首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 156 毫秒
1.
目的:探讨侧脑室注射orexins(食欲素)、NPY(神经肽Y)、MCH(黑色素聚集激素)和甘丙肽对大鼠摄食的影响及其机制。方法:将成年雄性Wistar大鼠随机分为对照组、侧脑室注射组和室旁核(PVN)注射组。通过套管将orexin-A、orexin-B、NPY、MCH和甘丙肽分别注射至侧脑室和PVN内,随后测量大鼠食物摄入量,并检测PVN、弓状核(ARC)和VMH内c-fos的表达。结果:与对照组比较,侧脑室注射NPY、MCH和orexin-B 2 h后,大鼠摄食量显著增多(P0.05)。相较于orexin-B和MCH,NPY对摄食的影响更显著(P0.05)。与NS对照组比较,侧脑室注射甘丙肽和orexin-A 1 h后,大鼠摄食量显著增多(P0.05)。与NS对照组比较,侧脑室注射orexin-A可显著增加c-fos在PVN和ARC中的表达,在VMH中效应较弱(P0.05)。与NS对照组比较,PVN注射NPY能显著增加大鼠2 h摄食量(P0.05),PVN注射orexin-A能显著增加大鼠2 h和4 h摄食量(P0.05)。结论:orexins与可促进大鼠摄食,此效应可能通过下丘脑参与摄食调控中枢PVN和ARC而实现的。  相似文献   

2.
Ghrelin是生长素促分泌受体的内源性配体,刺激摄食并增加体重。已有研究证实ghrelin刺激摄食的作用靶点主要是下丘脑弓状核(hypothalamic arcuate nucleus,ARC)内的神经肽Y(neuropeptide Y,NPY)/刺鼠色蛋白相关蛋白(agouti-related peptide,AgRP)神经元。除下丘脑外,脑干尾部迷走复合体具有ghrelin受体,是ghrelin调控摄食活动的另一靶点。本实验旨在验证ghrelin作用于脑干尾部所诱发的摄食增加是否需要下丘脑NPY/AgRP神经元参与。在大鼠延髓背侧迷走复合体(dorsal vagalcomplex,DVC)微量注射20pmol的ghrelin,用摄食自动分析仪测量大鼠的摄食反应,用荧光定量PCR技术测定ARC的NPY/AgRP mRNA的表达水平,同时利用免疫组化技术测定ARC的NPY阳性神经元数量及光密度。结果显示,与对照组(DVC微量注射生理盐水)相比,ghrelin微注射组大鼠摄食量增加,其累积摄食量在注射后2h达最高峰;ARC处NPY/AgRP mRNA的表达水平、NPY免疫阳性神经元的数量及光密度也明显增加,且均在ghrelin注射后2h增高达到高峰。以上结果提示,大鼠DVC注射ghrelin可能通过上行纤维激活弓状核NPY/AgRP神经元,介导大鼠的多食反应。  相似文献   

3.
目的:探讨Ghrelin对糖尿病大鼠下丘脑弓状核胃扩张敏感神经元和胃运动的影响。方法:逆行追踪结合免疫组化观察ARC中GHSR-1的表达,细胞外放电记录,观察ghrelin对GD神经元放电活动的影响及电刺激ARC对GD神经元放电活动和胃运动的影响。结果:电生理实验结果表明,在ARC Ghrelin能够能激发GD兴奋性神经元(GD-E)和GD抑制性神经元(GD-I)。然而,ghrelin可以兴奋更少的GD-E神经元,在正常大鼠中ghrelin对于GD-E的兴奋作用比在DM大鼠中的作用弱。在体胃运动研究表明,在ARC中微量注射ghrelin可以明显的增强胃运动,并且呈现剂量依赖关系。Ghrelin在糖尿病大鼠促胃动力作用低于正常大鼠。Ghrelin诱导的效应可被生长激素促分泌素受体(GHSR)拮抗剂阻断[d-lys-3]-GHRP-6或bim28163。放射免疫法和实时荧光定量PCR数据表明胃血浆ghrelin水平,在ARC ghrelin mRNA的表达水平先上升后下降,糖尿病大鼠(DM)中,在ARC中GHSR-1a mRNA表达保持在一个比较低的水平。结论:ghrelin可以调节GD敏感神经元以及胃运动,通过ARC中ghrelin受体。在糖尿病大鼠中,Ghrelin促进胃运动作用减弱可能与ARC中ghrelin受体表达减少有关。  相似文献   

