用户名: 密码: 验证码:
靶向干扰ghrelin基因对急性胰腺炎胰腺腺泡细胞炎症与钙通路的调节机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
实践及研究表明,急性胰腺炎(acute pancreatitis, AP)的发病机制是个复杂的、多因素参与的病理生理过程,其发病机制至今尚未完全阐明。新近提出的“炎症介质或细胞因子学说”和“钙超载学说”是近年关注的热点,在这些学说中有何种机制发挥重要作用呢?本课题组的前期动物实验表明ghrelin在大鼠AP胰腺组织中表达增高,且其表达水平随着AP病情程度的加重而增加[1]。既往已有动物实验研究证实外源性给予ghrelin可以减少AP炎症介质和炎症因子的释放,也可以引起胰腺腺泡细胞内钙离子增高。由于ghrelin的广泛分布及其所具有的多种生物学功能,我们推测内源性ghrelin在AP的发生发展中可能发挥了重要作用,而其作用机理是否与此类似?本文拟对此展开研究。
     第一部分体外急性胰腺炎胰腺腺泡细胞模型构建及细胞模型内ghrelin的表达
     目的:应用雨蛙肽或脂多糖诱导大鼠AR42J胰腺腺泡细胞,以构建体外AP胰腺腺泡细胞模型;研究ghrelin在正常AR42J细胞以及在雨蛙肽诱导AR42J细胞构建的AP胰腺腺泡细胞模型内的表达情况。方法:应用不同浓度雨蛙肽(0,10-8,10-7,10-6mol/L)或脂多糖(0,1,10,100mg/L)刺激大鼠AR42J细胞24h,采用底物酶法测定细胞上清液淀粉酶含量,RT-PCR方法测定细胞内核转录因子B (nuclear factor kappa B, NF-kB)p65、磷酸化IkB-α、L型钙通道(L-type calcium channel, Cav1.2,Cav1.3)、P/Q型钙通道(P/Q-type calcium channel, Cav2.1)、N型钙通道(N-type calcium channel, Cav2.2)、T型钙通道(T-type calcium channel,Cav3.1, Cav3.2)、 ghrelin mRNA表达水平,Western Blot方法测定细胞内NF-kB p65、磷酸化IkB-α、Cav1.2、Cav1.3、ghrelin蛋白表达水平,Elisa方法测定细胞培养上清液白介素(interleukin, IL)-1β、IL-6、肿瘤坏死因子-α (tumor necrosis factor-α, TNF-α)蛋白表达水平,Fluo-4-am钙离子荧光探针联合激光共聚焦扫描显微镜测定细胞内钙离子浓度([Ca2+]i)。
     结果:1.不同浓度雨蛙肽或脂多糖干预AR42J细胞24h后,细胞培养液上清中淀粉酶含量与对照组相比无显著差异(P>0.05)。2.与对照组相比,不同浓度雨蛙肽作用24小时后,AR42J细胞NF-kB p65、IKB-α, Cav1.2、 Cav1.3、Cav2.1、Cav3.1、Cav3.2mRNA (F=9.259,15.370,19.189,14.735,42.459,23.000,16.63, P<0.05)及NF-kB p65、IkB-α、Cav1.2、Cav1.3蛋白表达水平(F=7.325,11.341,7.862,42.616,P<0.05)均明显增高;部分因子组内比较(NF-kB p65、Cav1.3、Cav2.1、Cav3.2mRNA及NF-kB p65、 IkB-α、Cav1.2、Cav1.3)呈剂量依赖性增高(P<0.05), Cav2.2mRNA表达无明显变化(F=2.121,P=0.138>0.05);不同浓度脂多糖作用24小时后的AR42J细胞上述基因mRNA表达水平均无明显变化(F=0.552,0.174,0.491,0.182,0.185,0.509,0.125,0.350,P>0.05)。3.不同浓度雨蛙肽作用24小时后,AR42J细胞培养上清液中IL-1β、IL-6蛋白表达水平较对照组均明显增高,并呈剂量依赖性(F=102.185,349.787,P<0.05);不同浓度脂多糖对上述炎症因子蛋白表达无显著影响(F=1.720,0.514;P=0.203,0.679>0.05); Elisa检测各组AR42J细胞TNF-a的OD值过低,无法计算出相应的蛋白浓度。4.不同浓度雨蛙肽作用24小时后,AR42J细胞内[Ca2+]i较对照组均显著增高,且不同浓度雨蛙肽组间[Ca2+]i比较呈剂量依赖性增高(F=421.939,P<0.05)。5.雨蛙肽处理组AR42J细胞ghrelin mRNA和蛋白表达水平较对照组明显升高(F=45.128,67.2358,P<0.05),并呈剂量依赖性(P<0.05)。
     结论:1.雨蛙肽和脂多糖刺激AR42J细胞前后,细胞培养上清液淀粉酶无明显变化;2.雨蛙肽刺激能引起AR42J细胞多种炎症因子和钙通道基因mRNA、蛋白表达水平以及[Ca2+]i升高,提示AP胰腺腺泡细胞模型构建成功;3.脂多糖刺激对AR42J细胞多种炎症介质和钙通道基因mRNA及蛋白表达无显著影响;4. ghrelin在大鼠胰腺腺泡细胞AR42J中存在高表达,雨蛙肽刺激AR42J细胞后ghrelin mRNA和蛋白表达水平较对照组升高。
     第二部分ghrelin miRNA慢病毒干扰载体的构建、筛选及对AR42J细胞的靶向干扰效果
     目的:筛选有效的大鼠ghrelin miRNA序列,构建ghrelin miRNA慢病毒载体;应用构建的慢病毒ghrelin miRNA载体转染AR42J细胞,观察其对细胞内ghrelin基因表达的抑制情况。
     方法:1.针对ghrelin基因构建四个干扰质粒:pcDNATM6.2-X207-1-3, pcDNATM6.2-X207-2-1, pcDNATM6.2-X207-3-1, pcDNATM6.2-X207-4-1;构建高表达载体pCDNA3.1(+);干扰载体和高表达载体共转染HEK293细胞,qPCR检测目的基因的表达情况,筛选出最佳干扰载体X207-1-3;使用BP重组系统和LR重组系统将目的片段从miRNA载体重组到慢病毒载体pLenti6.3/v5DEST上,筛选阳性克隆并测序验证;用构建的慢病毒表达载体pLenti6.3-X207-1和包装质粒共转染293T细胞,包装病毒,收集病毒原液,超速离心浓缩,并测定滴度。2.将AR42J细胞分为5组:空白对照组、阴性对照组(MOI=100)、ghrelin-miRNA A组(MOI=50)、ghrelin-miRNA B组(MOI=80)、 ghrelin-miRNA C组(MOI=100);应用RT-PCR和Western Blot方法分别检测各组细胞内ghrelin mRNA和蛋白的表达水平。
