161-166, 2003

20. Wierup N, Svensson H, Mulder H, Sundler F: The ghrelin cell: a novel developmentally regulated islet cell in the human pancreas. Regul Pept 107:63-69, 2002

21. Wierup N, Yang S, McEvilly RJ, Mulder H, Sundler F: Ghrelin is expressed in a novel endocrine cell type in developing rat islets and inhibits insulin secretion from INS-1 (832/13) cells. J Histochem Cytochem 52:301-310, 2004

11

22. Prado CL, Pugh-Bernard AE, Elghazi L, Sosa-Pineda B, Sussel L: Ghrelin cells replace insulin-producing beta cells in two mouse models of pancreas development. Proc Natl Acad Sci U S A 101:2924-2929, 2004

23. Egido EM, Rodriguez-Gallardo J, Silvestre RA, Marco J: Inhibitory effect of ghrelin on insulin and pancreatic somatostatin secretion. Eur J Endocrinol 146:241-244, 2002

24. Reimer MK, Pacini G, Ahren B: Dose-dependent inhibition by ghrelin of insulin secretion in the mouse. Endocrinology 144:916-921, 2003

25. Salehi A, Dornonville de la Cour C, Hakanson R, Lundquist I: Effects of ghrelin on insulin and glucagon secretion: a study of isolated pancreatic islets and intact mice. Regul Pept 118:143-150, 2004

26. Dezaki K, Sone H, Koizumi M, Nakata M, Kakei M, Nagai H, Hosoda H, Kangawa K, Yada T: Blockade of pancreatic islet-derived ghrelin enhances insulin secretion to prevent high-fat diet-induced glucose intolerance. Diabetes 55:3486-3493, 2006

27. Sun Y, Asnicar M, Saha PK, Chan L, Smith RG: Ablation of ghrelin improves the diabetic but not obese phenotype of ob/ob mice. Cell Metab 3:379-386, 2006

28. Akamizu T, Takaya K, Irako T, Hosoda H, Teramukai S, Matsuyama A, Tada H, Miura K, Shimizu A, Fukushima M, Yokode M, Tanaka K, Kangawa K: Pharmacokinetics, safety, and endocrine and appetite effects of ghrelin administration in young healthy subjects. Eur J Endocrinol 150:447-455, 2004

29. Lucidi P, Murdolo G, Di Loreto C, Parlanti N, De Cicco A, Fatone C, Taglioni C, Fanelli C, Broglio F, Ghigo E, Bolli GB, Santeusanio F, De Feo P: Metabolic and endocrine effects of physiological increments in plasma ghrelin concentrations. Nutr Metab Cardiovasc Dis 15:410-417, 2005

30. Elder DA, Prigeon RL, Wadwa RP, Dolan LM, D'Alessio DA: Beta-cell function, insulin sensitivity, and glucose tolerance in obese diabetic and nondiabetic adolescents and young adults. J Clin Endocrinol Metab 91:185-191, 2006

31. Kahn SE, Montgomery B, Howell W, Ligueros-Saylan M, Hsu CH, Devineni D, McLeod JF, Horowitz A, Foley JE: Importance of early phase insulin secretion to intravenous glucose tolerance in subjects with type 2 diabetes mellitus. J Clin Endocrinol Metab 86:5824-5829, 2001 32. Damjanovic SS, Lalic NM, Pesko PM, Petakov MS, Jotic A, Miljic D, Lalic KS, Lukic L, Djurovic M, Djukic VB: Acute effects of ghrelin on insulin secretion and glucose disposal rate in gastrectomized patients. J Clin Endocrinol Metab 91:2574-2581, 2006

33. Tamagawa T, Henquin JC: Epinephrine modifications of insulin release and of 86Rb+ or 45Ca2+ fluxes in rat islets. Am J Physiol 244:E245-252, 1983

34. Sherwin RS, Sacca L: Effect of epinephrine on glucose metabolism in humans: contribution of the liver. Am J Physiol 247:E157-165, 1984

35. Barseghian G, Levine R: Effect of corticosterone on insulin and glucagon secretion by the isolated perfused rat pancreas. Endocrinology 106:547-552, 1980

