当前位置: 首页 > 期刊 > 《糖尿病学杂志》 > 2004年第1期 > 正文
编号:10584165
Plasma Ghrelin Concentrations Are Not Regulated by Glucose or InsulinA Double-Blind Placebo-Controlled Crossover Clamp Study
http://www.100md.com 《糖尿病学杂志》2004年第1期
     1 Department of Clinical Pharmacology, Vienna University, Vienna, Austria$2:^}8/, 百拇医药

    2 Department of Endocrinology and Metabolism, Vienna University, Vienna, Austria$2:^}8/, 百拇医药

    3 Ludwig Boltzmann Institute for Experimental Endocrinology, Vienna University, Vienna, Austria$2:^}8/, 百拇医药

    ABSTRACT$2:^}8/, 百拇医药

    Ghrelin plasma concentrations increase during fasting and fall rapidly after nutrient ingestion. We hypothesized that insulin or glucose could regulate ghrelin secretion by a feedback mechanism. In this randomized, double-blind, placebo-controlled crossover study, three different study days were carried out in nine healthy volunteers (age 26 ± 6 years). On each day, stepwise increasing systemic glucose concentrations of 5.0, 8.3, and 11.1 mmol/l were attained by intravenous infusion of glucose, representing fasting and postprandial conditions. Ghrelin plasma concentration was studied during concomitant exogenous hyperinsulinemia, inhibition of endogenous insulin production by somatostatin infusion, and placebo time control, respectively. Elevated glucose concentrations increased circulating insulin to 612 ± 85 pmol/l (P < 0.01), but they did not affect ghrelin concentrations. Prolonged hyperinsulinemia by exogenous infusion resulted in circulating insulin of 1,602 ± 261 pmol/l (P < 0.01) and suppressed plasma ghrelin to 49.6% of baseline (P < 0.01). During administration of somatostatin, insulin concentration remained constant, but an even greater decrease in ghrelin to 39.5% of baseline was noted (P < 0.01). Hyperglycemia does not decrease ghrelin, and a reduction in ghrelin is only seen at supraphysiological insulin concentrations. In contrast, systemic ghrelin concentrations are decreased by somatostatin. The meal-related suppression of ghrelin appears not directly regulated by glucose or insulin.

    The 28–amino acid peptide ghrelin has been identified as the endogenous ligand of the pituitary growth hormone (GH) secretagogue receptor (1). Ghrelin is produced by a distinct endocrine cell type primarily in the gastric fundus (2,3) and increases GH levels in both animals (4) and humans (5–7). In addition, ghrelin exerts potent orexigenic and adipogenic effects (8,9), which are thought to be mediated by action on hypothalamic neurons (10–12). Ghrelin is assumed to be involved in a feedback loop regulating food consumption and meal initiation. However, access of peripherally secreted ghrelin to regions in the brain is limited by specific and selective transport mechanisms through the blood-brain barrier (13)./gu, 百拇医药

    There is experimental evidence that food intake and body weight increase rapidly in rodents treated with ghrelin (4,10), and ghrelin levels rise in fasting animals and humans (10,11,14). A postprandial fall of ghrelin plasma levels has been demonstrated in both animals (15) and humans (16,17). Plasma ghrelin is elevated in cachectic patients with chronic heart failure (18), bulimia (19), or anorexia nervosa, where weight gain substantially reduces ghrelin concentrations (20). Conversely, ghrelin concentrations are suppressed in obese subjects (21) and increased by weight loss (22). However, food intake fails to decrease ghrelin levels in obese patients (23). Mutations in the human preproghrelin/ghrelin gene may be associated with obesity (24,25), but the influence of genetic polymorphisms on weight regulation is under debate (26).

    The mechanisms controlling ghrelin secretion during fasting and postprandial suppression are unknown. Ghrelin levels were found to be reciprocal to those of glucose and insulin (14). Glucose or insulin might therefore regulate ghrelin release. To test this hypothesis, plasma ghrelin and GH concentrations were quantified during a glucose clamp at normal and high blood glucose concentrations, representing fasting and postprandial states. This was studied on three separate study days, under conditions of concomitant hyperinsulinemia on the first day, during inhibition of endogenous insulin production by somatostatin on the second day, and serving as placebo control on the third study day.ty3/+%, http://www.100md.com

    RESEARCH DESIGN AND METHODSty3/+%, http://www.100md.com

    The study was approved by the ethics committee of the University of Vienna and complies with the principles outlined in the Declaration of Helsinki, including current revisions and the Good Clinical Practice guidelines. Written informed consent was obtained from all subjects before enrolment.