     结果:1.qPCR检测4个miRNA序列对ghrelin基因的沉默效应,其中以pcDNATM6.2-X207-1-3下调最为明显(86%)。2.选择沉默效应最明显(86%)的序列包装慢病毒,病毒滴度为1×108TU/ml。3. ghrelin miRNA能被重组慢病毒高效地转染入AR42J细胞,转染效率高达80%,RT-PCR和Western Blot结果显示靶基因慢病毒转染C组(MOI=100)细胞内ghrelin mRNA和蛋白表达的干扰效果最明显(P<0.05)。
     结论:成功构建慢病毒ghrelin miRNA载体,并包装出高滴度病毒,为后续实验奠定基础;构建的慢病毒ghrelin miRNA载体可以有效沉默AR42J细胞内的ghrelin基因。
     第三部分靶向干扰ghrelin基因对急性胰腺炎胰腺腺泡细胞炎症通路及钙通路的影响
     目的:研究靶向抑制ghrelin表达对雨蛙肽刺激AR42J细胞构建的AP胰腺腺泡细胞模型内炎症通路及钙通路的变化。
     方法:1.将AR42J细胞分为4组:空白对照组、雨蛙肽组、阴性对照+雨蛙肽组、ghrelin-miRNA+雨蛙肽组。2.应用RT-PCR和Western Blot方法分别检测各组细胞内NF-kB p65、磷酸化IkB-α、Cav1.2、Cav1.3. Cav2.1mRNA及蛋白的表达,Elisa方法检测各组细胞上清液IL-1β、IL-6、TNF-α蛋白的表达。3. Calcium CrimsonTM, AM钙离子荧光探针标记联合激光共聚焦扫描显微镜测定各组[Ca2+]i。
     结果:与雨蛙肽组和阴性对照+雨蛙肽组相比,ghrelin-miRNA+雨蛙肽组AR42J细胞:NF-KB p65、磷酸化IKB-αmRNA表达水平(F=51.255,9.266;P<0.05)和蛋白表达水平显著升高(F=7.533,18.876;P<0.05),细胞上清液IL-1β、IL-6蛋白表达水平显著升高(F=334.554,336.462;P<0.05);Cav1.2、Cav1.3、Cav2.1mRNA表达水平(F=37.208,55.651,42.640;P<0.05)及Cavl.2、Cav1.3蛋白表达水平明显降低(F=6.894,64.239; P<0.05);[Ca2+]i明显降低(F=61.358,P<0.05)。
     结论:靶向抑制AP胰腺腺泡细胞内ghrelin基因的表达,一方面可以上调细胞内炎症因子的表达,另一方面可以减轻其细胞内钙超载。
     第四部分全基因组基因芯片分析靶向干扰ghrelin基因对急性胰腺炎胰腺腺泡细胞基因表达谱特征的影响
     目的:筛选靶向抑制ghrelin表达的AP胰腺腺泡细胞内基因表达差异,并进一步分析相关信号通路,以探讨其分子生物学机制。
     方法:1.将AR42J细胞分为2组:ghrelin-miRNA+雨蛙肽组、阴性对照+雨蛙肽组。2.实验干预后收集细胞提取RNA,并进行RNA质量鉴定。将总RNA合成为ds-DNA后进行标记,并与NimbleGen大鼠全基因组基因芯片杂交,运用Axon GenePix4000B基因芯片扫描仪进行扫描。3.所有基因水平文件输入Agilent GeneSpring GX11.5.1软件进行聚类分级,进一步进行GO分析和通路分析。
     结果:1. NimbleGen全基因组基因芯片筛选出ghrelin基因沉默及雨蛙肽诱导的AR42J细胞差异表达基因上调或下调2倍以上基因共2938个,其中表达上调基因1435个,表达下调基因1503个。2.GO分析结果显示差异表达基因在biological process分类中,与各种代谢过程相关的基因出现最多,其次为生物调节、进化过程、细胞过程调节等;在cellular component分类中,改变最为显著的差异表达基因主要位于细胞及细胞内部分、细胞核、膜型细胞器等细胞成分中;在molecular function分类中,与结合相关的基因差异最多,包括蛋白结合、金属离子结合、阳离子结合、核苷酸结合、小分子结合等。3. Pathway分析结果显示表达上调基因涉及31个信号通路,表达下调基因涉及30个信号通路。4.通过筛查与炎症和钙通路相关的差异表达基因,结果显示,与CAMP/Ca2+信号通路相关的差异表达基因有60个,与炎症通路相关的差异表达基因有199个。
     结论:通过芯片结果、Pathway及GO分析等初步筛查发现:ghrelin对AP胰腺腺泡细胞的作用机制可能与细胞内代谢过程及离子、蛋白等结合功能有关;相关通路可能包括MAPK信号通路、细胞间粘附分子信号通路、焦点连接、Wnt信号通路、P53信号通路等。
Practice and studies have shown that the pathogenesis of acute pancreatitis (AP) was a pathophysiological process involved in complex and multi-factor. And its pathogenesis has not been fully elucidated. The theories of "Inflammatory mediators or cytokines" and "calcium overload" are the focus of attention in the recent years. What mechanism will play an important role in these theories? Our previous animal experiments showed that expression of ghrelin was increased in the rats'pancreatic tissue of AP, and its expression levels were increased with the severity of AP. Previous studies have verified that exogenous of ghrelin could reduce the release of inflammatory mediators and inflammatory factors in AP, furthermore, it could also increased calcium in pancreatic acinar cells. Because ghrelin is widely distributed and has a variety of biological functions, we hypothesize that endogenous ghrelin may play an important role in the development of AP, and whether its mechanism is similar to exogenous ghrelin? This article intends to study it.
