用户名: 密码: 验证码:
TNFRIBP-Fc融合蛋白治疗肥胖相关的胰岛素抵抗及其机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
当今肥胖发病率越来越高,已经成为世界上普遍的社会问题之一。肥胖与胰岛素抵抗密切相关,是非胰岛素依耐型糖尿病发病的高危因素之一。胰岛素抵抗(insulin resistance , IR)是指机体对一定量胰岛素的生物学反应低于正常水平,即胰岛素敏感性下降,常伴随2型糖尿病、脂代谢异常、高血压及心血管疾病,将之统称为胰岛素抵抗综合征,又称X综合征。
     TNF-α是由激活的单核/巨噬细胞、淋巴细胞等分泌的细胞因子,介导炎症反应、免疫应答、抗肿瘤等多种生物学效应。近来发现脂肪组织也合成和分泌TNF-α,它在胰岛素抵抗的发生机制中起关键性作用。已知TNF-α可通过胰岛素受体底物-1(IRS-1)丝、苏氨酸磷酸化抑制其酪氨酸磷酸化,从而抑制胰岛素的信号转导,这个效应究竟是TNFRI或TNFRII介导的,现在尚无定论。
     本室前期工作从噬菌体12线肽库中筛选出TNFRI封闭肽(blocking peptide,BP),它可与分泌型TNF-α竞争性结合TNFRI,发挥拮抗TNF-α的作用,为了增加该肽的亲合力和延长其半衰期,我们将该肽的核苷酸序列插入到分泌性表达IgG1Fc段的真核表达载体pIG/3C,得到TNFRIBP和人IgG1Fc融合蛋白的表达载体pIG/3C-TNFRIBP-Fc。本课题用该载体与带neo筛选标记的pcDNA3.1共转染CHO-K1和BHK-21建立稳定细胞系,得到大量纯化的融合蛋白用于治疗肥胖相关的胰岛素抵抗,为开发治疗TNF-α相关疾病的肽类药物奠定基础。
     一、TNFRIBP-Fc融合蛋白的稳定表达与纯化
     1.建立稳定细胞系:分别将pIG/3C-TNFRIBP-Fc和pIG/3C-IgG1Fc载体和带有G418筛选标记的pcDNA3.1载体共转染CHO-k1和BHK-21细胞,经G418筛选,挑取高表达株,经三次有限稀释,得到七株稳定表达TNFRIBP-Fc融合蛋白的细胞系,三株稳定表达Fc蛋白的细胞系。
     2.融合蛋白和Fc蛋白的纯化:无血清扩大培养这些细胞株,收集上清,经protein A-Sepharose CL-4B亲和层析,得到纯化的融合蛋白和Fc蛋白,经双抗体夹心ELISA定量,证实TNFRIBP-Fc融合蛋白的平均表达效率约为500ng/ml,Fc蛋白的平均表达效率约为400ng/ml。
     3.SDS-PAGE鉴定融合蛋白和Fc蛋白的分子量:SDS-PAGE显示纯化后的融合蛋白的分子量为37kD左右,Fc蛋白的分子量大约是36kD。
     4.纯化的融合蛋白生物活性的检测:间接免疫荧光和MTT试验证实融合蛋白能特异性结合鼠细胞系L929细胞上的TNFRI,且160ng/ml的融合蛋白就可完全阻断TNFRI介导的100U/ml的分泌性TNF-α(sTNF-α)引起的细胞毒效应。
     二、TNFRIBP-Fc融合蛋白的体外试验
     1.体外诱导前脂肪细胞3T3-L1分化成脂肪细胞:前脂肪细胞系3T3L1先用0.5mM IBMX、1μM地塞米松和10μg/ml的胰岛素诱导三天,再用10μg/ml的胰岛素诱导两天,撤去胰岛素培养两天,诱导其分化成脂肪细胞的效率达90%以上。
     2.体外高糖诱导脂肪细胞胰岛素抵抗:用不同浓度的葡萄糖作用上述分化的脂肪细胞24小时,结果显示25mM的葡萄糖即可诱导分化的3T3-L1发生胰岛素抵抗,表现为在胰岛素刺激下对3H-2-脱氧葡萄糖的摄取降低150倍左右(p<0.05)。
     3.内源性两型TNF-α与高糖诱导脂肪细胞胰岛素抵抗的关系:用不同浓度的葡萄糖作用上述分化的脂肪细胞24小时,结果显示与15mM葡萄糖作用组相比,25mM葡萄糖诱导胰岛素抵抗时,脂肪细胞表面TM-TNF-α表达从81.4%降至3.56%,而培养上清中sTNF-α从63.73升高113.03 (pg/ml)。在诱导胰岛素抵抗的同时加入160ng/ml的融合蛋白或TACE抗体,明显改善高糖诱导的胰岛素抵抗,与25mM葡萄糖诱导抵抗组比,前者把细胞在胰岛素刺激下对糖摄取提高了20倍,后者提高了50倍。
     4.外源性两型TNF-α对脂肪细胞胰岛素应答的影响:分别用sTNF-α和TM-TNF-α作用分化的脂肪细胞24小时,观察细胞在胰岛素的刺激下对糖的摄取。结果显示与未刺激的对照组比,sTNF-α使脂肪细胞对胰岛素刺激的糖摄取降低了96%,而TM-TNF-α却使之增加了约2倍;同时加入融合蛋白或抗TNF抗体,可几乎完全逆转sTNFα诱导的胰岛素抵抗,同时也几乎完全阻断TM-TNFα提高脂肪细胞对胰岛素的敏感性的作用。
     5.两型TNFα对脂肪细胞胰岛素信号转导的影响:分别用sTNFα和TMTNF-α作用分化的脂肪细胞24小时,用Western检测IRS-1酪氨酸和丝氨酸磷酸化水平。结果显示sTNFα使IRS-1270位丝氨酸磷酸化水平升高而酪氨酸磷酸化水平下降,TM-TNF-α不影响正常胰岛素诱导的丝氨酸/酪氨酸磷酸化水平。
     三、TNFRIBP-Fc融合蛋白的体内试验