36. Kalhan SC, Adam PA: Inhibitory effect of prednisone on insulin secretion in man: model for duplication of blood glucose concentration. J Clin Endocrinol Metab 41:600-610, 1975

37. Bratusch-Marrain PR, Smith D, DeFronzo RA: The effect of growth hormone on glucose metabolism and insulin secretion in man. J Clin Endocrinol Metab 55:973-982, 1982

38. Rizza RA, Mandarino LJ, Gerich JE: Effects of growth hormone on insulin action in man. Mechanisms of insulin resistance, impaired suppression of glucose production, and impaired stimulation of glucose utilization. Diabetes 31:663-669, 1982

12

39. Dezaki K, Kakei M, Yada T: Ghrelin uses Galphai2 and activates voltage-dependent K+ channels to attenuate glucose-induced Ca2+ signaling and insulin release in islet beta-cells: novel signal transduction of ghrelin. Diabetes 56:2319-2327, 2007

40. Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P, Bhattacharya S, Carpenter R, Grossman AB, Korbonits M: The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J Clin Endocrinol Metab 87:2988, 2002

41. Andralojc KM, Mercalli A, Nowak KW, Albarello L, Calcagno R, Luzi L, Bonifacio E, Doglioni C, Piemonti L: Ghrelin-producing epsilon cells in the developing and adult human pancreas. Diabetologia 52:486-493, 2009

42. Obici S, Zhang BB, Karkanias G, Rossetti L: Hypothalamic insulin signaling is required for inhibition of glucose production. Nat Med 8:1376-1382, 2002

43. Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L: Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci 5:566-572, 2002

44. Pocai A, Morgan K, Buettner C, Gutierrez-Juarez R, Obici S, Rossetti L: Central leptin acutely reverses diet-induced hepatic insulin resistance. Diabetes 54:3182-3189, 2005

45. Gronemeyer H: Control of transcription activation by steroid hormone receptors. FASEB J 6:2524-2529, 1992

46. Nagaya N, Kojima M, Uematsu M, Yamagishi M, Hosoda H, Oya H, Hayashi Y, Kangawa K: Hemodynamic and hormonal effects of human ghrelin in healthy volunteers. Am J Physiol Regul Integr Comp Physiol 280:R1483-1487, 2001

47. Vestergaard ET, Gormsen LC, Jessen N, Lund S, Hansen TK, Moller N, Jorgensen JO: Ghrelin infusion in humans induces acute insulin resistance and lipolysis independent of growth hormone signaling. Diabetes 57:3205-3210, 2008

48. Ahima RS: Ghrelin--a new player in glucose homeostasis? Cell Metab 3:301-302, 2006

49. van der Lely AJ: Ghrelin and new metabolic frontiers. Horm Res 71 Suppl 1:129-133, 2009 Table 1: Basal plasma glucose and insulin levels during continuous IV infusions of saline, 0.3, 0.9, or 1.5 nmol/kg/hr acyl ghrelin (0-54 minutes) prior to an IVGTT. Baseline plasma glucose and insulin concentration was calculated as the average of the -15 and -1 min values. Plasma glucose and insulin at steady-state ghrelin concentration was calculated as the average of 45, 50, and 54 min values.

Infusion rate

Plasma glucose (mg/dl) Plasma insulin (pM) Baseline

Ghrelin

steady state

(t=45-54 min)

Baseline

Ghrelin

steady state

(t=45-54 min)

Saline 83.8 ± 4.6 86.9 ± 1.0 34.1 ± 4.8 36.5 ± 5.8 Ghrelin (0.3 nmol/kg/h) 86.5 ± 2.4 96.7 ± 6.7 36.3 ± 4.8 30.4 ± 5.0 Ghrelin (0..9 nmol/kg/h) 91.4 ±2.1 93.5 ± 2.7 42.0 ± 6.6 36.1 ± 6.9 Ghrelin (1.5 nmol/kg/h) 87.6 ± 1.6 91.5 ± 2.4 39.1 ± 5.8 25.6 ± 3.9