     Part One
     Pancreatic Acinar Cell Model Building of Acute Pancreatitis in Vitro and Expression of Ghrelin in This Cell Model
     Objective:Rats'pancreatic acinar cells AR42J were induced by caerulin (CAE) or lipopolysaccharide (LPS) in order to build the pancreatic acinar cell model of AP in vitro. Study the expression of ghrelin in AR42J cells as well as builted in AR42J cells induced by caerulin.
     Methods:AR42J cells were induced by different concentration of CAE (0,10-8,10-7,10-6mol/L)or LPS(0,1,10,100mg/L)for24hrs.Amylase(AMS) content in cell supernatant was measured by bottom objects enzymatic. Expression of NF-kB p65, phosphorylation IkB-α, Cavl.2, Cav1.3, Cav2.1, Cav2.2Cav3.1Cav3.2and ghrelin mRNA was determined by RT-PCR. Proteins expression of NF-kB p65, phosphorylation IkB-α, Cav1.2, Cav1.3and ghrelin were determined by Western Blot. Proteins expression of IL-1β, IL-6, TNF-α in cell culture supernatant were determined by Elisa.[Ca2+]i was determined by calcium fluorescence probe Fluo-4-am loading combined with confocal laser scanning microscope (CLSM).
     Results:1.There was no statistically significant difference with AMS between control group cells and cells induced by CAE or LPS(P>0.05).2. Compared to control group, expression of NF-kB p65, IkB-α, Cav1.2, Cav1.3, Cav2.1,Cav3.1,Cav3.2mRNA(F=9.259,15.370,19.189,14.735,42.459,23.000,16.63, P<0.05) and NF-kB p65, IkB-α, Cav1.2, Cav1.3proteins (F=7.325,11.341,7.862,42.616, P<0.05) were increased significantly. Comparison in each group, some factors were dose-dependently increased (NF-kB p65, Cav1.3, Cav2.1, Cav3.2mRNA and NF-kB p65, IkB-α, Cavl.2, Cavl.3proteins, P<0.05). There was no significant difference (F=2.121, P=0.138>0.05) in Cav2.2mRNA. There was no significant difference in mRNA expression of aboved genes between control group cells and cells induced by LPS(F=0.552,0.174,0.491,0.182,0.185,0.509,0.125,0.350, P>0.05).3. Proteins expression of IL-1β and IL-6were increased significantly in AR42J cells induced by CAE, which was a dose-dependent manner (F=102.185,349.787, P<0.05). However, there was no significant difference in proteins expression of IL-1β and IL-6in AR42J cells induced by LPS (F=1.720,0.514, P=0.203,0.679>0.05). Because of OD value of TNF-a was too low, the corresponding protein concentration can not be calculated.4. Compared to control group,[Ca2+]i was increased significantly in AR42J cells induced by CAE, which was a dose-dependent manner (F=21.939, P<0.05).5. Expressions of ghrelin mRNA and protein were increased in AR42J cells induced by CAE, which was a dose-dependent manner (F=45.128,67.2358, P<0.05).
     Conclusions:1. There is no significant change of AMS in cell culture supernatant between normal AR42J cells and AR42J cells induced by CAE or LPS.2. CAE can enhance mRNA and protein levels of a variety of inflammatory cytokines and calcium channel, as well as [Ca2+]i in AR42J cells. It may indicate pancreatic acinar cell model of acute pancreatitis to be built successfully.3. LPS can not affect mRNA and protein levels of a variety of inflammatory cytokines and calcium channel in AR42J cells.4. High expression of ghrelin in AR42J cells. Compared with control group, ghrelin mRNA and protein expression levels are elevated in AR42J cells induced by CAE.
     Part Two
     Building and Screen of Lentiviral Interference Vector of Ghrelin miRNA and Its Targeted Interference Effect in AR42J Cells
     Objective:To screen the effective sequence of ghrelin miRNA of rat in order to build lentiviral vector of ghrelin miRNA. After AR42J cells were transfected with the lentiviral vector of ghrelin miRNA, we observed interference effect of ghrelin gene in cells.
     Methods:l.Four interference plasmids were constructed: pcDNATM6.2-X207-1-3,pcDNATM6.2-X207-2-1,pcDNATM6.2-X207-3-1,pcDN ATM6.2-X207-4-1. Expression vector pCDNA3.1(+) was constructed; After HEK293cells were co-transfected with the interference vector and expression vector, expression of target gene was determined by qPCR so as to screen out the best interference vector X207-1-3. Target fragment was restructured from miRNA vector to the lentiviral vector pLenti6.3/v5DEST through BP recombination system and LR recombination system, and positive clones were screened out and sequenced. After293T cells were cotransfected with lentiviral vector pLenti6.3-X207-1and packaging plasmid, packing virus, collecting virus stoste, ultracentrifugation to concentrate, and determining the titer.2. AR42J cells were divided into5groups:blank control group, negative control group (MOI=100), ghrelin-miRNA group A (MOI=50), ghrelin-miRNA group B (MOI=80), ghrelin-miRNA C group (MOI=100). Expression of ghrelin mRNA and protein in each group was detected by RT-PCR and Western Blot.