4.
目的:观察顺铂化疗对下丘脑、血浆ghrelin、orexin表达和摄食量的影响。方法:Real-time PCR、ELISA法观察顺铂对大鼠下丘脑、血浆ghrelin、orexin表达及摄食量的影响;19名接受顺铂经导管动脉灌注化疗(TAI)的肝细胞患者(HCC),ELISA法检测化疗前和化疗后血浆ghrelin、orexin的变化,用直观类比标度(VAS)(0-10)评估食欲和摄食量。结果:每日腹腔注射顺铂6 mg/kg,1-5 d大鼠摄食量均显著减少(P0.05),且1-4 d血浆酰化ghrelin显著降低(P0.05),5d时浓度仍低于对照组,但无统计学意义。血浆非酰化ghrelin和总的血浆ghrelin没有明显变化(P0.05),而1-5天血浆orexin水平均明显降低(P0.05);顺铂注射1 d后,大鼠下丘脑ghreilin和orexin的mRNA表达量均显著减少(P0.05),ghrelin mRNA变化持续3 d,orexin mRNA在化疗后5 d仍低于对照组(P0.05);肝细胞癌患者化疗后1至8 d的摄食量明显降低,1 d和2 d时的血浆酰化ghrelin显著低于化疗前水平(P0.05)。3 d时逐渐恢复,化疗后3 d、4 d和7 d时血浆酰化ghrelin浓度与化疗前无统计学差异(P0.05)。血浆非酰化ghrelin和总的血浆ghrelin没有明显变化(P0.05);化疗后1~4 d时血浆orexin浓度均显著降低(P0.05),化疗后7 d时orexin基本恢复到化疗前水平(P0.05)。结论:顺铂可降低大鼠下丘脑和血浆ghrelin、orexin的mRNA表达,HCC的TAI会降低血浆酰化ghrelin、orexin、和摄食量。  相似文献   

5.
何兵  王凯 《现代生物医学进展》2013,13(10):1983-1986
orexin是下丘脑的一种重要神经肽,在摄食、睡眠和药物成瘾等生理心理过程中起着重要作用.近年来发现下丘脑Orexin能神经纤维密集地投向参与应激调控的脑区;orexin基因敲除的老鼠表现为防御反应迟钝;另外,侧脑室微量注射orexin可导致血浆中ACTH水平的上升并诱导出应激样呼吸-心血管反应和行为反应;因此,下丘脑及中枢orexin系统对应激的调控可能起关键作用.其可能机制为强烈刺激能活化下丘脑orexin神经元,诱导中枢神经系统orexin的释放,从而激活CRF通路及蓝斑-交感-肾上腺髓质系统,提高血浆皮质酮与去甲肾上腺素的水平,诱导应激反应,维持机体的稳定.  相似文献   

6.
本研究旨在探讨外周注射ghrelin受体激动剂生长激素释放肽-6(growth hormone releasing peptide-6,GHRP-6)对NMRI小鼠摄食的影响以及摄食相关核团(弓状核、视上核和室旁核)的激活情况及其有效作用时间。腹腔注射GHRP-6 1,3,6 h后观察小鼠累计摄食量,同时用免疫组织化学方法检测GHRP-6对自由饮食小鼠和禁食小鼠下丘脑摄食相关核团c-fos表达影响,并观察GHRP-6作用的时间依从性。结果显示,腹腔注射了GHRP-6的小鼠摄食量明显大于生理盐水注射鼠,且在注射后3 h时观察到的摄食量的增加尤为显著,但注射后6 h内总的累计摄食量无显著变化;同时,GHRP-6能够在不依赖于摄食的情况下促进弓状核和室旁核中c-fos的表达,且c-fos的表达在注射后1 h时达到峰值,随后逐渐下降。以上结果提示,外源性注入GHRP-6可显著增加动物在给药后1、3 h的累积摄食量,该作用至少部分是通过上调弓状核和室旁核中的c-fos蛋白表达起作用的,而且具有时间依从性。本研究结果可为临床ghrelin受体激动剂的使用间隔提供理论依据。  相似文献   