13

14

15

Ghrelin and insulin secretion in healthy humans

16

Figure 3

Figure 4

Figure 5

Ghrelin在摄食及脂质代谢中的调节作用

万方数据

万方数据

万方数据

Ghrelin在摄食及脂质代谢中的调节作用 作者:顾辨辨, 严光 作者单位:安徽医科大学附属省立医院,安徽省立医院老年医学科,合肥,230001 刊名: 中国临床保健杂志 英文刊名:CHINESE JOURNAL OF CLINICAL HEALTHCARE 年,卷(期):2011,14(2) 参考文献(27条) 1.Wren AM;Small CJ;Ward HL The novel hypothalamic peptide Ghrelin stimulates food intake and growth hormone secretion 2000(11) 2.Yamamoto K;Takiguchi S;Miyata H Randomized phase II study of clinical effects of Ghrelin after esophagectomy with gastric tube reconstruction 2010(01) 3.Wang L;Basa NR;Shaikh A LPS inhibits fasted plasma Ghrelin levels in rats:role of IL-1 and PGs and functional implications 2006(04) https://www.360docs.net/doc/6c2539524.html,go F;Gonzalez-Juanatey JR;Casanueva FF Ghrelin,the same peptide for different functions:player or bystander 2005(14) 5.Chen HY;Trumbauer ME;Chen AS Orexigenic action of peripheral Ghrelin is mediated by neuropeptide Y and agouti-related protein[外文期刊] 2004(06) 6.Sakata I;Yamazaki M;Inoue K Growth hormone secretagogue receptor expression in the cells of the stomachprojected afferent nerve in the rat nodose ganglion[外文期刊] 2003(03) 7.le Roux CW;Neary NM;Halsey TJ Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy 2005(08) 8.Arnold M;Mura A;Langhans W Gut vagal afferents are not necessary for the eating-stimulatory effect of intraperitoneally injected Ghrelin in the rat[外文期刊] 2006(43) 9.Zigman JM;Nakano Y;Coppari RA Mice lacking Ghrelin receptors resist the development of diet-induced obesity 2005(12) 10.Gardiner JV;Campbell D;Patterson M The hyperphagic effect of Ghrelin is inhibited in mice by a diethigh in fat[外文期刊] 2010(07) 11.Arosio M;Ronchi CL;Beck-Peccoz P Effect of modified sham feeding on Ghrelin levels in healthy human subjects[外文期刊] 2004(10) 12.Reinehr T;de Sousa G;Roth CL Obestatin and Ghrelin levels in obese children and adolescents before and after reduction of overweight 2008(02) 13.Cummings DE;Clement K;Purnell JQ Elevated plasma Ghrelin levels in Prader Willi syndrome[外文期刊] 2002(07) 14.Inhoff T;Wiedenmann B;Klapp BF Is desacyl Ghrelin a modulator of food intake[外文期刊] 2009(05) 15.Chen CY;Inui A;Asakawa A Des-acyl Ghrelin acts by CRF type 2 receptors to disrupt fasted stomach motility in conscious rats[外文期刊] 2005(01) 16.Inhoff T;Noetzel S;Stengel A Desacyl Ghrelin inhibits the orexigenic effect of peripherally injected Ghrelin in rats[外文期刊] 2008(12) 17.Zhang IV;Ren PG;Avsian-Kretchmer 0Obestatin,a peptide encoded by the Ghrelin gene,opposes