     Results:1.The four miRNA sequences to ghrelin gene silencing effection were detected by qPCR and the result showed that pcDNA TM6.2-X207-1-3was the most significant silencing effected sequence (86%).2. To pack lentivirus with the most obvious silencing effected sequence (86%) and the viral titer was1×108TU/ml.3. Ghrelin miRNA could be transfected efficiently into AR42J cells. And the transfection efficiency was up to80%. The results of RT-PCR and Western Blot showed that the most significant interference effect of ghrelin mRNA and protein expression in AR42J cells transfected by ghrelin-miRNA C group (MOI=100)(P<0.05).
     Conclusions:We successfully built lentiviral vector of ghrelin-miRNA and packed high titers of virus, so as to lay the foundation for the subsequent experiments. The lentiviral vector of ghrelin-miRNA can effectively silence ghrelin gene in AR42J cells.
     Part Three
     Effects of Ghrelin Gene Silence on Inflammation Pathway and Calcium Channel in Pancreatic Acinar Cells of Acute Pancreatitis
     Objective:Study the changes of the inflammation pathway and calcium channel by ghrelin gene targeting suppression in pancreatic acinar cell model of acute pancreatitis induced by caerulin.
     Method:1. AR42J cells were divided into four groups:blank control group, CAE group, negative control (NG)+CAE group and miRNA+CAE group.2. Expression of NF-κB p65, phosphorylation IκB-α, Cav1.2, Cav1.3, Cav2.1mRNA and proteins was determined by RT-PCR and Western Blot. Proteins expression of IL-1β, IL-6and TNF-αwere determined by Elisa.3.[Ca2+]i in AR42J cells of each group was determined by Calcium Crimson TM, AM calcium fluorescent probe loading combined with confocal laser scanning microscope.
     Results:Compared with CAE group and NC+CAE group, the following results of miRNA+CAE group showed that:expression of NF-KBp65and IkB-α mRNA (F=51.255,9.266, P<0.05) and proteins (F=7.533,18.876, P<0.05) was elevated; proteins expression of IL-1β, IL-6in cell culture supernatant were elevated (F=334.554,336.462, P<0.05); expression of Cav1.2, Cav1.3, Cav2.1mRNA (F=37.208,55.651,42.640, P<0.05) and Cav1.2, Cavl.3proteins (F=6.894,64.239, P<0.05) were significantly decreased;[Ca2+]i in cells was significantly decreased (F=61.358, P<0.05).
     Conclusions:Targeting suppression of ghrelin gene in pancreatic acinar cells of acute pancreatitis can up-regulate the expression of inflammatory cytokines, as well as can reduce calcium overload in cells.
     Part Four
     Analysing the expression profiles characteristic of ghrelin gene silencing gene in pancreatic acinar cells of AP with whole genome microarray
     Objective:Screen out differentially expressed genes of targeting suppression of ghrelin gene in pancreatic acinar cells of acute pancreatitis. And further analysis of the relevant signal pathway was employed to explore the mechanism of molecular biology.
     Method:1. AR42J cells were divided into two groups:miRNA+CAE group and NC+CAE group.2. AR42J cells were collected to extract RNA after intervention of lentivirus and CAE. RNA quality was assessed. Synthesize total RNA into ds-DNA and mark it which was hybridized with NimbleGen rat whole genome microarray. Then, we scanned the microarray by Axon GenePix4000B gene chip scanner.3. All gene-level files were input Agilent GeneSpring GX11.5.1software to clustering classify, and further GO analysis and pathway analysis were worked.
     Results:1. There are2938genes of differentially expression including more than tow times of up-regulation (1435) and down-regulation (1503).2. The results of GO analysis showed that the differentially expressed genes which were associated with various metabolic processes appeared most frequently in the kind of biological process, followed by biological regulation, evolutionary process, regulation of cell process. In the kind of cellular component, the differentially expressed genes of the most significant change mainly located in the cells, nucleus, membrane type organelles and other cellular components. In the kind of molecular function, the most amount of differentially expression genes was associated with combination, including protein binding, metal ion binding, cation binding, nucleotide binding and small molecules binding.3. The results of pathway analysis showed that31signaling pathways involved in up-regulated genes and30signaling pathways involved in down-regulated genes.4. There are60differentially expressed genes involved in CAMP/Ca2+signal pathway and199differentially expressed genes involved in inflammation pathway.
     Conclusions:Through the results of gene chip, pathway and GO analysis, we preliminarily speculate that the mechanism of ghrelin on pancreatic acinar cells of acute pancreatitis may be related with metabolic processes in cells and binding of ion, protein, etc. And the related pathways of this mechanism may include MAPK signal pathway, cell adhesion molecules signal pathway, focal adhesion, Wnt signal pathway and p53signal pathway, etc.
引文
[l]方春云,唐国都,梁志海等.Ghrelin及GHSR在急性坏死性胰腺炎大鼠胰腺组织的表达.中华胰腺病杂志,2012;12(6):384-7.
    [2]Yousaf M, McCallion K, Diamond T. Management of severe acute pancreatitis. Br J Surg. 2003;90(4):407-20.
    [3]Toouli J, Brooke-Smith M, Bassi C, etc. Guidelines for the management of acute pancreatitis. J Gastroenterol Hepatol.2002;17 Suppl:S15-39.
    [4]Sileikis A, Beisa V, Simutis G, etc. Three-port retroperitoneoscopic necrosectomy in management of acute necrotic pancreatitis. Medicina (Kaunas).2010;46(3):176-9.
    [5]Andrius Karpavicius, Zilvinas Dambrauskas, Audrius Sileikis, etc. Value of adipokines in predicting the severity of acute pancreatitis:Comprehensive review. World J Gastroenterol. 20127; 18(45):6620-7.
