用户名: 密码: 验证码:
炎性反应与高脂饮食诱导的肥胖和胰岛素抵素抵抗关系的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
随着社会经济的发展和人们生活方式的改变,肥胖的发生率逐年上升,已经成为世界范围内的公共卫生问题。肥胖不仅仅是脂肪组织的过度聚集,同时与2型糖尿病、动脉粥样硬化、高血压和脂肪肝等慢性病密切相关。
     高脂饮食是肥胖发生的重要危险因素,但是高脂饮食条件下,并不是所有的个体都发生肥胖,而一部分发生肥胖,而另外一部分表现出了肥胖抵抗,目前相关的机制还未完全阐明。最近研究发现,高脂饮食与下丘脑炎性反应密切相关。下丘脑炎性因子表达的增加可以导致胰岛素和瘦素抵抗,从而引起机体的摄食紊乱和体重的增加。Toll样受体-4 (Toll like receptor 4, TLR4)作为保守的模式识别受体,在机体的免疫调控中有着重要作用。脑室注入饱和脂肪酸可以显著增加下丘脑TLR4和相关炎性基因的表达,TLR4抗体干预可逆转高脂饮食诱导的下丘脑炎性反应和胰岛素抵抗并降低食物的摄入量。核因子-κB (Nuclear factor-KB, NF-κB)作为TLR4的下游信号分子,在机体的炎性反应中起着关键的作用。最新研究发现NF-κB与下丘脑的胰岛素和瘦素抵抗也密切相关。利用外源性载体激活下丘脑的NF-κB可以引起下丘脑的胰岛素和瘦素敏感性降低,导致小鼠的摄食增加,而同样的方法抑制下丘脑NF-κB的活性后可使肥胖小鼠的摄食量降低,体重减轻。以上研究提示高脂饮食诱导的下丘脑炎性反应在机体的摄食紊乱和肥胖的发生中起着重要的作用。同样高脂饮食条件下,肥胖抵抗大鼠是否可以通过抑制下丘脑的炎性反应从而阻止肥胖的发生,目前还没有相关的报道。
     饮食诱导的肥胖个体通常伴随着胰岛素抵抗。但是有研究发现不同高脂饮食条件下,肥胖个体的胰岛素敏感性存在差异,富含多不饱和脂肪酸的高脂饮食可以改善肥胖个体的胰岛素敏感性。目前,相关的机制还不是很清楚。近年来,肥胖被认为是一种低度慢性炎症状态。长期低度的炎性反应可以导致外周的胰岛素抵抗,引起糖脂代谢紊乱。脂肪组织在肥胖个体伴随的炎性反应有着极其重要的作用,脂肪细胞与免疫细胞相互作用促进炎性因子分泌的增加,进一步研究发现脂肪组织中的巨噬细胞是炎性因子分泌的主要来源,伴随着巨噬细胞浸润的增加,炎性因子分泌增加。在介导巨噬细胞迁移和聚集的因素中,骨桥蛋白(osteopontin, OPN)因其较强的粘附趋化能力及在细胞迁移过程中的重要作用而备受关注。还有研究发现,高脂饮食条件下,OPN-/-小鼠与野生型小鼠相比体重无显著变化,但是胰岛素敏感性却显著增加,静脉注入OPN抗体可以明显改善饮食诱导肥胖小鼠的胰岛素敏感性。以上研究说明OPN介导的脂肪组织炎性反应在高脂饮食诱导的胰岛素抵抗的发生中起着重要的作用。但OPN介导的脂肪组织炎性反应是否参与了脂肪酸类型不同高脂饮食诱导的肥胖大鼠的胰岛素敏感性差异,目前尚不清楚。
     为了了解炎性反应和高脂饮食诱导的肥胖易感性及胰岛素敏感性差异的关系,进一步探讨肥胖和胰岛素抵抗发生的机制,我们分别建立了肥胖和肥胖抵抗动物模型以及脂肪酸类型不同的高脂饮食诱导的肥胖动物模型,分析了不同模型中大鼠下丘脑或脂肪组织炎性基因的表达变化,同时观察了膳食干预对炎性基因表达的影响。
     第一部分下丘脑炎性反应和高脂饮食诱导的肥胖易感性差异关系的研究
     目的:建立高脂饮食诱导的肥胖和肥胖抵抗动物模型,探讨下丘脑炎性反应与高脂饮食诱导的肥胖易感性差异的关系,同时观察低脂饮食干预对大鼠体重和下丘脑炎性反应的影响。
     方法:雄性清洁级SD大鼠55只,适应性喂养1周之后,随机分为高脂饲料组(HF,n=45)和基础饲料对照组(CF,n=10)。基础饲料由华中科技大学同济医学院实验动物中心提供;每100g高脂饲料含有基础饲料60g,猪油15g,蛋黄粉13g,干酪素10g,白糖2g。基础饲料总热能3.29kcal/g(碳水化合64.44%、蛋白质21.88%、脂肪13.68%);高脂饲料总热能4.62kcal/g(碳水化合物30.15%、蛋白质22.00%、脂肪49.85%)。第10周时,高脂实验组大鼠体重基本稳定,参照对照组大鼠的平均体重及标准差,将高脂饲料组大鼠分为饮食诱导肥胖(Dietary induced obesity, DIO)大鼠(体重大于对照组平均体重加1.96倍标准差)和饮食诱导肥胖抵抗大鼠(Dietary induced obesity resistant, DIO-R)大鼠(体重小于对照组平均体重加1倍标准差),体重介于二者之间的大鼠不纳入本次研究。再分别将DIO和DIO-R大鼠随机分为两组,一组继续高脂饲料喂养,一组转为基础饲料喂养,干预8周。各组大鼠自由摄食和饮水,动物房温度(20±2)℃,相对湿度(50+10)%,明暗周期12比12。每天记录进食量,每周记录体重。分别与实验开始、10周和实验结束时过夜禁食12h采集尾血,离心之后保存待测。实验结束时,大鼠断头处死,迅速分离下丘脑并置液氮冻存,24小时后转移至-80℃冰箱保存,以备RNA抽提。同时分离大鼠的肾周和睾周脂肪组织并准确称重。实时定量PCR法(Real-time PCR)检测各组大鼠TLR4、NF-κB、IL-1β和IL-6基因表达水平。结果:1.喂养10周后,高脂饲料组中18只大鼠判定为肥胖易感大鼠(DIO),12只判定为肥胖抵抗大鼠(DIO-R)。DIO组大鼠的能量摄入和能量利用率显著高于DIO-R和CF组,但DIO-R组与CF组相比无统计学差异。DIO组大鼠的TC、TG、LDL-C、FBG、FINS和HOMA-IR显著高于DIO-R和CF组大鼠(P<0.05),DIO-R大鼠的TC、LDL-C、FINS和HOMA-IR与CF组相比显著增高(P<0.05)
     2. DIO/HF组大鼠下丘脑TLR4、NF-κB、IL-6和IL-1β表达水平显著高于DIO-R/HF和CF组大鼠,DIO-R/HF与CF组相比(P<0.05),下丘脑TLR4、NF-κB、IL-1β和IL-6表达水平没有变化。