ghrelin在海马调控下丘脑室旁核胃牵张敏感神经元活动中作用

第48卷 第5期2012年10月 青岛大学医学院学报 ACTA ACADEMIAE MEDICINAE  QINGDAO UNIVERSITATISVol.48,No.5October 2 012·论著· [收稿日期]2012-05-07; [修订日期]2012-08- 10[基金项目]国家自然科学基金项目(No.30470642,No.306- 70780,No.31071014,No.81100260,No.81070305 );山东省科技攻关项目(2008GG10002006);山东省卫生厅项目(2007HZ026)和青岛市科技局项目(05-1-JC-93,11-2-3-3-(2)- nsh)[作者简介]齐玉霞(1973-),女,硕士研究生,讲师。[通讯作者]徐珞(1954-),女,博士,教授,博士生导师。E-mail:xu.luo@1 63.com。g hrelin在海马调控下丘脑室旁核胃牵张敏感神经元活动中作用 齐玉霞1,2,徐珞1 (1 青岛大学医学院病理生理学教研室,山东青岛 266021; 2 荏平县人民医院) [摘要] 目的 观察电刺激海马CA1区对下丘脑室旁核(PVN)胃牵张(GD)敏感神经元放电活动的影响,以及ghrelin在该通路中的调控作用。方法 采用细胞外记录神经元单位放电方法,观察电刺激海马CA1区、ghrelin及其受体阻断剂[D-Lys-3]-GHRP-6对大鼠下丘脑PVN内GD敏感神经元放电活动的影响。结果 在PVN记录到的109个GD敏感神经元中,有71个为GD兴奋性(GD-E)神经元,38个为GD抑制性(GD-I)神经元。在GD-E神经元中,微量注射ghrelin可兴奋其中72%的神经元,放电频率增加(38.9±7.3)%(t=2.85,P<0.01);而在GD-I神经元中,微量注射ghrelin可抑制其中60%的神经元,放电频率减少(45.2±6.3)%(t=3.08,P<0.01);gh-relin的效应可被[D-Lys-3]-GHRP-6阻断。在41个对ghrelin有兴奋反应的GD-E神经元和15个对ghrelin有抑制反应的GD-I神经元中,电刺激海马CA1区,可分别兴奋39%的GD-E和33%的GD-I神经元,其中44%的GD-E神经元的兴奋效应可被[D-Lys-3]-GHRP-6部分阻断。结论 海马CA1区可以调控PVN内GD敏感神经元的活性,g hrelin能神经纤维参与了该通路的调控。[关键词] 海马;下丘脑室旁核;g hrelin;胃牵张敏感神经元;大鼠[中图分类号] R338.2 [文献标志码] A [文章编号] 1672-4488(2012)05-0377- 04THE EFFECTS OF GHRELIN ON THE HIPPOCAMPUS REGULATING THE NEURONS OF PVN IN RATS QI Yuxia,XU Luo(Department of Pathophysiology,Qingdao University  Medical College,Qingdao 266021,China)[ABSTRACT] Objective To explore the effect of electric stimulation of CA1area on the activity of gastric distention(GD)sensitive neurons in PVN and the role of ghrelin in this nerve pathway. Methods The effects of ghrelin on GD sensitive neuronsin PVN and the effects of electric stimulation of CA1area on the activity of these neurons were observed by recording extracellularp otentials of single neurons.The effects of antagonist of ghrelin-[D-Lys-3]-GHRP-6were also observed to explore the receptor in-volved. Results In 109GD sensitivity neurons recorded by the PVN,71were classified as GD-excitatory(GD-E)neurons,and38were GD-inhibitory(GD-I).Microinjection of ghrelin excited 72%GD-E neurons,and discharge frequency increased(38.9±7.3)%(t=2.85,P<0.01);in GD-I neurons,microinjection of ghrelin could inhibit 60%of the neurons,and discharge frequencydecreased(45.2±6.3)%(t=3.08,P<0.01).The effect of ghrelin could be blocked by[D-Lys-3]-GHRP-6.Among 41GD-Eneurons excited by ghrelin and 15GD-I neurons inhibited by ghrelin,electric stimulation of CA1area of hippocampus could stimu-late 39%of GD-E neurons and 33%of GD-I neurons,respectively,in which,44%of excitement effect of GD-E could be partiallyblocked by[D-Lys-3]-GHRP-6. Conclusion The neurons of the hippocampus CA1area can regulate the activity of GD sensitiveneurons in PVN and this effect is mediated by ghrelin-energy  fibers.[KEY WORDS] hippocampus;paraventricular hypothalamic nucleus;ghrelin;g astric distention sensitive neurons;rats 海马是边缘系统的一个重要的整合中枢, 它主要参与认知及学习记忆功能的调节。近年的研究表明, 海马在摄食、胃运动、能量代谢的调节中同样发挥着重要的作用[1- 3]。形态学研究表明,海马与杏仁 核、下丘脑、延髓等中枢脑区有着丰富的纤维联系, 这些神经核团共同作用,通过对内脏传入传出信号 的整合处理, 完成对摄食相关功能的调控[4] 。下丘脑室旁核(PVN)是中枢内调节能量代谢的重要脑区,是消化功能传入传出信息的重要转换站。PVN内的多种神经肽参与摄食行为和能量平衡的调节, 主要包括NPY、g hrelin、motilin等[5-8] 。其中ghre-lin是1999年发现的一种由28个氨基酸组成的脑 肠肽,是一种生长激素促分泌激素受体(GHSs-R)的内源性配体。ghrelin除了分布在胃、肠、胰、肾等外周组织器官之外,在下丘脑、垂体等中枢部位亦有 表达[ 9- 10],主要参与摄食、能量平衡、胃酸分泌等的调