7.
本研究旨在探讨外周注射ghrelin受体激动剂生长激素释放肽-6(growth hormone releasing peptide-6,GHRP-6)对NMRI小鼠摄食的影响以及摄食相关核团(弓状核、视上核和室旁核)的激活情况及其有效作用时间。腹腔注射GHRP-6 1,3,6 h后观察小鼠累计摄食量,同时用免疫组织化学方法检测GHRP-6对自由饮食小鼠和禁食小鼠下丘脑摄食相关核团c-fos表达影响,并观察GHRP-6作用的时间依从性。结果显示,腹腔注射了GHRP-6的小鼠摄食量明显大于生理盐水注射鼠,且在注射后3 h时观察到的摄食量的增加尤为显著,但注射后6 h内总的累计摄食量无显著变化;同时,GHRP-6能够在不依赖于摄食的情况下促进弓状核和室旁核中c-fos的表达,且c-fos的表达在注射后1 h时达到峰值,随后逐渐下降。以上结果提示,外源性注入GHRP-6可显著增加动物在给药后1、3 h的累积摄食量,该作用至少部分是通过上调弓状核和室旁核中的c-fos蛋白表达起作用的,而且具有时间依从性。本研究结果可为临床ghrelin受体激动剂的使用间隔提供理论依据。  相似文献   

8.
目的:Ghrelin是一种主要由胃粘膜产生的28氨基酸肽.因为血浆中的ghrelin完全依赖于摄食获得,所以这种激素对食欲和能量平衡有显著影响.本研究侧脑室注射ghrelin后两小时,通过检测食物容器内剩余食物及水的量,观察中枢应用ghrelin对进食的影响.方法:每天侧脑室注射一次ghrelin(0.4 μg),连续注射4天.每天测量体重,同时实验结束后检测腹膜后、附睾脂肪(WAT)含量以及血液中瘦素和胰岛素水平.结果:在第二次侧脑室注射ghrelin后,大鼠第一个和第二个30分钟时间段内进食与对照组相比显著增多(P<0.05),分别为47.2%和63.8%.自第二次注射ghrelin直至治疗结束,大鼠体重显著增加(P<0.05),食物和水的摄入量显著增多(P<0.05).实验结束后,大鼠腹膜后和附睾WAT含量显著增加(231.4%,P<0.01;84.1%,P<0.05);血清胰岛素水平显著升高(42.3%,P<0.05),而血清瘦素水平显著下降(74.1%,P<0.05).结论:中枢多次注射ghrelin对自由摄食的成年大鼠摄食具有促进作用.  相似文献   

9.
糖皮质激素对癫痫大鼠海马c-fos表达的影响   总被引:3,自引:0,他引:3  
用免疫组织化学方法观察了糖皮质激素对马桑内酯致痫大鼠海马c-fos的影响。侧脑室注射马桑内酯2h后。导致动物急性癫痫样发作。海马内出现c-fos的高度表达。单纯向侧脑室注射糖皮质激素100μg以后,动物不出现癫痫行为。海马内也几科无c-fos表达,但先向侧脑室注射糖皮质激素100μg1h,再向侧脑室内注射马桑丙酯2h后,海马内c-fos表达明显减少。除双侧肾上腺一周,再向侧脑室注射马桑内酯2h后,动物癫痫发作更明显,海马内c-fos表达较单纯且马桑内酯致痫动物更强。结果提示:糖皮质激素对马桑内酯致痫有明显的抑制作用,为临床应用糖皮质激素治疗癫痫提供了形态学参考。  相似文献   