     3.低脂饲料干预后,DIO/CF组大鼠的体重和脂肪蓄积量与DIO/HF组相比显著下降,DIO-R/CF的体重与DIO-R/HF组大鼠相比无变化,但脂肪蓄积量显著降低。DIO/CF和DIO-R/CF与其相应的高脂组相比,能量摄入无显著变化,能量利用率显著降低。DIO/CF组大鼠的TC、FINS和HOMA-IR与相应的高脂组相比显著降低。DIO/CF和DIO-R/CF组大鼠下丘脑TLR4、NF-κB、IL-6和IL-1β的表达水平与其相对应的高脂组相比无显著差异,DIO/CF组大鼠下丘脑炎性基因的表示仍显著高于CF组大鼠。
     结论:猪油配制的高脂饲料喂养10周可以诱导大鼠肥胖易感性差异的发生,高脂饮食诱导的下丘脑炎性反应可能使肥胖易感大鼠的摄食紊乱,从而导致能量摄入增加和肥胖的发生,而肥胖抵抗大鼠可以通过抑制下丘脑炎性反应保持适宜的摄食和稳定的体重。低脂饮食干预可以显著降低肥胖易感大鼠的体重和脂肪蓄积量,但对肥胖大鼠下丘脑炎性基因的表达没有影响。
     第二部分脂肪组织炎性反应与脂肪酸类型不同的高脂饮食诱导的肥胖大鼠胰岛素敏感性差异的研究
     目的:建立富含饱和脂肪酸高脂饮食和富含多不饱和脂肪酸高脂饮食诱导的肥胖大鼠模型,探讨脂肪组织炎性反应与不同类型脂肪酸高脂饮食诱导的肥胖大鼠胰岛素敏感性差异的关系,并观察低脂饮食干预对胰岛素敏感性和脂肪组织炎性基因表达的影响。
     方法:雄性清洁级SD大鼠100只,适应性喂养1周之后,随机分为猪油高脂饲料组(SF,n=45)、大豆油高脂饲料组(PF,n=45)和基础饲料对照组(CF,n=10)。基础饲料由华中科技大学同济医学院实验动物中心提供;每100g高脂饲料含有基础饲料60g,猪油/大豆油15g,蛋黄粉13g,干酪素10g,白糖2g。基础饲料总热能3.29kcal/g(碳水化合64.44%、蛋白质21.88%、脂肪13.68%、);高脂饲料总热能4.62kcal/g(碳水化合物30.15%、蛋白质22.00%、脂肪49.85%)。第10周时,高脂实验组大鼠体重基本稳定,参照对照组大鼠的平均体重及标准差,分别筛选出富含饱和脂肪酸高脂饲料诱导的肥胖大鼠(Saturated fat diet induced obesity, SF-DIO)和富含多不饱和脂肪酸诱导的肥胖大鼠(polyunsaturated fat diet induced obesity, PF-DIO)大鼠。再分别将SF-DIO和PF-DIO大鼠随机分为两组,
     一组继续高脂饲料喂养,一组转为基础饲料喂养,继续喂养8周。实验期间,各组大鼠自由摄食和饮水,动物房温度(20+2)℃,相对湿度(50±10)%,明暗周期12比12。每天记录进食量,每周记录体重。分别与实验开始、10周和实验结束时过夜禁食12h采集尾血,离心之后保存待测。实验结束后,大鼠断头处死,迅速分离出大鼠肾周与睾周脂肪组织,准确称重后置液氮快速冷冻,-80℃冰箱保存待测。酶联免疫法和免疫印记法(Western Blotting)分别检测大鼠血清和脂肪组织OPN蛋白表达;实时定量PCR法(Real-time PCR)检测各组大鼠OPN、NF-κB、IL-6和IL-10基因表达水平。
     结果:1.猪油高脂饲料和大豆油高脂饲料组大鼠的肥胖发生率分别为42.2%(SF-DIO)和48.9%(PF-DIO),两组大鼠肥胖发生率无显著差异。SF-DIO组大鼠的FBG、FINS和HOMA-IR显著高于PF-DIO组大鼠和CF组大鼠(P<0.05);PF-DIO组大鼠的FINS和HOMA-IR显著高于CF组,而FBG在两组间的差异无显著性。
     2. SF-DIO/SF大鼠脂肪组织OPN的基因和蛋白表达水平以及NF-κB和IL-6的基因表达水平显著高于PF-DIO/PF和CF组大鼠,同时,PF-DIO/PF组OPN、NF-κB和IL-6的表达水平也显著高于CF组,SF-DIO/SF和PF-DIO/PF组大鼠脂肪组织IL-10表达无差异,并显著低于CF组大鼠。
     3.低脂干预以后,SF-DIO/CF和PF-DIO/CF组大鼠的体重和脂肪蓄积量与相应的高脂组相比显著下降,能量摄入无显著变化。同时,SF-DIO/CF的FINS和HOMA-IR与SF-DIO/SF组相比显著降低,PF-DIO/CF的HOMA-IR与PF-DIO/PF组相比显著降低。SF-DIO/CF和PF-DIO/CF组大鼠脂肪组织OPN的基因和蛋白表达以及NF-κB和IL-6的基因表达与其相应的高脂组相比显著降低,但SF-DIO/CF组脂肪组织OPN和NF-κB表达水平仍显著高于PF-DIO/CF与CF组大鼠,PF-DIO/CF与CF组相比无显著差异,SF-DIO/CF和PF-DIO/CF组大鼠脂肪组织的IL-10的表达出现了增加的趋势,但与其相应的高脂组相比,差异无显著意义,仍显著低于基础饲料对照组大鼠。血清OPN含量在各个组间无统计学差异。
     结论:能量密度相同而脂肪酸类型不同的高脂饲料对SD大鼠具有相同的肥胖诱导作用,但是对肥胖大鼠的胰岛素敏感性具有不同的影响。饱和脂肪酸高脂饮食通过上调OPN的表达,促进脂肪组织炎性反应从而诱导肥胖大鼠的胰岛素抵抗;而多不饱和脂肪酸高脂饮食则可以抑制脂肪组织OPN的表达,降低脂肪组织炎性反应从而改善肥胖大鼠的胰岛素敏感性。低脂饮食干预可以增强肥胖大鼠的胰岛素敏感性并降低脂肪组织炎性基因的表达。
     创新性:
     1.首次探讨了高脂饮食诱导的肥胖和肥胖抵抗大鼠下丘脑炎性基因表达的变化并进一步观察了低脂饮食干预对不同肥胖易感性大鼠下丘脑炎性表达的影响,为继续探讨肥胖发生的机制及预防和治疗途径提供了新的思路和方法。
     2.首次针对性的探讨了脂肪组织炎症与脂肪酸类型不同高脂饮食诱导的肥胖大鼠胰岛素敏感性差异的关系,为预防和改善胰岛素抵抗提供了一条新的途径,同时观察了低脂饮食干预后炎性基因表达的变化,进一步阐明了脂肪酸的含量和类型对肥胖大鼠胰岛素敏感性的影响。
The prevalence of obesity is growing rapidly and has become a major public health problem worldwide. In addition to the fat accumulation, obesity also plays a crucial role in the development of metabolic syndrome (MS).