     1.高糖高脂饮食建立大鼠胰岛素抵抗的模型:选取体重在110~120g的雄性Wistar大鼠,给与普通饮食(ND)或者高糖高脂饮食(HFS)喂养16周,结果显示与普通饮食相比,高糖高脂饮食大鼠体重每月增加18%,血糖维持正常的同时表现出明显的高胰岛素血症,胰岛素抵抗指数HOMAIR升高了3倍。提示成功建立肥胖引起的胰岛素抵抗。
     2.治疗分组:第17周开始HFS组随机分成四组:融合蛋白治疗组(HFS+Fusion)、Fc蛋白治疗组(HFS+Fc)、TNF-α兔多抗治疗组(HFS+Ab)和单纯高糖高脂饮食对照组(HFS),每只大鼠尾静脉注射0.1ml生理盐水含500μg纯化的TNFRIBP-Fc融合蛋白或Fc蛋白或TNF-α兔多抗或0.1ml生理盐水,每周两次持续4周。普通饮食组(ND)保持不变。
     3.融合蛋白对体重、脂肪沉积和血清甘油三酯、总胆固醇的影响:HFS组体重增加值是ND组的2倍,附睾周围脂肪组织的重量是ND组1.8倍,脂肪细胞明显增大,血中甘油三酯水平是ND组的3.7倍。融合蛋白治疗明显改善了HFS诱导的脂肪沉积和高甘油三酯血症:体重增加值下降了26%,脂肪组织重量下降了30%,脂肪细胞体积也明显减小,甘油三酯水平下降了54%。血清胆固醇水平各组没有差异。抗体组的治疗效果最好,上述指标抗体组与普通饮食组之间没有显著差异。
     4.融合蛋白对糖代谢及胰岛素敏感性的影响:各组治疗前后空腹葡萄糖都在正常水平,但HFS组胰岛素水平、C肽和HOMAIR是ND组2倍左右,胰岛代偿性肥大,胰岛素和葡萄糖耐受实验说明胰岛素的敏感性相对ND组显著下降。融合蛋白治疗后,与HFS组比,胰岛素、C肽和HOMAIR都几乎下降了50%,胰岛代偿性肥大也得到缓解。胰岛素和葡萄糖耐受试验直接说明融合蛋白治疗后胰岛素敏感性显著增加,而Fc治疗则没有效果。抗体组治疗后敏感性与普通饮食组之间几乎没有显著差异。
     5.ELISA显示融合蛋白治疗对TNF-α蛋白水平的影响:用ELISA检测血清和组织蛋白提取液中TNF-α的水平。结果显示HFS组血清sTNF-α和肌肉、脂肪组织中TNF-α水平都明显增高,融合蛋白治疗使血清和脂肪组织中TNF-α的水平下降50%,使肌肉中TNF-α的水平下降了80%。肝脏组织各组之间没有差异。抗体组脂肪和肌肉组织TNF-α的水平仍比ND组高2.5和1.4倍。
     6.免疫组化显示融合蛋白对脂肪和胰腺组织TNFα蛋白水平的影响:用免疫组化比较脂肪和胰腺组织之间TNF-α表达水平。结果显示HFS组这两个组织TNF-α表达水平比ND组明显升高,融合蛋白和抗体治疗后TNFα水平显著下降。
     7.融合蛋白对IRS-1酪氨酸磷酸化水平的影响:用免疫共沉淀和Western比较各组IRS-1酪氨酸磷酸化水平。结果显示HFS组脂肪、肌肉组织和肝脏组织IRS-1酪氨酸磷酸化水平相对与ND组下降80%、68%和32%,融合蛋白治疗使脂肪和肌肉IRS-1酪氨酸磷酸化水平相对HFS组分别升高了54%和50%,肝脏组织IRS-1酪氨酸磷酸化水平各治疗组之间没有区别。抗体治疗使脂肪和肌肉IRS-1酪氨酸磷酸化水平相对HFS组分别升高了75%和59%。
     四、sTNFRⅡ的原核表达、活性鉴定及其多克隆抗体的制备
     1.sTNFRⅡ的原核表达:采用RT-PCR方法,从人外周血的单核细胞中扩增出人TNFRⅡ基因的胞外区,将其克隆入pET28a(+)高效表达载体,测序鉴定后得到正确的重组子,用IPTG诱导其表达后经镍柱纯化,得到sTNFRⅡ重组蛋白。经10%SDS-PAGE分析其分子量约为32kD,扫描分析,其表达量占菌体蛋白总量的30%左右。
     2.纯化后sTNFRⅡ的生物活性鉴定:sTNFRⅡ经western鉴定可以与TNFRⅡ的抗体结合;间接免疫荧光显示sTNFRⅡ可以抑制GFP-sTNF-α融合蛋白与细胞表面TNFR的结合;MTT实验显示20μg/ml sTNFRⅡ几乎可完全阻断sTNF-α引起的细胞毒效应。
     3.多克隆抗体的制备:用纯化的sTNFRⅡ与完全佛氏佐剂免疫两只家兔后得到抗TNFRⅡ多克隆抗体,效价分别为1:32000和1:16000。结论:(1)成功真核表达和纯化具有生物活性的融合蛋白TNFRIBP-Fc;(2)体外实验证实融合蛋白可以改善高糖和sTNF-α诱导脂肪细胞胰岛素抵抗,其作用机制主要是下调sTNF-α的表达和阻断sTNF-α与TNFRI结合介导的胰岛素抵抗;(3)两型TNF-α在胰岛素抵抗中发挥不同的作用:sTNF-α导致脂肪细胞胰岛素抵抗,TMTNF-α提高脂肪细胞对胰岛素的敏感性;(4)体内实验证实融合蛋白通过下调和抑制sTNF-α作用而改善高糖高脂饮食造成的肥胖和胰岛素抵抗。
Obesity, with an increasing prevalence, has been become the most common metabolic disorder in the world. Obesity is highly associated with insulin resistance and is the biggest risk factor for non-insulin-dependent diabetes mellitus. Insulin resistance or the decrease in insulin sensitivity, defined as a smaller than expected biological response to a given dose of insulin, preceds overt diabetes and signifies a metabolic disorder called syndrome X (concurrence of type 2 diabetes, dyslipidemias, hypertension, and cardiovascular disease).