    [6]Harrison DA, D'Amico G, Singer M, etc. The Pancreatitis Outcome Prediction (POP) Score:a new prognostic index for patients with severe acute pancreatitis. Crit Care Med. 2007;35(7):1703-8.
    [7]张红,李永渝.急性胰腺炎的发病机制研究进展.中国危重病急救医学,2000;12(2):121-4.
    [8]薛平,邓力珲,张肇达等.柴芩承气汤减轻急性胰腺炎大鼠胰腺钙超载的机制研究.中西医结合学报,2008;6(10):1054-8.
    [9]Li YY, Lu XY, Li XJ, etc. Intervention of pyrrolidine dithiocarbamate and tetrandrine on cellular calcium overload of pancreatic acinar cells induced by serum and ascitic fluid from rats with acute pancreatitis. J Gastroenterol Hepatol.2009;24(1):155-65.
    [10]Bruce JI, Elliott AC. Oxidant-impaired intracellular Ca2+signaling in pancreatic acinar cells:role of the plasma membrane Ca2+ -ATPase. Am J Physiol Cell Physiol. 2007;293(3):C938-50.
    [11]Gukovskaya AS, Hosseini S, Satoh A, etc. Ethanol differentially regulates NF-kappaB activation in pancreatic acinar cells through calcium and protein kinase C pathways. Am J Physiol Gastrointest Liver Physiol.2004;286(2):G204-13.
    [12]Niederau C. Do cholecystokinin antagonists increase cytosolic calcium in pancreatic acinar cells and thereby promote pancreatitis? Dig Dis Sci.2004;49(2):266-9.
    [13]Kojima M, Hosoda H, Date Y, etc. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature.1999;402(6762):656-60.
    [14]Peino R, Baldelli R, Rodriguez-Garcia J, etc. Ghrelin-induced growth hormone secretion in humans. Eur J Endocrinol.2000;143(6):R11-4.
    [15]Mori K, Yoshimoto A, Takaya K, etc. Kidney produces a novel acylated peptide, ghrelin. FEBS Lett.2000;486(3):213-6.
    [16]Korbonits M, Bustin SA, Kojima M, etc. The expression of the growth hormone secretagogue receptor ligand ghrelin in normal and abnormal human pituitary and other neuroendocrine tumors. J Clin Endocrinol Metab.2001;86(2):881-7.
    [17]Hosoda H, Kojima M, Matsuo H, etc. Ghrelin and des-acyl ghrelin:two major forms of rat ghrelin peptide in gastrointestinal tissue. Biochem Biophys Res Commun. 2000;279(3):909-13.
    [18]Tolle V, Zizzari P, Tomasetto C, etc. In vivo and in vitro effects of ghrelin/motilin-related peptide on growth hormone secretion in the rat. Neuroendocrinology.2001;73(1):54-61.
    [19]Soares JB, Leite-Moreira AF. Ghrelin, des-acyl ghrelin and obestatin:three pieces of the same puzzle. Peptides,2008; 29(7):1255-70.
    [20]Warzecha Z, Ceranowicz P, Dembinski A, etc. Therapeutic effect of ghrelin in the course of cerulein-induced acute pancreatitis in rats. J Physiol Pharmacol.2010;61(4):419-27.
    [21]Zhou X, Xue C. Ghrelin inhibits the development of acute pancreatitis and nuclear factor kappaB activation in pancreas and liver. Pancreas.2009;38(7):752-7.
    [22]Kerem M, Bedirli A, Pasaoglu H, etc. Role of ghrelin and leptin in predicting the severity of acute pancreatitis. Dig Dis Sci.2007;52(4):950-5.
    [23]Lai JK, Cheng CH, Ko WH, etc. Ghrelin system in pancreatic AR42J cells:its ligand stimulation evokes calcium signalling through ghrelin receptors. Int J Biochem Cell Biol. 2005;37(4):887-900.
    [24]Lampel M, Kern HF. Acute interstitial pancreatitis in the rat induced by excessive doses of a pancreatic secretagogue. Virchows Arch A Pathol Anat Histol.1977;373(2):97-117.
    [25]Niederau C, Ferrell LD, Grendell JH. Caerulein-induced acute necrotizing pancreatitis in mice:protective effects of proglumide, benzotript, and secretin. Gastroenterology.1985;88(5 Pt 1):1192-204.
    [26]Tsang SW, Cheng CH, Leung PS. The role of the pancreatic renin-angiotensin system in acinar digestive enzyme secretion and in acute pancreatitis. Regul Pept.2004;119(3):213-9.
    [27]Bockman DE, Guo J, Miiller MW, etc. Cell wounding in early experimental acute pancreatitis. Lab Invest.2004;84(3):362-7.
    [28]Frossard JL, Pastor CM, Hadengue A. Effect of hyperthermia on NF-kappaB binding activity in cerulein-induced acute pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2001;280(6):G1157-62.
    [29]Hietaranta AJ, Singh VP, Bhagat L, etc. Water immersion stress prevents caerulein-induced pancreatic acinar cell nf-kappa b activation by attenuating caerulein-induced intracellular Ca2+ changes. J Biol Chem.2001;276(22):18742-7.
    [30]王兴鹏,王冰娴,徐选福等.肠源性内毒素血症促使水肿型胰腺炎向坏死型胰腺炎转化.中华消化杂志,2001;21(2):71-74.
    [31]Lee J, Seo JH, Lim JW, etc. Membrane proteome analysis of cerulein-stimulated pancreatic acinar cells:implication for early event of acute pancreatitis. Gut Liver. 2010;4(1):84-93.
    [32]Yu JH, Lim JW, Kim H. Altered gene expression in cerulein-stimulated pancreatic acinar cells:pathologic mechanism of acute pancreatitis. Korean J Physiol Pharmacol. 2009;13(6):409-16.