     Dietary fat has been recognized as a major risk factor for the development of obesity. However, both human beings and rodents appear to be different in developing obesity when they exposed to high fat diet, described as dietary induced obesity (DIO) and dietary induced obesity resistant (DIO-R). The potential mechanism related to the different susceptibility to obesity induced by high fat diet has not been clearly elucidated. Recently, many studies revealed that increased inflammatory response in hypothalamus produces insulin and leptin resistance and contributes to the energy imbalance. TLRs are pattern recognition receptors providing the first line of host defense. Emerging study proposed that TLR4 act as a predominant molecular target for saturated fatty acids in the hypothalamus trigger the inflammatory response and ultimately results to leptin resistance and weight gain. Moreover, pharmacological inhibition designed to disrupt TLR4 signaling could reverse leptin resistance and protect against high fat diet induced food intake. NF-κB was a master switch of innate inmmunity and also associated with diminished hypothalamic insulin and leptin signal transduction. Further, experimental and genetic interventions that block the hypothalamic NF-κB signaling reversed hypothalamic insulin and leptin resistance and was associated with reduced food intake and weight loss. These data collectively implicated that the activation of hypothalamic inflammation is necessary and sufficient for the control of energy intake and likely involved in the mechanism underlying the pathogenesis of obesity during high fat diet feeding. We wonder wether the hypothalamic inflammatory response was involved in the different obesity susceptibility induced by high fat diet.
     Additionally, high fat diet induced obesity usually was associated with peripheral insulin resistance. Many studies have demonstrated that saturated fat diet and polyunsaturated fat diet had different effect on insulin sensitivity and the related mechanism remain unclear. Recently, obesity is considered to be a low grade chronic inflammatory state and associated with insulin resistance. Adipose tissue is major endocrine tissue promoting the interaction between metabolism and inflammation. Further research showed that the macrophages in adipose tissue are a major source of inflammatory cytokine. Among all the factors associated with migration and aggregation of macrophages, OPN (osteopontin) received more attention for its strong chemotactic ability. Further research found that OPN was also related with insulin resistance. OPN-/- mice could prevent the insulin resistance induced by high fat diet compared with the OPN+/+mice which hadthe same body weight. Moreover, the antibody of OPN could improve the insulin resistance in obese mice. These findings collectively demonstrated that OPN play an important role in the development of insulin resistance. However, it remains unclear wether OPN is involved in the different insulin sesentivity induced by saturated fat diet and polyunsaturated fat diet.
     Therefore, we established different animal model for investigate the underlying mechanism of different obesity susceptibility and insulin sensitivity induced by high fat diet and observe the influence of low fat diet on inflammation in hypothalamus and adipose tissue.
     Part I
     Hypothalamic inflammatory response and different obesity susceptibility induced by high fat diet
     Objective: To investigate the hypothalamic inflammation in dietary induced obesity (DIO) and dietary induced obesity resistant (DIO-R) rats for investigating the potential mechanism related to different susceptibility to obesity and their responses to low fat diet intervention.
     Methods:Fifty-five six-week-old outbred male SD rats (purchased from Shanghai Sippr-BK lab animal Co. Ltd.) weighing 150-160g were housed individually with regulated temperature (22±5℃) and humidity (50±10%) on a daily cycle of 12 h light and darkness (08:00-20:00 h). All rats were allowed ad libitum access to water and food throughout the experimental period. After a week acclimation, tail blood was collected and serum was stored under-80℃for further assay. Then the rats were randomly divided into two groups:the HF group (n=45) were placed on a high-fat diet containing 4.62kcal/g (49.85% fat,20.00% protein, and 30.15% carbohydrate) and the CF group (n=10) were remained on normal laboratory chow food (purchased from Tongji Medical college lab animal center, Wuhan) containing 3.29kcal/g (13.68% fat,21.88% protein, and 64.44% carbohydrate). Dietary intake was recorded daily and body weight was measured weekly in the morning throughout the study. After 10 weeks of free access to their corresponding diet, rats in HF group with body weights more than x+1.96s of CF group were designated as DIO and those with body weight less than x+1.0s of CF group designated as DIO-R rats. Then one half of the DIO and DIO-R rats were switched to chow diet and the other half of the DIO rats and DIO-R rats were kept on HF diets for the following 8 weeks. All rats were provided water and food ad libitum. Terminally, all animals were killed after 12h fasting. Trunk blood was collected and centrifuged and serum was stored for further use. Perirenal and epididymal white adipose tissue was dissected and weighed. Hypothalamus was located and isolated according to brain coronal plane iconography of rat and related articles Samples of hypothalamic tissues were snap-frozen in liquid nitrogen immediately and stored at-80℃for RNA extraction.
     Results:1. Rats fed high fat diet had a wide distribution in body weight and weight gain.18 of the 45 were designated as DIO rats and 12 of which were DIO-R rats. The DIO rats had higher energy intake and energy efficiency compared with DIO-R and CF rats. TC、TG、LDL-C、FBG、FINS和HOMA-IR in DIO rats was increased compared with DIO-R and CF rats, TC、LDL-C and FINS in DIO-R rats was higher than that in CF rats.
     2. At the end of experiment, Hypothalamic TLR4, NF-κB, IL-1βand IL-6 mRNA expression in DIO/HF rats was significantly increased compared to DIO-R/HF and CF rats (P<0.05).
     3. Switching to chow food from high fat diet reduced the body weight and fat mass in DIO-R rats. The TC、FINS and HOMA-IR in DIO/CF rats was decreased compared with their counterpart on high fat diet. Low fat diet intervention failed to affect hypothalamic TLR4, NF-κB, IL-1βand IL-6 mRNA expression in DIO rats.
     Conclusion:SD rat fed a high fat diet showed different susceptibility to obesity. Upregulated hypothalamic inflammation may contribute to the overeating and development of obesity susceptibility induced by high fat diet and obesity resistant rats could main the appropriate energy intake and body weight by inhibiting the increased hypothalamic inflammatory response. While reduced body weight in DIO rats by chow food was not through correcting hypothalamic inflammation during the intervention period.
     PartⅡ
     Inflammatory response in adipose tissuce and different insulin sensitivity induced by saturated fat diet and polyunsaturated fat diet
     Objective:To explore the expression of OPN and related inflammatory factor in adipose tissue for investigating the underlying mechanism of the different insulin sensitivity induced by saturated and polyunsaturated fatty acid and their response to low fat diet.