     Tumor necrosis factor (TNF-α), produced chiefly by activated monocytes/macrophages and T lymphocytes, is a pleiotropic cytokine with a wide range of biological effects, such as anti-infection, antitumor, immune regulation and inflammation. Recently TNF-αhas been found is synthesised and secreted by adipose tissue and plays a key role in insulin resistance. The general mechanism of TNF-α-induced insulin resistance involves inhibition of insulin receptor signaling as demonstrated in variety of cell types and obese animals. TNF-αinhibits insulin-stimulated tyrosine phosphorylation of the insulin receptor and insulin receptor substrate-1 (IRS-1) at least in part by induction of serine phosphorylation of IRS-1. Although it has been previously shown that the lack of TNF-αfunction improve insulin sensitivity in obese animals, no definite conclusion about which TNFR mediates this action of TNF-α.
     TNFRI blocking peptide (TNFRIBP) was obtained by screening a phage displayed random 12 amino acid peptide library using soluble recombinant human TNFRI in our previous study. TNFRIBP was able to block TNF-αbioactivity by competitively binding to TNFRI. In order to enhance its affinity and prolong its half-life time, we inserted the oligonucleotide sequence of this peptide into the pIg/3C eukaryotic expression vector that contains an upsteam signal peptide sequence necessary for the secretion of chimeras and a downstream human IgG1-Fc fragment sequence, so we got the recombinant vector pIg/3C-TNFRIBP-Fc to secretively express TNFRIBP-Fc fusion protein. The aim of this study to establish stable cell lines to express TNFRIBP-Fc fusion protein by cotransfection CHO-K1 and BHK-21 cells with this recombinant vector and pcDNA3.1 which contains neo gene for selection, gain plenty of purified protein for treatment insulin resistance linked to obesity in vitro and in vivo. This study is expected to lay the foundaton for development of new peptide drug to treat TNF-αrelated diseases.