10.
目的:探讨侧脑室注射obestatin对大鼠血浆酰基化ghrelin、去酰基化ghrelin、nesfatin-1水平的影响以及对胃排空的调控。方法:侧脑室注射obestatin,采用酶免疫测定(EIA)法检测血浆酰基化ghrelin、去酰基化ghrelin、nesfatin-1水平以及胃排空率的变化。结果:侧脑室分别注射0.1、0.3或1.0 nmol obestatin,大鼠血浆酰基化ghrelin、去酰基化ghrelin以及nesfatin-1水平无显著改变(P0.05),且酰基化ghrelin与去酰基化ghrelin比率无显著改变(P0.05);侧脑室注射obestatin,大鼠摄食量无显著改变,但胃排空率明显增加(P0.05);胃排空率明显延迟(P0.05)。与侧脑室注射1.0 nmol Obestatin组相比,注射1.0 nmol Obestatin+CRF,大鼠摄食量无显著改变,胃排空率明显延迟(P0.05)。各组摄食量及进入十二指肠内食物量无明显差异(P0.05)。结论:中枢obestatin促进大鼠的胃排空,可能与h/r CRF通路有关。  相似文献   

11.
It is known that, in rats, central and peripheral ghrelin increases food intake mainly through activation of neuropeptide Y (NPY) neurons. In contrast, intracerebroventricular (ICV) injection of ghrelin inhibits food intake in neonatal chicks. We examined the mechanism governing this inhibitory effect in chicks. The ICV injection of ghrelin or corticotropin-releasing factor (CRF), which also inhibits feeding and causes hyperactivity in chicks. Thus, we examined the interaction of ghrelin with CRF and the hypothalamo-pituitary-adrenal (HPA) axis. The ICV injection of ghrelin increased plasma corticosterone levels in a dose-dependent or a time-dependent manner. Co-injection of a CRF receptor antagonist, astressin, attenuated ghrelin-induced plasma corticosterone increase and anorexia. In addition, we also investigated the effect of ghrelin on NPY-induced food intake and on expression of hypothalamic NPY mRNA. Co-injection of ghrelin with NPY inhibited NPY-induced increase in food intake, and the ICV injection of ghrelin did not change NPY mRNA expression. These results indicate that central ghrelin does not interact with NPY as seen in rodents, but instead inhibits food intake by interacting with the endogenous CRF and its receptor.  相似文献   

12.
Intracerebroventricular (ICV) administration of ghrelin, orexin and neuropeptide Y (NPY) stimulates food intake in goldfish. Orexin and NPY interact with each other in the regulation of feeding, while ghrelin-induced feeding has also shown to be mediated by NPY in the goldfish model. To investigate the interaction between ghrelin and orexin, we examined the effects of a selective orexin receptor-1 antagonist, SB334867, and a growth hormone secretagogue-receptor antagonist, [D-Lys(3)]-GHRP-6, on ghrelin- and orexin-A-induced feeding. Ghrelin-induced food intake was completely inhibited for 1h following ICV preinjection of SB334867, while [D-Lys(3)]-GHRP-6 attenuated orexin-A stimulated feeding. Furthermore, ICV administration of ghrelin or orexin-A at a dose sufficient to stimulate food intake increased the expression of each other's mRNA in the diencephalon. These results indicate that, in goldfish, ghrelin and orexin-A have interacting orexigenic effects in the central nervous system. This is the first report that orexin-A-induced feeding is mediated by the ghrelin signaling in any animal model.  相似文献   

13.