    Subjects./io, http://www.100md.com

    Nine healthy male volunteers were included in this study. The subjects were between the ages of 19 and 36 years (mean ± SD, 26 ± 6 years) and had a BMI between the 15th and 85th percentile (27). All subjects were nonsmokers and drug free. In a complete health examination (including physical examination, electrocardiography, laboratory screening, and an oral glucose tolerance test) within 14 days before the first study day, no clinically relevant abnormalities were detected. Subjects were studied after overnight fasting, and experiments were started between 0800 and 0900. Studies were conducted in a quiet room with an ambient temperature of 22°C./io, http://www.100md.com

    Experimental protocol./io, http://www.100md.com

    The study used a double-blind, placebo-controlled three-way crossover design. On three different study days separated by a washout period of at least 2 days, all subjects received infusions of insulin, somatostatin, or placebo, respectively. This was accompanied by a coinfusion of glucose to achieve stepwise increased venous glucose concentrations of 5.0, 8.3, and 11.1 mmol/l for a period of 90 min each on all three study days.

    Intravenous cannulas were inserted into opposite arms for blood sampling and drug infusions, respectively. Baseline blood samples were drawn, and infusions were started and maintained for 270 min: insulin mixed with heparinized blood (to avoid adsorption to the syringe and to connecting tubing) (28) at 1.0 mU · kg-1 · min-1 (Huminsulin; Lilly, Fegersheim, France), somatostatin at 61.1 pmol · kg-1 · min-1 (UCB Pharma, Vienna, Austria), or placebo (0.9% saline). Blood glucose concentrations were determined every 5 min throughout the study (glucose analyzer; Beckman, Fullerton, CA), and glucose infusion rates (10% glucose; Mayerhofer Pharmazeutika, Linz, Austria) were adjusted accordingly.?jkv&, 百拇医药

    Laboratory parameters.?jkv&, 百拇医药

    Blood samples for quantification of ghrelin were drawn every 30 min. Ghrelin was analyzed using a commercially available kit (Peninsula Laboratories, San Carlos, CA). Venous insulin, human GH, and IGF-1 concentrations were determined at baseline and at the end of the three different glucose level periods. Laboratory analyses were carried out according to standard procedures at the Department of Medical and Chemical Laboratory Diagnostics, Allgemeines Krankenhaus, Vienna.

    Systemic hemodynamics.\e!t, http://www.100md.com

    Blood pressure was measured by an automated oscillometric device on the upper arm at 15-min intervals during the study periods, and the pulse rate was monitored continuously by a finger pulse–oxymetric device (CMS patient monitor; Hewlett Packard, Palo Alto, CA) (29).\e!t, http://www.100md.com

    Statistical analysis.\e!t, http://www.100md.com

    The individual area under the ghrelin concentration versus time curve (AUCG) was calculated for each 90-min interval of different glucose concentrations using Kinetica software (release 3.0; InnaPhase, Philadelphia, PA). Because of the skewed distribution of the parameters under study, nonparametric tests were used. Between-group comparisons were performed using the Mann-Whitney U test. Within groups, effects were tested by Friedman’s ANOVA and the Wilcoxon’s signed-rank test, respectively. All calculations were performed using a Statistica software package (release 5.1; StatSoft, Tulsa, OK). P ≤ 0.05 was considered significant. Values are expressed as the means ± SE unless indicated otherwise.

    RESULTS4, http://www.100md.com

    The drugs under study were well tolerated, and no side effects were reported. No differences in blood pressure or pulse rate were observed at baseline between the three study days, and the drugs under study had no effect on systemic hemodynamics (data not shown). Fasting glucose concentrations were comparable at baseline and in the normal range. The scheduled systemic glucose concentrations of 5.0, 8.3, and 11.1 mmol/l were achieved on all trial days . However, the dose of exogenous glucose required was much higher during coinfusion of insulin than during placebo or somatostatin. During hyperinsulinemia, a mean of 2,269 ± 168 ml of a 10% glucose solution was infused over 270 min, whereas 753 ± 69 ml were administered during placebo and 382 ± 32 ml during somatostatin, respectively (P < 0.001 between groups). Baseline insulin or ghrelin concentrations were not different between the study days .4, http://www.100md.com

    fig.ommtted4, http://www.100md.com

    Coinfusion of glucose with placebo. Changes in glucose (, insulin, and ghrelin plasma concentrations over time are presented as means ± SE (n = 9).