    [33]Chen P, Huang L, Zhang Y, etc. The antagonist of the JAK-1/STAT-1 signaling pathway improves the severity of cerulein-stimulated pancreatic injury via inhibition of NF-kB activity. Int J Mol Med.2011;27(5):731-8.
    [34]龙友明,刘学,进陈垦等.脂多糖对AR42J细胞核因子-кB、肿瘤坏死因子-α表达的影响.中华急诊医学杂志,2008;17(1):54-8.
    [35]Kim D, Kolch W, Cho KH. Multiple roles of the NF-kappaB signaling pathway regulated by coupled negative feedback circuits. FASEB J.2009;23(9):2796-802.
    [36]Baker RG, Hayden MS, Ghosh S. NF-kB, inflammation, and metabolic disease. Cell Metab.2011;13(1):11-22.
    [37]Goebeler M, Gillitzer R, Kilian K, etc. Multiple signaling pathways regulate NF-kappaB-dependent transcription of the monocyte chemoattractant protein-1 gene in primary endothelial cells. Blood.2001;97(1):46-55.
    [38]Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol.2009; 1(6):a001651.
    [39]袁洪峰,贺翔鸽.核因子kB与眼科疾病的研究进展.国际眼科杂志,2008;8(1):132-136.
    [40]张红,李永渝.维拉帕米减轻脂多糖导致的大鼠胰腺腺泡细胞损伤机制的研究.中国病理生理杂志,2007;23(3):444-8.
    [41]Abe R, Shimosegawa T, Kimura K, etc. Lipopolysaccharide-induced desensitization to pancreatic edema formation in rat cerulein pancreatitis. Pancreas.1998;16(4):539-44.
    [42]Kimura K, Shimosegawa T, Abe R, etc. Low doses of lipopolysaccharide upregulate acinar cell apoptosis in cerulein pancreatitis. Pancreas.1998; 17(2):120-6.
    [43]Oruc N, Ozutemiz AO, Yukselen V, etc. Infliximab:a new therapeutic agent in acute pancreatitis? Pancreas.2004;28(1):e1-8.
    [44]Xiping Z, Dijiong W, Jianfeng L, etc. Effects of Salvia miltiorrhizae on ICAM-1, TLR4, NF-kappaB and Bax proteins expression in multiple organs of rats with severe acute pancreatitis or obstructive jaundice. Inflammation.2009;32(4):218-32.
    [45]Lu XS, Qiu F, Li JQ, etc. Low molecular weight heparin in the treatment of severe acute pancreatitis:a multiple centre prospective clinical study. Asian J Surg.2009;32(2):89-94.
    [46]Nikolopoulos D, Theocharis S, Kouraklis G. Ghrelin's role on gastrointestinal tract cancer. Surg Oncol.2010;19(1):e2-10.
    [47]Root AW, Root MJ. Clinical pharmacology of human growth hormone and its secretagogues. Curr Drug Targets Immune Endocr Metabol Disord.2002;2(1):27-52.
    [48]Ortiz RM, Noren DP, Ortiz CL, etc. GH and ghrelin increase with fasting in a naturally adapted species, the northern elephant seal (Mirounga angustirostris). J Endocrinol. 2003;178(3):533-9.
    [49]Gnanapavan S, Kola B, Bustin SA, etc. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J Clin Endocrinol Metab.2002;87(6):2988.
    [50]Date Y, Nakazato M, Hashiguchi S, etc. Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes.2002;51 (1):124-9.
    [51]Volante M, Allia E, Gugliotta P, etc. Expression of ghrelin and of the GH secretagogue receptor by pancreatic islet cells and related endocrine tumors. J Clin Endocrinol Metab. 2002;87(3):1300-8.
    [52]Wierup N, Svensson H, Mulder H, etc. The ghrelin cell:a novel developmentally regulated islet cell in the human pancreas. Regul Pept.2002;107(1-3):63-9.
    [53]Wierup N, Yang S, McEvilly RJ, etc. 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.2004;52(3):301-10.
    [54]Prado CL, Pugh-Bernard AE, Elghazi L, etc. Ghrelin cells replace insulin-producing beta cells in two mouse models of pancreas development. Proc Natl Acad Sci U S A. 2004;101(9):2924-9.
    [55]Hannon GJ, Rossi JJ. Unlocking the potential of the human genome with RNA interference. Nature.2004;431(7006):371-8.
    [56]Brennecke J, Hipfner DR, Stark A, etc. bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell.2003;113(1):25-36.
    [57]李继霞,周克元.mjRNA的研究进展.生物化学与生物物理进展,2003;30(5):702-4.
    [58]Sumimoto H, Kawakami Y. Lentiviral vector-mediated RNAi and its use for cancer research. Future Oncol.2007;3(6):655-64.
    [59]Frecha C, Szecsi J, Cosset FL, etc. Strategies for targeting lentiviral vectors. Curr Gene Ther.2008;8(6):449-60.
    [60]Schambach A, Baum C. Clinical application of lentiviral vectors-concepts and practice Curr Gene Ther.2008;8(6):474-82.
    [61]Singer O, Verma IM. Applications of lentiviral vectors for shRNA delivery and transgenesis. Curr Gene Ther.2008;8(6):483-8.
    [62]Fang H, Hong Z, Zhang J, Effects of ghrelin on the intracellular calcium concentration in rat aorta vascular smooth muscle cells. Cell Physiol Biochem.2012;30(5):1299-309.
    [63]Yamazaki M, Aizawa S, Tanaka T, etc. Ghrelin increases intracellular Ca2□ concentration in the various hormone-producing cell types of the rat pituitary gland. Neurosci Lett. 2012;526(1):29-32.
    [64]Hattori N. Expression, regulation and biological actions of growth hormone (GH) and ghrelin in the immune system. Growth Horm IGF Res.2009; 19(3):187-97.