Ghrelin采食作用及分泌调节的研究进展

Ghrelin采食作用及分泌调节的研究进展 耿春银1,杨连玉2,蒋友3,张敏1,严昌国1 1. 延边大学农学院动科系,吉林龙井(133400) 2.吉林农业大学动物科技学院,长春(130118) 3.新万发镇中学,吉林松原(131215) E-mail:gcy1011@https://www.360docs.net/doc/6c2539524.html, 摘要:Ghrelin是从鼠和人的胃中分离得到的一种生长激素释放激素受体的天然的内源性配体,它由28个氨基酸残基组成,且氮端第三位丝氨酸发生了辛酰基化,Ghrelin 具有广泛的生物学功能,包括调控动物采食、调节生长激素释放、调节胃肠功能及调节能量平衡等,对Ghrelin的研究具有重要的理论意义和广阔的应用前景,本文综述了Ghrelin采食作用及对其分泌调节的研究进展。 关键词:Ghrelin;生长激素;生长激素促分泌素;综述 生长激素(GH)是由脑垂体释放的一种调控动物生长的激素,它的释放主要由促生长激素释放激素(GHRH)和生长抑素(SS)进行调控。近年来,人们通过对生长激素促释放素(GHS)的研究,发现了一种新的调控GH释放的激素—Ghrelin。Ghrelin是生长激素促释放激素受体(GHS-R)的内源性配体,通过与其受体结合,能强烈的促进动物的采食,并能刺激GH的释放及促进胃酸分泌。除上述功能外,人们又陆续的发现,Ghrelin在能量平衡、胃功能、心脑血管系统、记忆、睡眠,以及肿瘤生长方面都显现出了重要的生物学效应。随着Ghrelin的发现、分离及对其研究的深入,Ghrelin在人类疾病的诊治及在动物生产中已展现出广阔的应用前景,本文对Ghrelin对动物的采食作用及对其分泌调节的研究进展情况进行了综述,为对其在生产中的应用奠定理论基础。 1. Ghrelin的结构及其分布 Ghrelin由28个aa残基组成,分子量是3314。人和大鼠的Ghrelin前体蛋白由117个氨基酸组成,N-端前23肽呈现分泌信号肽的特征。GhrelinN端前4个氨基酸片段为其最小的活性中心,C末端的P-R结构(脯氨酸一精氨酸)为其识别部位。人和大鼠Ghrelin除了两个氨基酸不同外,其余都是相同的,而且,哺乳动物氮端前10个氨基酸是完全一致的。由于不同的结合机制,体内有两种Ghrelin的前体,一种为Ghrelin,另一种被命名为des-Gnl14-ghrelin,它除了氮端第14位上谷氨酸缺失外,与Ghrelin结构及功能基本一致,并且此两种活性形式都可在胃中产生,但胃中des-Gnl14-ghrelin的浓度很低,表明正常的Ghrelin为其主要的活性形式。Ghrelin在体内有两种分泌形式,一种是Ghrelin氮端第三位丝氨酸发生了辛酰基化,一种没有发生辛酰基化,辛酰基化是Ghrelin发挥生物学功能的实质部位,而去辛酰基化的Ghrelin(des-acyl ghrelin)的作用还不是很清楚,但在大鼠胃中的浓度也很高。 Ghrelin主要在动物的胃内产生,而肠、胰腺、垂体、肾和胎盘也可以分泌少量。切除老鼠的胃或胃酸产生部位减少80%的循环ghrelin,这进一步的说明胃是ghrelin的主要来源部位。在老鼠体内,从胃到结肠,ghrelin在胃基底部的量是最大的,Ghrelin 的免疫反应性的细胞在十二指肠,空肠,回肠,结肠的黏膜上都存在,在肠内,ghrelin的浓度从十二指肠到结肠逐渐的减少。下丘脑的神经元到邻近的第三脑室之间的背侧,腹侧室旁核(PVN)和弓形的下丘脑神经核也发现存在ghrelin,此外在唾液中也检测到Ghrelin的存在。随着研究的深入,可能在动物的其他组织中还会检测到Ghrelin的存在。有趣的是,最近有人报道了原本只在动