    fig.ommtted*:q^%], http://www.100md.com

    Coinfusion of glucose with somatostatin. Changes in glucose , insulin , and ghrelin plasma concentrations over time are presented as means ± SE (n = 9).*:q^%], http://www.100md.com

    Effect of hyperglycemia.*:q^%], http://www.100md.com

    During placebo, hyperglycemia caused an increase of endogenous insulin, from 69 ± 13 pmol/l to a maximum of 612 ± 85 pmol/l (P < 0.01) at the end of the study . Increased glucose concentrations did not affect ghrelin concentrations, which were 193 ± 29 pmol/l at baseline and 220 ± 49 pmol/l at the end of the study (P = NS). The AUCG was 16.74 ± 2.18, 18.03 ± 2.81, and 17.27 ± 2.24 nmol · min-1 · l-1 at glucose concentrations of 5.0, 8.3, and 11.1 mmol/l, respectively (P = NS). GH concentrations decreased slightly during hyperglycemia (P = NS). This was paralleled by a significant decrease in IGF-1 concentrations (P < 0.05).*:q^%], http://www.100md.com

    fig.ommtted*:q^%], http://www.100md.com

    Human GH and IGF-1 plasma concentrations at baseline and during various venous glucose concentrations with coinfusion of placebo, insulin, or somatostatin

    Effect of exogenous hyperinsulinemia.q*zh'[m, 百拇医药

    Exogenous insulin increased circulating insulin concentrations, from a baseline of 63 ± 7 pmol/l to a maximum of 1,602 ± 261 pmol/l during hyperglycemia (P < 0.01) . Ghrelin concentrations decreased over time to 49.6% of baseline, from a mean of 246 ± 43 pmol/l under euglycemic conditions to 122 ± 19 pmol/l during hyperglycemia (P < 0.01). The AUCG was 13.06 ± 1.58 nmol · min-1 · l-1 during euglycemia (P = NS), 16.31 ± 2.44 nmol · min-1 · l-1 at a glucose concentration of 8.3 mmol/l (P = NS), and 10.52 ± 1.39 nmol · min-1 · l-1 during hyperglycemia at a glucose concentration of 11.1 mmol/l, which was significantly smaller than during placebo conditions (P < 0.05). GH and IGF-1 concentrations significantly decreased during hyperglycemia (P < 0.05).q*zh'[m, 百拇医药

    fig.ommttedq*zh'[m, 百拇医药

    Coinfusion of glucose with insulin. Changes in glucose (, insulin , and ghrelin plasma concentrations over time are presented as means ± SE (n = 9).

    Effect of somatostatin.s;, http://www.100md.com

    Systemic insulin concentrations were unchanged during somatostatin administration . Ghrelin concentrations decreased from 147 ± 22 pmol/l at baseline to 58 ± 10 pmol/l at the end of glucose infusion (39.5% of baseline, P < 0.01). The AUCG was 11.28 ± 1.61 nmol · min-1 · l-1 during euglycemia (P < 0.06 vs. placebo), 8.41 ± 1.11 nmol · min-1 · l-1 at glucose concentrations of 8.3 mmol/l (P < 0.01), and 6.58 ± 1.01 nmol · min-1 · l-1 at glucose concentrations of 11.1 mmol/l (P < 0.001 vs. placebo). GH and IGF-1 concentrations decreased by coinfusion of somatostatin with glucose (P < 0.05) .s;, http://www.100md.com

    DISCUSSIONs;, http://www.100md.com

    This randomized controlled study suggests that systemic ghrelin concentrations are not directly regulated by changes in circulating glucose or insulin at physiological concentrations, but rather by somatostatin. Ghrelin concentrations were not affected by glucose concentrations of up to 11.1 mmol/l and an increase of insulin to ~ 620 pmol/l.

    Our findings also confirm results of a recent study in which intravenous glucose coadministered with subcutaneous insulin failed to suppress ghrelin concentrations (30). In contrast, other studies have reported a rapid, but transient, decrease in plasma ghrelin after a high-dose glucose bolus (21) or after food intake (17). This reciprocal pattern between glucose and ghrelin concentrations was not reproducible in our controlled study. Although it is possible that changes in ghrelin within periods shorter than 30 min could have been missed in our experiments, it is unlikely that increased glucose results in ghrelin suppression in healthy subjects when elevated to supraphysiological concentrations.8yx5(, http://www.100md.com