     Methods:100 male SD rats weighing 150-160 g were housed individually in cages under controlled conditions. All rats had ad libitum access to their respective food and water throughout the study. After a week acclimation, tail blood was collected and serum was stored under-80℃for further assay. Then the rats were randomly divided into three groups, the SF group (n=45) were placed on a saturated fat diet containing 4.62kcal/g (49.85% fat,20.00% protein, and 30.15% carbohydrate), PF group (n=45) were placed on a polyunsaturated fat diet containing 4.62kcal/g (49.85% fat,20.00% protein, and 30.15% carbohydrate)and the CF group (n=10) remained on normal laboratory chow food (purchased from Tongji Medical college lab animal center, Wuhan) containing 3.29kcal/g (13.68% fat,21.88% protein, and 64.44% carbohydrate).At the end of 10th week, rats that fed high-fat diet with body weights more than x+1.96s of CF group were classified as saturated fat diet induced obesity (SF-DIO) and polyunsaturated fat diet induced obesity (PF-DIO). The SF-DIO and PF-DIO rats were then randomly subdivided into two groups respectively:one subgroup of each was shifted to chow food (SF-DIO/CF and PF-DIO/CF) and the other maintained on their respective high-fat diet (SF-DIO/SF and PF-DIO/PF). After another 8 weeks of feeding, all rats were sacrificed after 12 h fasting and trunk blood was collected and serum samples were stored at-80℃. The epididymal adipose tissue were rapidly separated, immediately frozen in liquid nitrogen, and then stored at-80℃until analysis. OPN, NF-κB, IL-6 and IL-10 mRNA expression in adipose tissue were determined by RT-PCR. Western blotting was used to determine OPN protein expression in adipose tissue and ELISA for serum OPN.
     Results:1. At the end of the 10th week,19 of SF group (42.2%) and 22 of PF group (48.9%) rats were designated as SF-DIO and PF-DIO respectively according to the body weight as compared with that of CF group. Serum FBG, FINS and HOMA-IR in SF-DIO group were significantly higher than PF-DIO and CF group, while the FINS and HOMA-IR in PF-DIO group were significantly higher than CF group.
     2. SF-DIO/SF rats had higher OPN, NF-κB and IL-6 expression compared with PF-DIO/PF and CF group, and PF-DIO/PF also had higher OPN, NF-κB and IL-6 expression compared with CF group, IL-10 expression in SF-DIO and PF-DIO rats was significantly reduced compared with CF control.
     3. Switching to chow food from high fat diet reduced the body weight and fat mass both in SF-DIO and PF-DIO rats without alteration of energy. FINS and HOMA-IR in SF-DIO/CF rats was reduced compared with SF-DIO/SF rats, and HOMA-IR in PF-DIO/CF rats was lower than that in PF-DIO/PF rats. The expression of OPN, NF-κB and IL-6 in SF-DIO/CF and PF-DIO/CF rat was decreased compared with their counterpart on high fat diet, SF-DIO/CF rats remained higher OPN and NF-κB expression than PF-DIO/CF rats. No difference in serum OPN was found among the group.
     Conclusion:Polyunsaturated fat diet showed improved insulin sensitivity compared with the saturated fat diet although they had the same effect on the development of obesity. The increased expression of OPN and related inflammatory factor in SF-DIO rats might be responsible for the development of insulin resistance, while inhibited expression of OPN and related inflammatory factors in PF-DIO rats compared with SF-DIO rats might be contributed to improved insulin sensitivity. And the improved insulin sensitivity by chow food both in SF-DIO and PF-DIO rats may be through the reduced the inflammatory response in adipose tissue during the intervention period.
引文
[1]International besity taskforce. http://www.iaso.org/iotf/.2008.
    [2]Wu Y F, Ma G S, Hu Y H, et al. The current prevalence status of body overweight and obesity in China: data from the China National Nutrition and Health Survey. Zhonghua Yu Fang Yi Xue Za Zhi 2005.39(5):316-320.
    [3]中华人民共和国教育部、国家体育总局、卫生部等.2005年中国学生体质与健康研究报告.2008.
    [4]Hu F B, Manson J E, Stampfer M J, et al. Diet, lifestyle, and the risk of type 2 diabetes mellitus in women. N Engl J Med 2001.345(11):790-797.
    [5]Ginsberg H N, Zhang Y L, and Hernandez-Ono A. Metabolic syndrome:focus on dyslipidemia. Obesity (Silver Spring) 2006.14 Suppl 1:41S-49S.
    [6]Kolovou G D, Anagnostopoulou K K, and Cokkinos D V. Pathophysiology of dyslipidaemia in the metabolic syndrome. Postgrad Med J 2005.81(956): 358-366.
    [7]Ora J, Laveneziana P, Wadell K, et al. Effect of Obesity on Respiratory Mechanics during Rest and Exercise in COPD. J Appl Physiol.
    [8]Calle E E, Rodriguez C, Walker-Thurmond K, et al.. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 2003.348(17):1625-1638.
    [9]Boutelle K N, Hannan P, Fulkerson J A, et al.. Obesity as a prospective predictor of depression in adolescent females. Health Psychol.29(3):293-298.
    [10]Bray G A and Popkin B M. Dietary fat intake does affect obesity! Am J Clin Nutr 1998.68(6):1157-1173.
    [11]Seidell J C. Dietary fat and obesity:an epidemiologic perspective. Am J Clin Nutr 1998.67(3 Suppl):546S-550S.
    [12]Levin B E, Triscari J, Hogan S, et al. Resistance to diet-induced obesity:food intake, pancreatic sympathetic tone, and insulin. Am J Physiol 1987.252(3 Pt 2): R471-478.
    [13]Bouchard C and Tremblay A. Genetic influences on the response of body fat and fat distribution to positive and negative energy balances in human identical twins. J Nutr 1997.127(5 Suppl):943S-947S.
    [14]Cancello R and Clement K. Is obesity an inflammatory illness? Role of low-grade inflammation and macrophage infiltration in human white adipose tissue. Bjog 2006.113(10):1141-1147.
    [15]De Souza C T, Araujo E P, Bordin S, et al. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 2005.146(10):4192-4199.
    [16]Carvalheira J B, Ribeiro E B, Araujo E P, et al. Selective impairment of insulin signalling in the hypothalamus of obese Zucker rats. Diabetologia 2003.46(12): 1629-1640.
    [17]Howard J K, Cave B J, Oksanen L J, et al. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med 2004.10(7):734-738.
    [18]Bouchard C, Tremblay A, Despres J P, et al. The response to long-term overfeeding in identical twins. N Engl J Med 1990.322(21):1477-1482.
    [19]West D B, Boozer C N, Moody D L, et al.. Dietary obesity in nine inbred mouse strains. Am J Physiol 1992.262(6 Pt 2):R1025-1032.
    [20]Wang C, Yang N, Wu S, et al. Difference of NPY and its receptor gene expressions between obesity and obesity-resistant rats in response to high-fat diet. Horm Metab Res 2007.39(4):262-267.
    [21]Zhang Y, Proenca R, Maffei M, et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994.372(6505):425-432.
    [22]Posey K A, Clegg D J, Printz R L, et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. Am J Physiol Endocrinol Metab 2009.296(5):E1003-1012.
    [23]Means T K, Golenbock D T, and Fenton M J. The biology of Toll-like receptors. Cytokine Growth Factor Rev 2000.11(3):219-232.