    [65]Kwan RO, Cureton E, Dozier K, etc. Ghrelin decreases microvascular leak during inflammation. J Trauma.2010;68(5):1186-91
    [66]Dembinski A, Warzecha Z, Ceranowicz P, etc. Ghrelin attenuates the development of acute pancreatitis in rat. J Physiol Pharmacol.2003;54(4):561-73.
    [67]Delgado M, Ganea D. Anti-inflammatory neuropeptides:a new class of endogenous immunoregulatory agents. Brain Behav Immun.2008;22(8):1146-51.
    [68]Liu SP, Li XY, Li Z, etc. Octanoylated Ghrelin Inhibits the Activation of the Palmitic Acid-Induced TLR4/NF-κB Signaling Pathway in THP-1 Macrophages. ISRN Endocrinol. 2012;2012:237613.
    [69]Dixit VD, Schaffer EM, Pyle RS, etc. Ghrelin inhibits leptin-and activation-induced proinflammatory cytokine expression by human monocytes and T cells. J Clin Invest. 2004;114(1):57-66.
    [70]Vila G, Maier C, Riedl M, etc. Bacterial endotoxin induces biphasic changes in plasma ghrelin in healthy humans. J Clin Endocrinol Metab.2007;92(10):3930-4.
    [71]Daniel P, Lesniowski B, Jasinska A, etc. Usefulness of assessing circulating levels of resistin, ghrelin, and IL-18 in alcoholic acute pancreatitis. Dig Dis Sci.2010;55(10):2982-7.
    [72]Kohno D, Nakata M, Maekawa F, etc. Leptin suppresses ghrel in-induced activation of neuropeptide Y neurons in the arcuate nucleus via phosphatidylinositol 3-kinase-and phosphodiesterase 3-mediated pathway. Endocrinology.2007;148(5):2251-63.
    [73]Kohno D, Sone H, Minokoshi Y, etc. Ghrelin raises [Ca2+]i via AMPK in hypothalamic arcuate nucleus NPY neurons. Biochem Biophys Res Commun.2008;366(2):388-92.
    [74]Liu Y, Zhou YB, Zhang GG, etc. Cortistatin attenuates vascular calcification in rats. Regul Pept.2010;159(1-3):35-43.
    [75]Grey CL, Chang JP. Ghrelin-induced growth hormone release from goldfish pituitary cells involves voltage-sensitive calcium channels. Gen Comp Endocrinol. 2009; 160(2):148-57.
    [76]许燕萍,梁黎,王秀敏等.不同出生体重新生大鼠胰腺ghrelin和钙通道表达的研究.浙江大学学报(医学版),2008;37(3):233-9.
    [77]Ju KD, Yu JH, Kim H,etc. Role of mitogen-activated protein kinases, NF-kappaB, and AP-1 on cerulein-induced IL-8 expression in pancreatic acinar cells. Ann N Y Acad Sci. 2006;1090:368-74.
    [78]Mazzon E, Impellizzeri D, Di Paola R, etc. Effects of mitogen-activated protein kinase signaling pathway inhibition on the development of cerulein-induced acute pancreatitis in mice. Pancreas.2012;41(4):560-70.
    [79]Feng Z, Fei J, Wenjian X, Jiachen J, etc. Rhubarb attenuates the severity of acute necrotizing pancreatitis by inhibiting MAPKs in rats. Immunotherapy.2012;4(12):1817-21.
    [80]Lee JK, Ryu JK, Park JK, etc. Effects of nafamostat mesilate on the prevention of cerulein-induced acute pancreatitis. Pancreas.2008;36(3):255-60.
    [81]Perejaslov A, Chooklin S, Bihalskyy I. Implication of interleukin 18 and intercellular adhesion molecule (ICAM)-1 in acute pancreatitis. Hepatogastroenterology. 2008;55(86-87):1806-13.
    [82]Kleinhans H, Kaifi JT, Mann O, etc. The role of vascular adhesion molecules PECAM-1 (CD 31), VCAM-1 (CD 106), E-selectin (CD62E) and P-selectin (CD62P) in severe porcine pancreatitis. Histol Histopathol.2009;24(5):551-7.
    [83]Ramudo L, Yubero S, Manso MA, etc. Effects of dexamethasone on intercellular adhesion molecule 1 expression and inflammatory response in necrotizing acute pancreatitis in rats. Pancreas.2010;39(7):1057-63.
    [84]Zhu HH, Jiang LL. Serum inter-cellular adhesion molecule 1 is an early marker of diagnosis and prediction of severe acute pancreatitis. World J Gastroenterol. 2012;18(20):2554-60.
    [85]Nakamura Y, Do JH, Yuan J, etc. Inflammatory cells regulate p53 and caspases in acute pancreatitis. Am J Physiol Gastrointest Liver Physiol.2010;298(1):G92-100.
    [86]Rizvi IA, Robinson K, McFadden DW, etc. Peptide YY reverses TNF-alpha induced transcription factor binding of interferon regulatory factor-1 and p53 in pancreatic acinar cells. J Surg Res.2006;136(1):25-30.
    [87]Ju KD, Lim JW, Kim KH, etc. Potential role of NADPH oxidase-mediated activation of Jak2/Stat3 and mitogen-activated protein kinases and expression of TGF-β1 in the pathophysiology of acute pancreatitis. Inflamm Res.2011;60(8):791-800.
    [88]Weng TI, Wu HY, Chen BL, etc. Honokiol attenuates the severity of acute pancreatitis and associated lung injury via acceleration of acinar cell apoptosis. Shock. 2012;37(5):478-84.
    [89]Liu Y, Zhou ZG, Chen KL, etc. The ER chaperone GRP78 is associated with the severity of cerulein-induced pancreatic inflammation via regulating apoptosis of pancreatic acinar cells. Hepatogastroenterology.2012;59(118):1670-6.