    A reduction in ghrelin was seen during pharmacological hyperinsulinemia, at an insulin concentration of 1,600 pmol/l, with concomitant hyperglycemia. However, hyperinsulinemia at concentrations typically seen in insulin-resistant subjects did not affect plasma ghrelin in our experiments. This is compatible with findings in patients with type 2 diabetes, where ghrelin concentrations were in the normal range in lean subjects and only decreased in obese patients (21). In contrast, a recent uncontrolled study reports a decrease in ghrelin concentrations during euglycemia at insulin concentrations of ~ 600 pmol/l (31). Of note, there is substantial variability of circulating ghrelin concentrations, and changes over time can be easily overinterpreted. On the basis of our data using similar insulin concentrations during euglycemia and hyperglycemia, it seems that insulin substantially decreased plasma ghrelin only at pharmacological concentrations. Glucose in combination with supraphysiological insulin concentrations might cross the blood-brain barrier more easily and influence central regulation of gastric ghrelin release. On the other hand, upregulation of ghrelin expression (11) and an increase of plasma ghrelin (32) have also been reported in animals after the administration of insulin. This was not confirmed in the healthy subjects under study, indicating that insulin is not responsible for the reduction in ghrelin concentrations after food intake, nor is it responsible for the observed increase after insulin administration.

    Somatostatin suppressed the hyperglycemia-induced increase in endogenous insulin and reduced ghrelin concentrations significantly. This effect on ghrelin was apparent during euglycemia, and it was more pronounced during hyperglycemia. The placebo control experiment suggests that this finding is due to a time-dependent effect rather than to hyperglycemia. The decrease in ghrelin concentrations may reflect a direct inhibitory action of somatostatin on ghrelin-releasing peripheral cells and may indicate a continuous constitutive release of ghrelin under normal conditions. It is well known that somatostatin inhibits hormone release from neuroendocrine cells (33) and that the gastrointestinal mucosa expresses somatostatin receptors (34). Thus, there could also be an interaction between somatostatin and the ghrelin-producing cells, but so far there is no direct evidence to support this hypothesis. Furthermore, the functional results obtained do not elucidate which cells are involved in the endocrine responses. The tissue distribution of ghrelin is widespread (35), and the physiological significance of this distribution is still unclear.

    Apart from glucose or insulin, several other regulatory mechanisms appear to be involved in the regulation of plasma ghrelin after ingestion. The autonomic nervous system may play a role because vagotomy increases plasma ghrelin levels, and the vagal tone is low in fasting animals (32). Stimulation of gastric mechanoreceptors is probably of minor clinical importance, because ghrelin plasma concentrations were only reduced by ingestion of nutrients and not by the same volume of water in animals (8) and in humans (21). However, gastric bypass surgery abrogates the normal meal-related fluctuations and diurnal rhythm and decreases ghrelin concentrations (17). The potential roles of amino acids or lipids have been less investigated, but they may alter ghrelin release directly by luminal contact (36) or by increased plasma concentrations. This possibility needs to be addressed in future studies.n!'d, 百拇医药

    Hyperglycemia is known to inhibit GH secretion in normal subjects (37). This was also seen in our study, in which a small decrease in GH and IGF-1 was observed during hyperglycemia. This effect was more pronounced during insulin or somatostatin coinfusion, suggesting the involvement of different mechanisms. Because IGF-1 concentrations were also decreased at glucose concentrations at which no changes in GH could be detected, a direct action of glucose on IGF-1 production seems likely.

    In conclusion, ghrelin plasma concentrations were significantly decreased by somatostatin. In contrast, no direct involvement of glucose or insulin at physiological concentrations in the regulation of ghrelin plasma concentrations in healthy humans was demonstrable.1'uw, http://www.100md.com

    ACKNOWLEDGMENTS1'uw, http://www.100md.com

    We are grateful for the assistance and administrative work of Carola Fuchs, RN.1'uw, http://www.100md.com

    REFERENCES1'uw, http://www.100md.com

    Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K: Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature402 :656 –660,19991'uw, http://www.100md.com

    Date Y, Kojima M, Hosoda H, Sawaguchi A, Mondal MS, Suganuma T, Matsukura S, Kangawa K, Nakazato M: Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology141 :4255 –4261,20001'uw, http://www.100md.com

    Rindi G, Necchi V, Savio A, Torsello A, Zoli M, Locatelli V, Raimondo F, Cocchi D, Solcia E: Characterisation of gastric ghrelin cells in man and other mammals: studies in adult and fetal tissues. Histochem Cell Biol117 :511 –519,2002