神经肽Ghrelin和Nesfatin-1的研究进展

神经肽Ghrelin和Nesfatin-1的研究进展(作者:___________单位: ___________邮编: ___________) 作者:刘诤,王峰,孙涛,齐江华 【关键词】 Ghreli;Nesfatin-1;摄食;生长激素;酰基化 具有调节能量平衡和摄食功能的肽类激素Ghrelin和Nesfatin-1近年来被越来越多地证实参与、影响了学习、记忆、睡眠、癫痫等多种脑功能,已引起人们广泛的关注,成为研究的热点。 1发现及命名 自20世纪70年代末Momany和Bowers合成了一种能够促进生长激素分泌的多肽——生长激素释放肽(Growth Hormone Releasing Peptides,GHRP)以来,人们陆续合成了一组具有相同作用的物质:1993年,Smith等根据GHP-6三维结构合成出非肽类的生长激素促分泌素(Growth Hormone Secretagogues,GHSs),并根据GHSs在体内的信号传导机制与GHRP的不同,推测体内存在着GHSs的特殊受体;果然,1996年Howards等在人类垂体等部位发现并成功克隆出GHSs 的受体——生长激素促分泌素受体(Growth Hormone Secretagogues Receptor, GHSR)[1];在此基础上,1999年日本科学家KOJIMA利用免疫组化方法在小鼠和人的胃内分泌细胞及下丘脑弓状核新发现

了一种含有28个氨基酸残基的多肽,它是GHSR天然的内源性配体,当这种多肽与GHSR结合后, 它能通过一种G蛋白偶联受体刺激垂体释放生长激素,因此将其命名为Ghrelin[2]。Ghre在古印欧语系中有“生长(grow)”之意,所以国内将Ghrelin翻译为生长素。Ghrelin 成为继GHRP和生长抑素(SS)之后调节生长激素分泌的又一个主要激素。 2006年,日本OH-I教授在《Nature》上首次报告了作为核组蛋白NUCB2派生前体的神经肽Nesfatin-1存在于下丘脑中。最初的实验发现如果将NUCB2重组蛋白注入大鼠脑室内,其体重会明显降低,且摄食呈剂量依赖性减少,故将此分泌蛋白命名为Nesfatin(有“减肥”之意)。进一步研究发现在激素原转化——酶作用下,Nesfatin可以水解为3个片段,分别被命名为:Nesfatin-1、Nesfatin-2和Nesfatin-3,Nesfatin-1位于NUCB2的N端,是由前82个氨基酸组成的肽段,在下丘脑分泌后进入脑脊液中,最终进入血循环[3]。2结构及分泌 Ghrelin由28个氨基酸组成,分子量3,314D。人类Ghrelin氨基酸排列顺序是G-S-S-F-L-S-P-E-H-Q-R-V-Q-Q-R-K-E-S-K-K-P-P-A-K-L-Q-P-R,基因排序位于3号染色体短臂(3p25-26),包含6个外显子和5个内含子。它在体内有两种存在形式,一种是N端去辛酰基化的,另一种是N端第3位丝氨酸残基辛酰基化的(即该位丝氨酸的N端与十八辛酸以酯键连接),后者是其促GH分泌活性所必需的。辛酰基化后的