Background

Ghrelin and obestatin are two gut-derived peptides originating from the same ghrelin/obestatin prepropeptide gene (GHRL). While ghrelin stimulates growth hormone (GH) secretion and food intake and inhibits γ-aminobutyric-acid synaptic transmission onto GHRH (Growth Hormone Releasing Hormone) neurons, obestatin blocks these effects. In Humans, GHRL gene polymorphisms have been associated with pathologies linked to an unbalanced energy homeostasis. We hypothesized that one polymorphism located in the obestatin sequence (Q to L substitution in position 90 of the ghrelin/obestatin prepropeptide, rs4684677) may impact on the function of obestatin. In the present study, we tested the activity of native and Q90L obestatin to modulate ghrelin-induced food intake, GH secretion, cFos activity in GHRH and Neuropeptide Y (NPY) neurons and γ-aminobutyric-acid activity onto GHRH neurons.

Methodology/Principal findings

Food intake, GH secretion and electrophysiological recordings were assessed in C57BL/6 mice. cFos activity was measured in NPY-Renilla-GFP and GHRH-eGFP mice. Mice received saline, ghrelin or ghrelin combined to native or Q90L obestatin (30 nmol each) in the early light phase. Ghrelin stimulation of food intake and GH secretion varied considerably among individual mice with 59–77% eliciting a robust response. In these high-responders, ghrelin-induced food intake and GH secretion were reduced equally by native and Q90L obestatin. In contrast to in vivo observations, Q90L was slightly more efficient than native obestatin in inhibiting ghrelin-induced cFos activation within the hypothalamic arcuate nucleus and the nucleus tractus solitarius of the brainstem. After ghrelin injection, 26% of NPY neurons in the arcuate nucleus expressed cFos protein and this number was significantly reduced by co-administration of Q90L obestatin. Q90L was also more potent that native obestatin in reducing ghrelin-induced inhibition of γ-aminobutyric-acid synaptic transmission onto GHRH neurons.

Conclusions/Significance

These data support the hypothesis that Q90L obestatin partially blocks ghrelin-induced food intake and GH secretion by acting through NPY and GHRH neurons.  相似文献   

14.
目的:探究下丘脑外侧区(LHA)-腹内侧核(VMH)ghrelin信号通路对肥胖大鼠的摄食选择、胃肠道运动及自发活动的影响。方法:采用免疫组织化学方法检测大鼠LHA中ghrelin受体的表达;观察LHA注射ghrelin对大鼠摄食选择胃肠道运动及自发活动的影响;电损毁VMH,观察LHA注射ghrelin对大鼠摄食的影响。结果:免疫组化结果显示,大鼠下丘脑LHA中存在ghrelin受体,且LHA-VMH之间存在纤维投射;大鼠LHA微量注射ghrelin后,肥胖(DIO)大鼠及肥胖抵抗(DR)大鼠的正常饮食、高脂饮食及高糖饮食均高于正常大鼠,但预注射ghrelin受体拮抗剂[D-Lys3]-GHRP-6 (DLS)能够阻断这种作用;而电损毁大鼠VMH,显著减弱了ghrelin对正常大鼠、DIO大鼠及DR大鼠的促摄食作用。大鼠LHA微量注射ghrelin后,正常大鼠、DIO大鼠及DR大鼠的自发活动中,X轴、Y轴方向上的活动增加,且总活动增加,但Z轴方向上活动无明显改变;此外,LHA注射ghrelin,DIO大鼠及DR大鼠的胃肠道转运速率明显加快,且DR大鼠胃肠道转运速率增加更为明显,而预注射ghrelin受体拮抗剂[D-Lys3]-GHRP-6(DLS)显著阻断ghrelin的促胃肠道转运作用。结论:下丘脑LHA-VMH ghrelin信号通路参与调节正常大鼠、DIO及DR大鼠的摄食选择、胃肠道运动及自发活动。  相似文献   