    [24]Akira S and Takeda K. Toll-like receptor signalling. Nat Rev Immunol 2004.4(7): 499-511.
    [25]Akira S, Uematsu S, and Takeuchi O. Pathogen recognition and innate immunity. Cell 2006.124(4):783-801.
    [26]Milanski M, Degasperi G, Coope A, et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus:implications for the pathogenesis of obesity. J Neurosci 2009. 29(2):359-370.
    [27]Tsukumo D M, Carvalho-Filho M A, Carvalheira J B, et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 2007.56(8):1986-1998.
    [28]Zhang X, Zhang G, Zhang H, et al. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell 2008.135(1): 61-73.
    [29]Allen Y S, Adrian T E, Allen J M, et al. Neuropeptide Y distribution in the rat brain. Science 1983.221(4613):877-879.
    [30]Beck B. Intracerebroventricular injection of proinsulin C-peptide does not influence food consumption in male Long-Evans rats. Horm Metab Res 2006. 38(5):314-316.
    [31]Larionov A, Krause A, and Miller W. A standard curve based method for relative real time PCR data processing. BMC Bioinformatics 2005.6:62.
    [32]Sondergaard L. Homology between the mammalian liver and the Drosophila fat body. Trends Genet 1993.9(6):193.
    [331 Wellen K E and Hotamisligil G S. Inflammation, stress, and diabetes. J Clin Invest 2005.115(5):1111-1119.
    [34]Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Immunol 2001. 1(2):135-145.
    [35]Lee J Y, Sohn K H, Rhee S H, et al.. Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through Toll-like receptor 4. J Biol Chem 2001.276(20):16683-16689.
    [36]Hayden M S and Ghosh S. Shared principles in NF-kappaB signaling. Cell 2008. 132(3):344-362.
    [37]Aguirre V, Uchida T, Yenush L, et al. The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). J Biol Chem 2000.275(12):9047-9054.
    [38]Banks W A, Willoughby L M, Thomas D R, et al. Insulin resistance syndrome in the elderly: assessment of functional, biochemical, metabolic, and inflammatory status. Diabetes Care 2007.30(9):2369-2373.
    [39]Rotter V, Nagaev I, and Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem 2003.278(46):45777-45784.
    [40]Emanuelli B, Glondu M, Filloux C, et al. The potential role of SOCS-3 in the interleukin-1beta-induced desensitization of insulin signaling in pancreatic beta-cells. Diabetes 2004.53 Suppl 3:S97-S103.
    [41]Rosenblum C I, Tota M, Cully D, et al. Functional STAT 1 and 3 signaling by the leptin receptor (OB-R); reduced expression of the rat fatty leptin receptor in transfected cells. Endocrinology 1996.137(11):5178-5181.
    [42]Bullen J W, Ziotopoulou M, Ungsunan, et al. Short- term resistance to diet induced obesity in A/J mice is not associated with regulation of hypothalamic neuropeptides. Am J Physiol endocrinol Metab.2004.287(4):E662-670.
    [43]Kendall A, Levitsky D A, Strupp B J, et al. Weight loss on a low-fat diet: consequence of the imprecision of the control of food intake in humans. Am J Clin Nutr 1991.53(5):1124-1129.
    [44]Hill J O, Dorton J, Sykes M N, et al. Reversal of dietary obesity is influenced by its duration and severity. Int J Obes 1989.13(5):711-722.
    [45]Enriori P J, Evans A E, Sinnayah P, et al. Diet-induced obesity causes severe but reversible leptin resistance in arcuate melanocortin neurons. Cell Metab 2007. 5(3):181-194.
    [46]Ford E S, Giles W H, and Dietz W H. Prevalence of the metabolic syndrome among US adults:findings from the third National Health and Nutrition Examination Survey. Jama 2002.287(3):356-359.
    [47]Gu D, Reynolds K, Wu X, et al. Prevalence of the metabolic syndrome and overweight among adults in China. Lancet 2005.365(9468):1398-1405.
    [48]Reaven G. The metabolic syndrome or the insulin resistance syndrome? Different names, different concepts, and different goals. Endocrinol Metab Clin North Am 2004.33(2):283-303.
    [49]Golay A and Ybarra J. Link between obesity and type 2 diabetes. Best Pract Res Clin Endocrinol Metab 2005.19(4):649-663.
    [50]Bastard J P, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006. 17(1):4-12.
    [51]Ogston D and McAndrew G M. Fibrinolysis in Obesity. Lancet 1964.2(7371): 1205-1207.
    [52]Hotamisligil G S, Shargill N S, and Spiegelman B M. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993.259(5091):87-91.
    [53]Feinstein R, Kanety H, Papa M Z, Lunenfeld B, and Karasik A. Tumor necrosis factor-alpha suppresses insulin-induced tyrosine phosphorylation of insulin receptor and its substrates. J Biol Chem 1993.268(35):26055-26058.
    [54]Hotamisligil G S. Inflammatory pathways and insulin action. Int J Obes Relat Metab Disord 2003.27 Suppl 3:S53-55.
    [55]Stephens J M, Lee J, and Pilch P F. Tumor necrosis factor-alpha-induced insulin resistance in 3T3-L1 adipocytes is accompanied by a loss of insulin receptor substrate-1 and GLUT4 expression without a loss of insulin receptor-mediated signal transduction. J Biol Chem 1997.272(2):971-976.
    [56]Duncan B B, Schmidt M I, Pankow J S, et al. Low-grade systemic inflammation and the development of type 2 diabetes:the atherosclerosis risk in communities study. Diabetes 2003.52(7):1799-1805.
    [57]Spranger J, Kroke A, Mohlig M, et al. Inflammatory cytokines and the risk to develop type 2 diabetes:results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes 2003.52(3):812-817.
    [58]Permana P A, Menge C, and Reaven P D. Macrophage-secreted factors induce adipocyte inflammation and insulin resistance. Biochem Biophys Res Commun 2006.341(2):507-514.
    [59]Denhardt D T and Guo X. Osteopontin:a protein with diverse functions. Faseb J 1993.7(15):1475-1482.
    [60]Todoric J, Loffler M, Huber J, et al. Adipose tissue inflammation induced by high-fat diet in obese diabetic mice is prevented by n-3 polyunsaturated fatty acids. Diabetologia 2006.49(9):2109-2119.
    [61]Levin B E, Hogan S, and Sullivan A C. Initiation and perpetuation of obesity and obesity resistance in rats. Am J Physiol 1989.256(3 Pt 2):R766-771.
    [62]da Silva M H, Pithon T C, and do Nascimento C M. Effect of saturated and polyunsaturated fatty acids rich diets on hepatic and adipose tissue lipid metabolism in rats. Int J Vitam Nutr Res 1996.66(3):258-262.
    [63]Schroder H. Protective mechanisms of the Mediterranean diet in obesity and type 2 diabetes. J Nutr Biochem 2007.18(3):149-160.