    Wren AM, Small CJ, Ward HL, Murphy KG, Dakin CL, Taheri S, Kennedy AR, Roberts GH, Morgan DG, Ghatei MA, Bloom SR: The novel hypothalamic peptide ghrelin stimulates food intake and growth hormone secretion. Endocrinology141 :4325 –4328,2000|!dno, 百拇医药

    Arvat E, Di Vito L, Broglio F, Papotti M, Muccioli G, Dieguez C, Casanueva FF, Deghengi R, Camanni F, Ghigo E: Preliminary evidence that ghrelin, the natural secretagogue receptor ligand, strongly stimulates growth hormone secretion in humans. J Endocrinol Invest23 :493 –495,2000|!dno, 百拇医药

    Peino R, Baldelli R, Rodriguez-Garcia J, Rodriguez-Segade S, Kojima M, Kangawa K, Arvat E, Ghigo E, Dieguez C, Casanueva FF: Ghrelin-induced growth hormone secretion in humans. Eur J Endocrinol143 :R11 –R14,2000|!dno, 百拇医药

    Takaya K, Ariyasu H, Kanamoto N, Iwakura H, Yoshimoto A, Harada M, Mori K, Komatsu Y, Usui T, Shimatsu A, Ogawa Y, Hosoda K, Akamizu T, Kojima M, Kanagawa K, Nakao K: Ghrelin strongly stimulates growth hormone release in humans. J Clin Endocrinol Metab85 :4908 –4911,2000|!dno, 百拇医药

    Tschop M, Smiley DL, Heiman ML: Ghrelin induces adiposity in rodents. Nature407 :908 –913,2000

    Nakazato M, Murakami N, Date Y, Kojima M, Matsuo H, Kangawa K, Matsukura S: A role for ghrelin in the central regulation of feeding. Nature409 :194 –198,20011a)0]im, 百拇医药

    Asakawa A, Inui A, Kaga T, Yuzuriha H, Nagata T, Ueno N, Makino S, Fujimiya M, Nijima A, Fujino MA, Kasuga M: Ghrelin is an appetite stimulatory signal from the stomach with structural resemblance to motilin. Gastroenterology120 :337 –345,20011a)0]im, 百拇医药

    Toshinai K, Mondal MS, Nakazato M, Date Y, Murakami N, Kojima M, Kangawa K, Matsukura S: Upregulation of ghrelin expression in the stomach upon fasting, insulin-induced hypoglycemia, and leptin administration. Biochem Biophys Res Commun281 :1220 –1225,20011a)0]im, 百拇医药

    Shintani M, Ogawa Y, Ebihara K, Aizawa-Abe M, Miyanaga F, Takaya K, Hayashi T, Inoue G, Hosoda K, Kojima M, Kangawa K, Nakao K: Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes50 :227 –232,20011a)0]im, 百拇医药

    Banks WA, Tschop M, Robinson SM, Heiman ML: Extent and direction of ghrelin transport across the blood-brain barrier is determined by its unique primary structure. J Pharmacol Exp Ther302 :822 –827,2002

    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. Diabetes50 :1714 –1719,20012?zfh^, http://www.100md.com

    Hayashida T, Murakami K, Mogi K, Nishihara M, Nakazato M, Mondal Ms, Horii Y, Kojima M, Kangawa K, Murakami N: Ghrelin in domestic animals: distribution in stomach and its possible role. Domest Anim Endocrinol21 :17 –24,20012?zfh^, http://www.100md.com

    Tschop M, Wawarta R, Riepl RL, Friedrich S, Bidlingmaier M, Landgraf R, Folwaczny C: Post-prandial decrease of circulating human ghrelin levels. J Endocrinol Invest24 :RC19 –RC21,20012?zfh^, http://www.100md.com

    Cummings DE, Weigle DS, Frayo RS, Breen PA, Ma MK, Dellinger EP, Purnell JQ: Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med346 :1623 –1630,20022?zfh^, http://www.100md.com

    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. Circulation104 :2034 –2038,2001

    Tanaka M, Naruo T, Muranaga T, Yasuhara D, Shiiya T, Nakazato M, Matsukura S, Nozoe S: Increased fasting plasma ghrelin levels in patients with bulimia nervosa. Eur J Endocrinol146 :R1 –R3,2002var{{, 百拇医药

    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 Endocrinol145 :669 –673,2001var{{, 百拇医药

    Shiiya T, Nakazato M, Mizuta M, Date Y, Mondal MS, Tanaka M, Nozoe S, Hosoda H, Kangawa K, Matsukura S: Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J Clin Endocrinol Metab87 :240 –244,2002var{{, 百拇医药