Ghrelin 与高级神经功能的研究进展

Ghrelin 与高级神经功能的研究进展 Ghrelin 是一种由28 个氨基酸残基组成的新型内源性脑肠肽,主要由胃底黏膜分泌,是生长激素促泌 素受体(growth hormone secretagogue receptor,GHSR)的内源性配体。Ghrelin 及其受体在外周组织及中枢神经系统中广泛分布,发挥着重要的生物学作用,如促进生长激素的释放、增加食欲、调节能量代谢、抑制炎症反应及调节免疫功能等。近年研究表明,ghrelin 能够增强学习和记忆能力,具有神经保护作用,在中枢神经系统疾病治疗中显示出良好的应用前景。本文就ghrelin 结构、分布、在高级神经功能方面的研 究进展等做一综述。 1 Ghrelin 及GHSR 的结构 1999 年日本学者Kojima 等[1]在大鼠胃中发现了一种新的内源性脑肠肽并将其命名为ghrelin。“ghre” 在古印欧语中是“生长”的意思,“lin”则是多肽类激素的后缀(如insulin,vasopressin 等)。我国学者将其翻 译为胃促生长素、饥饿激素、胃饥素等。Ghrelin 是生长激素促泌素受体(growth hormone secretagogues receptor,GHSR)的内源性配体,是除生长激素释放激素和生长抑素外,目前人们发现的第3 种调节腺垂体生长激素分泌的内源性物质。ghrelin 前体是由117 个氨基酸组成的多肽,人和大鼠具有高度的同源性(82.9%)。大鼠ghrelin 的分子量为3314Da,其前体N 端的前23 个氨基酸具有分泌信号肽的特征,从24 位 的甘氨酸开始到第51 位的28 个氨基酸为ghrelin 序列: H-Gly-Ser-Ser-Phe-Leu-Ser-Pro-Glu-His-Gln-Lys-Ala-Gln-Gln-Arg-Lys-Glu-Ser-Lys-Lys-Pro-Pro-Ala-Lys-Leu- Gln-Pro-Arg-OH。Ghrelin 有两种存在形式,即第3 位的丝氨酸残基N 端辛酰基化和去N 端辛酰基化,且 第 3 位的丝氨酸残基N 端辛酰基化是保持其活性所必需的。只有辛酰基化的ghrelin 才能与其受体结合,进入中枢神经系统发挥作用[2]。GHSR 是一种G 蛋白偶联的蛋白质受体超级家族的一员,具有1α和l β两 种亚型。GHSR1α含有366 个氨基酸,有7 个跨膜域,是GHSR 的完整功能形式,研究表明ghrelin 的大 多数生理作用是通过GHSR-lα介导的,而GHSR-lβ缺少第 6 和第7 个跨膜链,无法与ghrelin 结合,也不 具备信号传递能力,其确切功能有待进一步研究[3]。 2 Ghrelin 及GHSR 的分布 Ghrelin 主要由胃黏膜的内分泌细胞产生,人类甲状腺、小肠、胰腺、心脏、肺、肝、肾、卵巢、胎盘 及免疫系统等处也有少量分泌[4,5]。在中枢神经系统,ghrelin 的主要合成部位在下丘脑[6]。研究表明,ghrelin 及GHSR 主要分布在丘脑弓状核、腹内侧核、背内侧核、漏斗核、室旁核和第三脑室的室管膜层。此外,大脑皮质、海马区、感觉运动神经层及背侧迷走神经核等也有一定的表达[7,8]。这提示ghrelin 及GHSR 可能在中枢神经系统发挥重要的生物功能。 3 Ghrelin 与认知功能 Ghrelin 与GHSR 结合后产生一系列生物学效应包括刺激垂体前叶释放生长激素、调节机体生长发育、 增加食欲[9]、调节能量平衡[10]、调节胃肠道功能[11]、抑制炎症反应及调节免疫功能等[12]。Ghrelin 在高级神经功能方面的作用主要表现在以下两个方面: 3.1 增强学习和记忆能力 Carlini 等人[13]将ghrelin 注入大鼠海马、杏仁核及中缝背核,发现ghrelin 能增加大鼠的记忆保留时间 1