    [64]Lovejoy J C, Smith S R, Champagne C M, et al. Effects of diets enriched in saturated (palmitic), monounsaturated (oleic), or trans (elaidic) fatty acids on insulin sensitivity and substrate oxidation in healthy adults. Diabetes Care 2002. 25(8):1283-1288.
    [65]Kraegen E W, Clark P W, Jenkins A B, et al. Development of muscle insulin resistance after liver insulin resistance in high-fat-fed rats. Diabetes 1991.40(11): 1397-1403.
    [66]许岭翎,向红丁,张荣,等.高脂饮食诱发血糖升高的动物模型中瘦素与胰岛素变化的关系.基础医学与临床2001.21(5):441-444.
    [67]史虹莉,陆肇曾,方京冲,等.不同脂肪酸组分饮食对OLETF雄性大鼠胰岛素抵抗的影响.中华内分泌代谢杂志2005.21(6):565-566.
    [68]Brake D K, Smith E O, Mersmann H, et al. ICAM-1 expression in adipose tissue: effects of diet-induced obesity in mice. Am J Physiol Cell Physiol 2006.291(6): C1232-1239.
    [69]Chen A, Mumick S, Zhang C, et al. Diet induction of monocyte chemoattractant protein-1 and its impact on obesity. Obes Res 2005.13(8):1311-1320.
    [70]Zhu B, Suzuki K, Goldberg H A, et al. Osteopontin modulates CD44-dependent chemotaxis of peritoneal macrophages through G-protein-coupled receptors: evidence of a role for an intracellular form of osteopontin. J Cell Physiol 2004. 198(1):155-167.
    [71]Rollo E E and Denhardt D T. Differential effects of osteopontin on the cytotoxic activity of macrophages from young and old mice. Immunology 1996.88(4): 642-647.
    [72]Nomiyama T, Perez-Tilve D, Ogawa D, et al. Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J Clin Invest 2007.117(10):2877-2888.
    [73]Zheng W, Li R, Pan H, et al. Role of osteopontin in induction of monocyte chemoattractant protein 1 and macrophage inflammatory protein lbeta through the NF-kappaB and MAPK pathways in rheumatoid arthritis. Arthritis Rheum 2009.60(7):1957-1965.
    [74]Rice J, Courter D L, Giachelli C M, et al.. Molecular mediators of alphavbeta3-induced endothelial cell survival. J Vasc Res 2006.43(5):422-436.
    [75]Kiefer F W, Zeyda M, Todoric J, et al. Osteopontin expression in human and murine obesity:extensive local up-regulation in adipose tissue but minimal systemic alterations. Endocrinology 2008.149(3):1350-1357.
    [76]Chapman J, Miles P D, Ofrecio J M, et al. Osteopontin is required for the early onset of high fat diet-induced insulin resistance in mice. PLoS One 2010.5(11): e13959.
    [77]DeLany J P, Windhauser M M, Champagne C M, et al. Differential oxidation of individual dietary fatty acids in humans. Am J Clin Nutr 2000.72(4):905-911.
    [78]Fried S K, Bunkin D A, and Greenberg A S. Omental and subcutaneous adipose tissues of obese subjects release interleukin-6:depot difference and regulation by glucocorticoid. J Clin Endocrinol Metab 1998.83(3):847-850.
    [79]Steppan C M, Bailey S T, Bhat S, et al. The hormone resistin links obesity to diabetes. Nature 2001.409(6818):307-312.
    [80]Skurk T, Alberti-Huber C, Herder C, et al. Relationship between adipocyte size and adipokine expression and secretion. J Clin Endocrinol Metab 2007.92(3): 1023-1033.
    [81]Maziere C, Gomila C, and Maziere J C. Oxidized low-density lipoprotein increases osteopontin expression by generation of oxidative stress. Free Radic Biol Med.48(10):1382-1387.
    [82]Arai T, Kim H J, Chiba H, and Matsumoto A. Anti-obesity effect of fish oil and fish oil-fenofibrate combination in female KK mice. J Atheroscler Thromb 2009. 16(5):674-683.
    [83]Ashkar S, Weber G F, Panoutsakopoulou V, et al. Eta-1 (osteopontin):an early component of type-1 (cell-mediated) immunity. Science 2000.287(5454): 860-864.
    [84]Rosenfalck A M, Almdal T, Viggers L, et al. A low-fat diet improves peripheral insulin sensitivity in patients with Type 1 diabetes. Diabet Med 2006.23(4): 384-392.
    [85]Halber N K T, Trujillo ME. Hypoxia-inducible factor 1 induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 2009.29(16):4467-4483.
    [86]Benomar Y N N, Aubourgl A, et al. Insulin and leptin induce Glut4 plasmamembrane translocation and glucose uptake in a human neuronal cell line by a PI3-Kinase dependent. Enocrinology 2006.147(5):2550-2556.
    [87]Xu H, Barnes G T, Yang Q, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 2003. 112(12):1821-1830.
    [88]Boring L, Gosling J, Chensue S W, et al. Impaired monocyte migration and reduced type 1 (Thl) cytokine responses in C-C chemokine receptor 2 knockout mice. J Clin Invest 1997.100(10):2552-2561.
    [1]Medzhitov R, Preston-Hurlburt P, and Janeway C A, Jr. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 1997.388(6640):394-397.
    [2]Takeda K, Kaisho T, and Akira S. Toll-like receptors. Annu Rev Immunol 2003.21:335-376.
    [3]Kumar H, Kawai T, and Akira S. Toll-like receptors and innate immunity. Biochem Biophys Res Commun 2009.388(4):621-625.
    [4]Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Immunol 2001.1(2):135-145.
    [5]Erridge C, Moncayo-Nieto O L, Morgan R, Young M, and Poxton I R. Acinetobacter baumannii lipopolysaccharides are potent stimulators of human monocyte activation via Toll-like receptor 4 signalling. J Med Microbiol 2007.56(Pt 2):165-171.
    [6]Schnappinger D, Schoolnik G K, and Ehrt S. Expression profiling of host pathogen interactions:how Mycobacterium tuberculosis and the macrophage adapt to one another. Microbes Infect 2006.8(4):1132-1140.
    [7]Haeberle H A, Takizawa R, Casola A, et al. Respiratory syncytial virus-induced activation of nuclear factor-kappaB in the lung involves alveolar macrophages and toll-like receptor 4-dependent pathways. J Infect Dis 2002.186(9):1199-1206.
    [8]Bachar O, Adner M, Uddman R, and Cardell L O. Toll-like receptor stimulation induces airway hyper-responsiveness to bradykinin, an effect mediated by JNK and NF-kappa B signaling pathways. Eur J Immunol 2004. 34(4):1196-1207.
    [9]Kawai T and Akira S. TLR signaling. Cell Death Differ 2006.13(5): 816-825.
    [10]Yamamoto M, Sato S, Hemmi H, et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science 2003. 301(5633):640-643.
    [11]Hotamisligil G S. Inflammation and metabolic disorders. Nature 2006. 444(7121):860-867.
    [12]Hotamisligil G S, Shargill N S, and Spiegelman B M. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993.259(5091):87-91.