    Hansen TK, Dall R, Hosoda H, Kojima M, Kangawa K, Christiansen JS, Jorgensen JO: Weight loss increases circulating levels of ghrelin in human obesity. Clin Endocrinol56 :203 –206,2002var{{, 百拇医药

    English PJ, Ghatei MA, Malik IA, Bloom SR, Wilding JP: Food fails to suppress ghrelin levels in obese humans. J Clin Endocrinol Metab87 :2984 –2987,2002var{{, 百拇医药

    Ukkola O, Ravussin E, Jacobson P, Snyder EE, Chagnon M, Sjostrom L, Bouchard C: Mutations in the preproghrelin/ghrelin gene associated with obesity in humans. J Clin Endocrinol Metab86 :3996 –3999,2001

    Korbonits M, Gueorguiev M, O’Grady E, Lecoeur C, Swan DC, Mein CA, Weill J, Grossman AB, Froguel P: A variation in the ghrelin gene increases weight and decreases insulin secretion in tall, obese children. J Clin Endocrinol Metab87 :4005 –4008,2002o+n:c^c, 百拇医药

    Hinney A, Hoch A, Geller F, Schafer H, Siegfried W, Goldschmidt H, Remschmidt H, Hebebrand J: Ghrelin gene: identification of missense variants and a frameshift mutation in extremely obese children and adolescents and healthy normal weight students. J Clin Endocrinol Metab87 :2716 –2719,2002o+n:c^c, 百拇医药

    Must A, Dallal GE, Dietz WH: Reference data for obesity: 85th and 95th percentiles of body mass index (wt/ht2) and triceps skinfold thickness. Am J Clin Nutr53 :839 –846,1991o+n:c^c, 百拇医药

    Kerchner J, Colaluca DM, Juhl RP: Effect of whole blood on insulin adsorption onto intravenous infusion systems. Am J Hosp Pharm37 :1323 –1325,1980o+n:c^c, 百拇医药

    Wolzt M, Schmetterer L, Rheinberger A, Salomon A, Unfried C, Breiteneder H, Ehringer H, Eichler HG, Fercher AF: Comparison of non-invasive methods for the assessment of haemodynamic drug effects in healthy male and female volunteers: sex differences in cardiovascular responsiveness. Br J Clin Pharmacol39 :347 –359,1995

    Caixas A, Bashore C, Nash W, Pi-Sunyer F, Laferrere B: Insulin, unlike food intake, does not suppress ghrelin in human subjects. J Clin Endocrinol Metab87 :1902 –1906,2002k-r$-, 百拇医药

    Saad MF, Bernaba B, Hwu CM, Jinagouda S, Fahmi S, Kogosov E, Boyadjian R: Insulin regulates plasma ghrelin concentration. J Clin Endocrinol Metab87 :3997 –4000,2002k-r$-, 百拇医药

    Lee HM, Wang G, Englander EW, Kojima M, Greeley GH Jr: Ghrelin, a new gastrointestinal endocrine peptide that stimulates insulin secretion: enteric distribution, ontogeny, influence of endocrine, and dietary anipulations. Endocrinology143 :185 –190,2002k-r$-, 百拇医药

    Zaki M, Harrington L, McCuen R, Coy DH, Arimura A, Schubert ML: Somatostatin receptor subtype 2 mediates inhibition of gastrin and histamine secretion from human, dog, and rat antrum. Gastroenterology111 :919 –924,1996k-r$-, 百拇医药

    Reubi JC: Somatostatin receptors in the gastrointestinal tract in health and disease. Yale J Biol Med65 :493 –503 [discussion 531–536],1992k-r$-, 百拇医药

    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 Metab87 :2988 –2991,2002k-r$-, 百拇医药

    Sakata I, Nakamura K, Yamazaki M, Matsubara M, Hayashi Y, Kangawa K, Sakai T: Ghrelin-producing cells exist as two types of cells, closed- and opened-type cells, in the rat gastrointestinal tract. Peptides23 :531 –536,2002k-r$-, 百拇医药

    Diamond MP, Hallarman L, Starick-Zych K, Jones TW, Connolly-Howard M, Tamborlane WV, Sherwin RS: Suppression of counterregulatory hormone response to hypoglycemia by insulin per se. J Clin Endocrinol Metab72 :1388 –1390,1991(Georg Schaller Adele Schmidt Johannes Pleiner Wolfgang Woloszczuk Michael Wolztand Anton Luger)