15.
Ghrelin promotes growth hormone (GH) secretion and feeding. Recent studies further showed that ghrelin displayed a defending effect against the depressive-like symptoms and affected sleep in animals and humans. Serotonergic system is considered to be implicated in feeding, depression and other mood disorders, and sleep. The dorsal raphe nucleus (DRN) utilizes serotonin (5-HT) as its major neurotransmitter and expresses GH secretagogue receptors (GHS-Rs). Therefore, the present study was carried out to examine the electrophysiological effect of ghrelin on rat DRN neurons in vitro and determine the ionic mechanism involved. Whole-cell recording revealed that ghrelin depolarized DRN neurons dose-dependently in tetrodotoxin-containing artificial cerebrospinal fluid (TTX ACSF). Pretreatment with [d-Lys3]-GHRP-6, a selective antagonist for GHS-Rs, antagonized the ghrelin-induced depolarization. The depolarization was significantly reduced in a low-Na+ TTX ACSF and in a high-K+ TTX ACSF and was abolished in the combination of both ACSFs, suggesting that the ghrelin-induced depolarization is mediated by a dual ionic mechanism including an increase in nonselective cationic conductance and a decrease in K+ conductance. The experiments on the reversal potential also supported an involvement of the dual ionic mechanism in the ghrelin-induced depolarization. On the basis of their electrophysiological and pharmacological properties, approximately 80% of DRN neurons were classified as putative 5-HT-containing neurons and ghrelin depolarized 75% of them. These results suggest that DRN neurons, especially 5-HT-containing neurons, might be involved in the neural mechanisms through which ghrelin participates in the development and/or regulation of feeding behavior, sleep-wake states and depressive-like symptoms.  相似文献   

16.
Ghrelin is an endogenous regulator of energy homeostasis synthesized by the stomach to stimulate appetite and positive energy balance. Similarly, the endocannabinoid system is part of our internal machinery controlling food intake and energy expenditure. Both peripheral and central mechanisms regulate CB1-mediated control of food intake and a functional relationship between hypothalamic ghrelin and cannabinoid CB1 receptor has been proposed. First of all, we investigated brain ghrelin actions on food intake in rats with different metabolic status (negative or equilibrate energy balance). Secondly, we tested a sub-anxiogenic ultra-low dose of the CB1 antagonist SR141716A (Rimonabant) and the peripheral-acting CB1 antagonist LH-21 on ghrelin orexigenic actions. We found that: 1) central administration of ghrelin promotes food intake in free feeding animals but not in 24 h food-deprived or chronically food-restricted animals; 2) an ultra-low dose of SR141716A (a subthreshold dose 75 folds lower than the EC50 for induction of anxiety) completely counteracts the orexigenic actions of central ghrelin in free feeding animals; 3) the peripheral-restricted CB1 antagonist LH-21 blocks ghrelin-induced hyperphagia in free feeding animals. Our study highlights the importance of the animaĺs metabolic status for the effectiveness of ghrelin in promoting feeding, and suggests that the peripheral endocannabinoid system may interact with ghrelińs signal in the control of food intake under equilibrate energy balance conditions.  相似文献   

17.
CCK and ghrelin exert antagonistic effects on ingestive behavior. The aim of the present study was to investigate the interaction between ghrelin and CCK administered peripherally on food intake and neuronal activity in specific hypothalamic and brain stem nuclei, as assessed by c-Fos-like immunoreactivity (c-FLI) in nonfasted rats. Ghrelin (13 microg/kg body wt) injected intraperitoneally significantly increased the cumulative food intake when measured at 30 min and 1 h after injection, compared with the vehicle group (2.9 +/- 1.0 g/kg body wt vs. 1.2 +/- 0.5 g/kg body wt, P < 0.028). Sulfated CCK octapeptide (CCK-8S) (2 or 25 microg/kg body wt) injected simultaneously blocked the orexigenic effect of ghrelin (0.22 +/- 0.13 g/kg body wt, P < 0.001 and 0.33 +/- 0.23 g/kg body wt, P < 0.0008), while injected alone, both doses of CCK-8S exerted a nonsignificant trend to reduce food intake. Ghrelin (13 microg/kg body wt ip) markedly increased the number of c-FLI-positive neurons per section in the arcuate nucleus (ARC) compared with vehicle (median: 31.35 vs. 9.86, P < 0.0001). CCK-8S (2 or 25 microg/kg body wt ip) had no effect on neuronal activity in the ARC, as assessed by c-FLI (median: 5.33 and 11.21 cells per section), but blocked the ghrelin-induced increase of c-fos expression in this area when both peptides were administered simultaneously (median: 13.33 and 12.86 cells per section, respectively). Ghrelin at this dose had no effect on CCK-induced stimulation of c-fos expression in the paraventricular nucleus of the hypothalamus and the nucleus of the solitary tract. These results suggest that CCK abolishes ghrelin-induced food intake through dampening increased ARC neuronal activity.  相似文献   