    [13]Zhang Y, Proenca R, Maffei M, et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994.372(6505):425-432.
    [14]Fried S K, Bunkin D A, and Greenberg A S. Omental and subcutaneous adipose tissues of obese subjects release interleukin-6:depot difference and regulation by glucocorticoid J Clin Endocrinol Metab 1998.83(3): 847-850.
    [15]Steppan C M, Bailey S T, Bhat S, et al. The hormone resistin links obesity to diabetes. Nature 2001.409(6818):307-312.
    [16]Fukuhara A, Matsuda M, Nishizawa M, et al. Visfatin:a protein secreted by visceral fat that mimics the effects of insulin. Science 2005.307(5708): 426-430.
    [17]Skurk T, Alberti-Huber C, Herder C, and Hauner H. Relationship between adipocyte size and adipokine expression and secretion. J Clin Endocrinol Metab 2007.92(3):1023-1033.
    [18]Galic S, Oakhill J S, and Steinberg G R. Adipose tissue as an endocrine organ. Mol Cell Endocrinol 2010.316(2):129-139.
    [19]Wellen K E and Hotamisligil G S. Inflammation, stress, and diabetes. J Clin Invest 2005.115(5):1111-1119.
    [20]Lee J Y, Sohn K H, Rhee S H, and Hwang D. Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through Toll-like receptor 4. J Biol Chem 2001.276(20): 16683-16689.
    [21]Lee J Y, Ye J, Gao Z, et al. Reciprocal modulation of Toll-like receptor-4 signaling pathways involving MyD88 and phosphatidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids. J Biol Chem 2003.278(39):37041-37051.
    [22]Hwang D. Modulation of the expression of cyclooxygenase-2 by fatty acids mediated through toll-like receptor 4-derived signaling pathways. Faseb J 2001.15(14):2556-2564.
    [23]Shi H, Kokoeva M V, Inouye K, et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 2006.116(11):3015-3025.
    [24]Kahn C R. Banting Lecture. Insulin action, diabetogenes, and the cause of type II diabetes. Diabetes 1994.43(8):1066-1084.
    [25]White M F. Insulin signaling in health and disease. Science 2003. 302(5651):1710-1711.
    [26]陈瑜,李伶,杨刚毅。游离脂肪酸在2型糖尿病发病机制中的作用.国际检验医学杂志2008.28(10):915-917.
    [27]Arner P. Insulin resistance in type 2 diabetes--role of the adipokines. Curr Mol Med 2005.5(3):333-339.
    [28]Hotamisligil G S. Inflammatory pathways and insulin action. Int J Obes Relat Metab Disord 2003.27 Suppl 3:S53-55.
    [29]Stephens J M, Lee J, and Pilch P F. Tumor necrosis factor-alpha-induced insulin resistance in 3T3-L1 adipocytes is accompanied by a loss of insulin receptor substrate-1 and GLUT4 expression without a loss of insulin receptor-mediated signal transduction. J Biol Chem 1997.272(2):971-976.
    [30]Rotter V, Nagaev I, and Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem 2003.278(46):45777-45784.
    [31]Banks W A, Willoughby L M, Thomas D R, and Morley J E. Insulin resistance syndrome in the elderly:assessment of functional, biochemical, metabolic, and inflammatory status. Diabetes Care 2007.30(9):2369-2373.
    [32]Stein C J and Colditz G A. The epidemic of obesity. J Clin Endocrinol Metab 2004.89(6):2522-2525.
    [33]Poggi M, Bastelica D, Gual P, et al. C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia 2007.50(6):1267-1276.
    [34]Tsukumo D M, Carvalho-Filho M A, Carvalheira J B, et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 2007.56(8):1986-1998.
    [35]Kim F, Pham M, Luttrell I, et al. Toll-like receptor-4 mediates vascular inflammation and insulin resistance in diet-induced obesity. Circ Res 2007. 100(11):1589-1596.
    [36]Staiger H, Staiger K, Stefan N, et al. Palmitate-induced interleukin-6 expression in human coronary artery endothelial cells. Diabetes 2004. 53(12):3209-3216.
    [37]Senn J J. Toll-like receptor-2 is essential for the development of palmitate-induced insulin resistance in myotubes. J Biol Chem 2006. 281(37):26865-26875.
    [38]Nguyen M T, Favelyukis S, Nguyen A K, et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J Biol Chem 2007.282(48):35279-35292.
    [39]Schaeffler A, Gross P, Buettner R, et al. Fatty acid-induced induction of Toll-like receptor-4/nuclear factor-kappaB pathway in adipocytes links nutritional signalling with innate immunity. Immunology 2009.126(2): 233-245.
    [40]Hommelberg P P, Plat J, Langen R C, Schols A M, and Mensink R P. Fatty acid-induced NF-kappaB activation and insulin resistance in skeletal muscle are chain length dependent Am J Physiol Endocrinol Metab 2009.296(1): E114-120.
    [41]Lockridge J B, Sailors M L, Durgan D J, et al. Bioinformatic profiling of the transcriptional response of adult rat cardiomyocytes to distinct fatty acids. J Lipid Res 2008.49(7):1395-1408.
    [42]Reyna S M, Ghosh S, Tantiwong P, et al. Elevated toll-like receptor 4 expression and signaling in muscle from insulin-resistant subjects. Diabetes 2008.57(10):2595-2602.
    [43]Song M J, Kim K H, Yoon J M, and Kim J B. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun 2006.346(3):739-745.
    [44]Davis J E, Gabler N K, Walker-Daniels J, and Spurlock M E. Tlr-4 deficiency selectively protects against obesity induced by diets high in saturated fat. Obesity (Silver Spring) 2008.16(6):1248-1255.
    [45]Frisard M I, McMillan R P, Marchand J, et al. Toll-like receptor 4 modulates skeletal muscle substrate metabolism. Am J Physiol Endocrinol Metab.298(5):E988-998.
    [46]Takeuchi O and Akira S. Pattern recognition receptors and inflammation. Cell 2010.140(6):805-820.
    [47]Caricilli A M, Nascimento P H, Pauli J R, et al. Inhibition of toll-like receptor 2 expression improves insulin sensitivity and signaling in muscle and white adipose tissue of mice fed a high-fat diet J Endocrinol 2008. 199(3):399-406.
    [48]Erridge C and Samani N J. Saturated fatty acids do not directly stimulate Toll-like receptor signaling. Arterioscler Thromb Vase Biol 2009.29(11): 1944-1949.
    [49]Summers S A and Nelson D H. A role for sphingolipids in producing the common features of type 2 diabetes, metabolic syndrome X, and Cushing's syndrome. Diabetes 2005.54(3):591-602.
    [50]Fischer H, Ellstrom P, Ekstrom K, et al. Ceramide as a TLR4 agonist; a putative signalling intermediate between sphingolipid receptors for microbial ligands and TLR4. Cell Microbiol 2007.9(5):1239-1251.
    [51]Holland W L, Brozinick J T, Wang L P, et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metab 2007.5(3):167-179.