人饥饿素ghrelinELISA试剂盒使用说明书

人饥饿素 (ghrelin)ELISA试剂盒使用说明书我司ELISA试剂盒品质保证,质量优,价格实惠,是您生物实验的首选,如有需要可与我司销售人员联系。 本试剂仅供研究使用目的:本试剂盒用于测定人血清,血浆及相关液体样本中人饥饿素(ghrelin)含量。 实验原理: 本试剂盒应用双抗体夹心法测定标本中人饥饿素(ghrelin)水平。用纯化的人饥饿素(ghrelin)抗体包被微孔板,制成固相抗体,往包被单抗的微孔中依次加入饥饿素(ghrelin),再与HRP标记的饥饿素(ghrelin)抗体结合,形成抗体-抗原-酶标抗体复合物,经过彻底洗涤后加底物TMB显色。TMB在HRP酶的催化下转化成蓝色,并在酸的作用下转化成最终的黄色。颜色的深浅和样品中的饥饿素(ghrelin)呈正相关。用酶标仪在450nm波长下测定吸光度(OD值),通过标准曲线计算样品中人饥饿素(ghrelin)浓度。 试剂盒组成: 1.血清:室温血液自然凝固10-20分钟,离心20分钟左右(2000-3000转/分)。仔细收集上 清,保存过程中如出现沉淀,应再次离心。 2.血浆:应根据标本的要求选择EDTA、者柠檬酸钠或肝素作为抗凝剂,混合10-20分钟后, 离心20分钟左右(2000-3000转/分)。仔细收集上清,保存过程中如有沉淀形成,应该再次离心。 3.尿液:用无菌管收集,离心20分钟左右(2000-3000转/分)。仔细收集上清,保存过程中 如有沉淀形成,应再次离心。胸腹水、脑脊液参照实行。 4.细胞培养上清:检测分泌性的成份时,用无菌管收集。离心20分钟左右(2000-3000转/分)。仔细收集上清。检测细胞内的成份时,用PBS(PH7.2-7.4)稀释细胞悬液,细胞浓

Ghrelin与心血管病研究进展

Ghrelin与心血管疾病的研究进展 学号:14214289 姓名:李菲菲院系:附属第一医院心内科 字数3707 摘要 Ghrelin是一种新型的脑肠肽,其受体广泛的表达于心血管系统,发挥抑制炎症因子释放、保护血管内皮细胞、降低血压、扩张冠状动脉、增加心肌供血、增强心肌收缩力、改善心力衰竭症状等多种生物学效应。作为一种小分子多肽, ghrelin参与高血压病、冠状动脉粥样硬化性心脏病、心力衰竭等多种心血管疾病的发生、发展。 Abstract Ghrelin is a new type of brain-gut peptide, and its receptor is widely expressed in the cardiovascular system.It can inhibit the inflammatory factors release,protect vascular endothelial cells,reduce blood pressure,expand the coronary artery, increase myocardial blood supply, enhance myocardial contractility, improve heart failure symptoms and other biological effects. As a kind of small peptide, ghrelin is involved in heart failure, coronary heart disease, hypertension and other cardiovascular diseases’development. 关键字 Ghrelin 高血压病冠状动脉粥样硬化性心脏病心力衰竭 Keywords Ghrelin Hypertension Coronary heart disease Heart failure Ghrelin是 Kojima等[1]发现的一种肠肽类激素, 是含有28个氨基酸残基的多肽,是生长激素促分泌物受体的内源性配体。研究发现,ghrelin通过与其受体生长激素促分泌物受体( growth hormone secrelagogue receptor, GHS-R) 相结合而发挥多种生物学作用, 不仅可促进生长激素释放、增加摄食、调节脂肪代谢,还具有抑制炎症因子释放、保护血管内皮细胞、降低血压、扩张冠状动脉、增加心肌供血、增强心肌收缩力、改善心力衰竭症状等多种生物学效应。作为一种小分子多肽, ghrelin 在心血管疾病的发生、发展中发挥一定作用。本文就近年来 ghrelin与心血管疾病的研究进展作一综述。 1.Ghrelin的简介 1.1 Ghrelin来源及结构胃肠道黏膜层的内分泌腺细胞是ghrelin的主要来源, 循环中的ghrelin主要来自胃[2]。下丘脑弓状核、垂体、脑干等中枢神经系统含有ghrelin免疫活性神经元。有研究显示,人胎盘组织、胰岛细胞、甲状腺滤泡旁细胞、淋巴细胞、小鼠肾小球肾细胞, 以及大鼠睾丸细胞,均能分泌 ghrelin或有ghrelin mRNA的表达[3]。人和大鼠的ghrelin前体( proghrelin)均由 117个氨基酸组成, 前23位为信号肽。Ghrelin由第24 52

相关主题
相关文档
最新文档