18.
Ghrelin is a stomach-derived hormone that regulates food intake and neuroendocrine function by acting on its receptor, GHSR (Growth Hormone Secretagogue Receptor). Recent evidence indicates that a key function of ghrelin is to signal stress to the brain. It has been suggested that one of the potential stress-related ghrelin targets is the CRF (Corticotropin-Releasing Factor)-producing neurons of the hypothalamic paraventricular nucleus, which secrete the CRF neuropeptide into the median eminence and activate the hypothalamic-pituitary-adrenal axis. However, the neural circuits that mediate the ghrelin-induced activation of this neuroendocrine axis are mostly uncharacterized. In the current study, we characterized in vivo the mechanism by which ghrelin activates the hypophysiotropic CRF neurons in mice. We found that peripheral or intra-cerebro-ventricular administration of ghrelin strongly activates c-fos--a marker of cellular activation--in CRF-producing neurons. Also, ghrelin activates CRF gene expression in the paraventricular nucleus of the hypothalamus and the hypothalamic-pituitary-adrenal axis at peripheral level. Ghrelin administration directly into the paraventricular nucleus of the hypothalamus also induces c-fos within the CRF-producing neurons and the hypothalamic-pituitary-adrenal axis, without any significant effect on the food intake. Interestingly, dual-label immunohistochemical analysis and ghrelin binding studies failed to show GHSR expression in CRF neurons. Thus, we conclude that ghrelin activates hypophysiotropic CRF neurons, albeit indirectly.  相似文献   

19.
Objective: The novel gastric hormone ghrelin has recently been identified as an important modulator of energy homeostasis. Leptin-responsive hypothalamic neuropeptide Y/Agouti-related protein neurons are believed to mediate afferent ghrelin signals. Little is known, however, about ghrelin-induced efferent signals. We therefore investigated if hypothalamic-pituitary axes have a role in transferring ghrelin-induced changes of energy balance to the periphery. Research Methods and Procedures: We subcutaneously injected hypophysectomized, as well as adrenalectomized, thyroidectomized, and sham-operated control rats with GH secretagogues [ghrelin, growth hormone (GH)-releasing peptide] for 1 week. Body weight, food intake, and body composition (chemical carcass analysis) were analyzed and compared with vehicle-treated controls. In addition, we quantified circulating levels of endogenous ghrelin in hypophysectomized and GH–treated normal rats. Results: GH-secretagogue treatment of sham-operated control rats dose-proportionally increased food intake, body weight, and fat mass compared with vehicle-injected controls (p < 0.01). These effects, however, were not observed in ghrelin-treated hypophysectomized, thyroidectomized, or adrenalectomized rats, indicating an essential role for the pituitary axis in ghrelin-induced adiposity. Circulating levels of endogenous ghrelin were reduced by administration of GH in normal rats and were about 3-fold higher in hypophysectomized rats (n = 20, p = 0.001), suggesting a regulatory feedback loop involving the stomach and the pituitary to regulate gastric ghrelin secretion. Discussion: According to these results, the endocrine pituitary is mediating ghrelin-induced changes toward a positive energy balance and is involved in the regulation of ghrelin secretion through a gastro-hypophyseal feedback loop.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号