    [90]Wang G, Han B, Zhou H, etc. Inhibition of hydrogen sulfide synthesis provides protection for severe acute pancreatitis rats via apoptosis pathway. Apoptosis. 2013;18(1):28-42.
    [1]Kojima M, Hosoda H, Date Y,et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature.1999;402(6762):656-60.
    [2]Hosoda H, Kojima M, Matsuo H,et al. Ghrelin and des-acyl ghrelin:two major forms of rat ghrelin peptide in gastrointestinal tissue. Biochem Biophys Res Commun. 2000;279(3):909-13.
    [3]Tolle V, Zizzari P, Tomasetto C, et al. In vivo and in vitro effects of ghrelin/motilin-related peptide on growth hormone secretion in the rat. Neuroendocrinology.2001;73(1):54-61.
    [4]Peino R, Baldelli R, Rodriguez-Garcia J, et al. Ghrelin-induced growth hormone secretion in humans. Eur J Endocrinol.2000;143(6):R11-4.
    [5]Mori K, Yoshimoto A, Takaya K, et al. Kidney produces a novel acylated peptide, ghrelin. FEBS Lett.2000;486(3):213-6.
    [6]Korbonits M, Bustin SA, Kojima M, et al. The expression of the growth hormone secretagogue receptor ligand ghrelin in normal and abnormal human pituitary and other neuroendocrine tumors. J Clin Endocrinol Metab.2001;86(2):881-7.
    [7]Dezaki K, Sone H, Yada T. Ghrelin is a physiological regulator of insulin release in pancreatic islets and glucose homeostasis. Pharmacol Ther.2008; 118(2):239-49.
    [8]Dezaki K, Hosoda H, Kakei M, et al. Endogenous ghrelin in pancreatic islets restricts insulin release by attenuating Ca2+ signaling in beta-cells:implication in the glycemic control in rodents. Diabetes.2004;53(12):3142-51.
    [9]Qader SS, Hakanson R, Rehfeld JF, et al. Proghrelin-derived peptides influence the secretion of insulin, glucagon, pancreatic polypeptide and somatostatin:a study on isolated islets from mouse and rat pancreas. Regul Pept.2008;146(1-3):230-7.
    [10]Poykko SM, Kellokoski E, Horkko S, et al. Low plasma ghrelin is associated with insulin resistance, hypertension, and the prevalence of type 2 diabetes. Diabetes. 2003;52(10):2546-53.
    [11]Poykko S, Ukkola O, Kauma H, et al. Ghrelin Arg51Gln mutation is a risk factor for Type 2 diabetes and hypertension in a random sample of middle-aged subjects. Diabetologia.2003;46(4):455-8.
    [12]Katsuki A, Urakawa H, Gabazza EC, et al. Circulating levels of active ghrelin is associated with abdominal adiposity, hyperinsulinemia and insulin resistance in patients with type 2 diabetes mellitus. Eur J Endocrinol.2004;151(5):573-7.
    [13]Asakawa A, Inui A, Kaga T, et al. Antagonism of ghrelin receptor reduces food intake and body weight gain in mice. Gut.2003;52(7):947-52.
    [14]Volante M, Allia E, Gugliotta P, et al. Expression of ghrelin and of the GH secretagogue receptor by pancreatic islet cells and related endocrine tumors. J Clin Endocrinol Metab. 2002;87(3):1300-8.
    [15]Ekeblad S, Lejonklou MH, Grimfjard P, et al. Co-expression of ghrelin and its receptor in pancreatic endocrine tumours. Clin Endocrinol (Oxf).2007;66(1):115-22.
    [16]Walter T, Chardon L, Hervieu V, et al. Major hyperghrelinemia in advanced well-differentiated neuroendocrine carcinomas:report of three cases. Eur J Endocrinol. 2009;161(4):639-45.
    [17]Corbetta S, Peracchi M, Cappiello V, et al. Circulating ghrelin levels in patients with pancreatic and gastrointestinal neuroendocrine tumors:identification of one pancreatic ghrelinoma. J Clin Endocrinol Metab.2003;88(7):3117-20.
    [18]Walter T, Chardon L, Hervieu V, et al. Major hyperghrelinemia in advanced well-differentiated neuroendocrine carcinomas:report of three cases. Eur J Endocrinol. 2009;161(4):639-45.
    [19]Kerem M, Bedirli A, Pasaoglu H, et al. Role of ghrelin and leptin in predicting the severity of acute pancreatitis. Dig Dis Sci.2007;52(4):950-5.
    [20]Warzecha Z, Ceranowicz P, Dembinski A, et al. Therapeutic effect of ghrelin in the course of cerulein-induced acute pancreatitis in rats. J Physiol Pharmacol. 2010;61(4):419-27.
    [21]Zhou X, Xue C. Ghrelin inhibits the development of acute pancreatitis and nuclear factor kappaB activation in pancreas and liver. Pancreas.2009;38(7):752-7.
    [22]Zhou X, Xue C. Ghrelin attenuates acute pancreatitis-induced lung injury and inhibits substance P expression. Am J Med Sci.2010;339(1):49-54.
    [23]Lai KC, Cheng CH, Leung PS. The ghrelin system in acinar cells:localization, expression, and regulation in the exocrine pancreas. Pancreas.2007;35(3):e1-8.
    [24]Lai JK, Cheng CH, Ko WH, et al. Ghrelin system in pancreatic AR42J cells:its ligand stimulation evokes calcium signalling through ghrelin receptors. Int J Biochem Cell Biol. 2005;37(4):887-900.
    [25]Duxbury MS, Waseem T, Ito H, et al. Ghrelin promotes pancreatic adenocarcinoma cellular proliferation and invasiveness. Biochem Biophys Res Commun. 2003;309(2):464-8.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700