    [52]Cani P D and Delzenne N M. Interplay between obesity and associated metabolic disorders:new insights into the gut microbiota Curr Opin Pharmacol 2009.9(6):737-743.
    [53]Turnbaugh P J, Ley R E, Mahowald M A, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006. 444(7122):1027-1031.
    [54]Arkan M C, Hevener A L, Greten F R, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med 2005.11(2):191-198.
    [55]Cani P D, Amar J, Iglesias M A, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007.56(7):1761-1772.
    [56]Friedman J M. Leptin at 14 y of age:an ongoing story. Am J Clin Nutr 2009. 89(3):973S-979S.
    [57]Friedman J M. Obesity:Causes and control of excess body fat Nature 2009. 459(7245):340-342.
    [58]Gropp E, Shanabrough M, Borok E, et al. Agouti-related peptide-expressing neurons are mandatory for feeding. Nat Neurosci 2005.8(10):1289-1291.
    [59]Luquet S, Perez F A, Hnasko T S, and Palmiter R D. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 2005.310(5748):683-685.
    [60]Gao Q and Horvath T L. Neurobiology of feeding and energy expenditure. Annu Rev Neurosci 2007.30:367-398.
    [61]Plum L, Belgardt B F, and Bruning J C. Central insulin action in energy and glucose homeostasis. J Clin Invest 2006.116(7):1761-1766.
    [62]Belgardt B F, Husch A, Rother E, et al. PDK1 deficiency in POMC-expressing cells reveals FOXO1-dependent and -independent pathways in control of energy homeostasis and stress response. Cell Metab 2008.7(4):291-301.
    [63]Hill J W, Williams K W, Ye C, et al. Acute effects of leptin require PI3K signaling in hypothalamic proopiomelanocortin neurons in mice. J Clin Invest 2008.118(5):1796-1805.
    [64]Niswender K D, Morton G J, Stearns W H, et al. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature 2001.413(6858):794-795.
    [65]Plum L, Ma X, Hampel B, et al. Enhanced PIP3 signaling in POMC neurons causes KATP channel activation and leads to diet-sensitive obesity. J Clin Invest 2006.116(7):1886-1901.
    [66]Plum L, Rother E, Munzberg H, et al. Enhanced leptin-stimulated Pi3k activation in the CNS promotes white adipose tissue transdifferentiation. Cell Metab 2007.6(6):431-445.
    [67]Saxena N K, Sharma D, Ding X, et al. Concomitant activation of the JAK/STAT, PI3K/AKT, and ERK signaling is involved in leptin-mediated promotion of invasion and migration of hepatocellular carcinoma cells. Cancer Res 2007.67(6):2497-2507.
    [68]Uddin S, Bavi P, Siraj A K, et al. Leptin-R and its association with PI3K/AKT signaling pathway in papillary thyroid carcinoma Endocr Relat Cancer.17(1):191-202.
    [69]Plum L, Schubert M, and Bruning J C. The role of insulin receptor signaling in the brain. Trends Endocrinol Metab 2005.16(2):59-65.
    [70]Schwartz M W. Central nervous system regulation of food intake. Obesity (Silver Spring) 2006.14 Suppl 1:1S-8S.
    [71]Bruning J C, Gautam D, Burks D J, et al. Role of brain insulin receptor in control of body weight and reproduction. Science 2000.289(5487): 2122-2125.
    [72]Koch L, Wunderlich F T, Seibler J, et al. Central insulin action regulates peripheral glucose and fat metabolism in mice. J Clin Invest 2008.118(6): 2132-2147.
    [73]Konner A C, Janoschek R, Plum L, et al. Insulin action in AgRP-expressing neurons is required for suppression of hepatic glucose production. Cell Metab 2007.5(6):438-449.
    [74]Obici S, Zhang B B, Karkanias G, and Rossetti L. Hypothalamic insulin signaling is required for inhibition of glucose production. Nat Med 2002. 8(12):1376-1382.
    [75]Konner A C, Klockener T, and Bruning J C. Control of energy homeostasis by insulin and leptin:targeting the arcuate nucleus and beyond. Physiol Behav 2009.97(5):632-638.
    [76]Niswender K D and Schwartz M W. Insulin and leptin revisited:adiposity signals with overlapping physiological and intracellular signaling capabilities. Front Neuroendocrinol 2003.24(1):1-10.
    [77]Seino S and Miki T. Physiological and pathophysiological roles of ATP-sensitive K+ channels. Prog Biophys Mol Biol 2003.81(2):133-176.
    [78]Gao Q, Wolfgang M J, Neschen S, et al. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc Natl Acad Sci USA 2004. 101(13):4661-4666.
    [79]Bates S H, Stearns W H, Dundon T A, et al. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature 2003. 421(6925):856-859.
    [80]Banks W A, Kastin A J, Huang W, Jaspan J B, and Maness L M. Leptin enters the brain by a saturable system independent of insulin. Peptides 1996. 17(2):305-311.
    [81]Diamond F B, Jr., Eichler D C, Duckett G, et al. Demonstration of a leptin binding factor in human serum. Biochem Biophys Res Commun 1997. 233(3):818-822.
    [82]Carpenter L R, Farruggella T J, Symes A, et al. Enhancing leptin response by preventing SH2-containing phosphatase 2 interaction with Ob receptor. Proc Natl Acad Sci U S A 1998.95(11):6061-6066.
    [83]Crouse J A, Elliott G E, Burgess T L, et al. Altered cell surface expression and signaling of leptin receptors containing the fatty mutation. J Biol Chem 1998.273(29):18365-18373.
    [84]De Souza C T, Araujo E P, Bordin S, et al. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 2005.146(10):4192-4199.
    [85]Picardi P K, Calegari V C, Prada Pde O, et al. Reduction of hypothalamic protein tyrosine phosphatase improves insulin and leptin resistance in diet-induced obese rats. Endocrinology 2008.149(8):3870-3880.
    [86]Milanski M, Degasperi G, Coope A, et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus:implications for the pathogenesis of obesity. J Neurosci 2009.29(2):359-370.
    [87]Zhang X, Zhang G, Zhang H, et al. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell 2008. 135(1):61-73.
    [88]Posey K A, Clegg D J, Printz R L, et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet Am J Physiol Endocrinol Metab 2009.296(5):E1003-1012.
    [89]Bjorbaek C, El-Haschimi K, Frantz J D, and Flier J S. The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem 1999.274(42): 30059-30065.
    [90]Bence K K, Delibegovic M, Xue B, et al. Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med 2006.12(8):917-924.
    [91]Ernst M B, Wunderlich C M, Hess S, et al. Enhanced Stat3 activation in POMC neurons provokes negative feedback inhibition of leptin and insulin signaling in obesity. J Neurosci 2009.29(37):11582-11593.
    [92]Enriori P J, Evans A E, Sinnayah P, et al. Diet-induced obesity causes severe but reversible leptin resistance in arcuate melanocortin neurons. Cell Metab 2007.5(3):181-194.
    [93]Kleinridders A, Schenten D, Konner A C, et al. MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell Metab 2009.10(4):249-259.

© 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