     1. Stable express and purify fusion protein TNFRIBP-Fc
     ⑴Establish stable cell lines: CHO-K1 and BHK-21 cells were cotransfected with pIg/3C-TNFRIBP-Fc vector and pcDNA3.1 which contained neo gene for selection, then G418 resistant transformants were selected. After subcloning through three limited-dilution , seven stable cell lines which had high productivity of fusion protein and three stable cell lines which had high productivity of Fc protein were picked out, assessed by ELISA.
     ⑵Purify fusion protein and Fc protein: We cultured these stable cell lines in quantity in the medium without serum and collected the culture supernatants. Fusion protein and Fc protein from supernatants were purified by chromatography on protein A-sepharose CL-4B. Approximately 50μg of fusion protein or 40μg of Fc protein was purified from 100ml of cultural supernatant, quantitied by a sandwich ELISA.
     ⑶Identify molecular weight by SDS-PAGE: The molecule weight of the purified protein and Fc protein were about 37kD and 36kD respectively analyzed by SDS-PAGE.
     ⑷Identify its biological activities: Indirect immunofluorescene showed that fusion protein was able to bind murine TNFRI. TNF-αbioassay suggested that fusion protein, but not Fc protein, suppressed TNF-α-induced cell death in a dose dependent manner. As low as 160ng/ml, the fusion protein completely inhibited the cytotoxicity induced by 100U/ml TNF-α.
     2. Effect of fusion protein on insulin resistance in vitro
     ⑴Induce 3T3-L1 preadipocytes to differentiate into adipocytes: Briefly, the monolayer cell were maintained in medium A (DMEM with 10% fetal calf serum). Differentiation was induced by incubating the cells with medium A supplemented with 10μg/ml insulin, 1μM dexamethasone, 0.5mM IBMX for 3 days, followed by another 2 days incubation with medium A supplemented with 10μg/ml insulin. The cells were further incubated in medium A for an additional 2 days to complete the adipocyte conversion. Followed this procedure, great than 90% of the cells had the morphological and biochemical properties of adipocytes.
     ⑵High glucose induce insulin resistance in 3T3-L1 adipocytes: 3T3-L1 adipocytes were treated with different concentration glucose for 24 hours, the insulin-stimulated 3H-2-deoxyglucose incorporation test were used to determin insulin sensitivity of 3T3-L1 cells. Results showed that 25mM glucose induced the adipocytes insulin resistance, for decreasing insulin-stimulated increase in glucose uptake by 150 times.
     ⑶The effect of the two forms of endogenous TNF-αon insulin resistance induce by high glucose: 3T3-L1 adipocytes treated with different concentration glucose for 24 hours. Results showed when the cells were induced insulin resistance by 25mM glucose, the TM TNF-αprotein level was declined from 81.4% to 3.56% (p<0.01) and sTNF-αlevel was increased from 63.7 to 113.0 pg/ml (p<0.01), compared to group with 15mM glucose. Insulin resistance could be blocked in part by 160ng/ml anti-TACE antibody or 160ng/ml TNFR1BP-Fc fusion protein, compared to group induced insulin resistance by 25mM glucose, fusion protein and anti-TACE antibody increased insulin-stimulated glucose uptake by 20 and 50 times, respectively.
     ⑷The effect of the two forms of exogenous TNF-αon reponse of 3T3-L1 adipocytes to insulin: 3T3-L1 adipocytes were treated with the two forms of exogenous TNF-αfor 24 hours and detected glucose uptake with insulin stimulation. Compared to the control group without any stimulation, sTNF-αdecreased insulin-stimulated glucose uptake by 96%, but TMTNF-αincreased two times. We alse found that fusion protein or anti-TNF-αantibody can completely block both of sTNF-αand TMTNF-αeffects when they were put together alone.
     ⑸The effect of the two forms of exogenous TNF-αon insulin signaling: 3T3-L1 adipocytes were treated with the two forms of TNF-αfor 24 hours and after that we detected the level of IRS-1 tyrosine phospharylation and serine phopharylation on 270 site (Ser270) to find out its molecular mechanism. We found that sTNF-αstimulated serine phospharylation of IRS-1 and inhibited tyrosine phopharylation, TM TNF-αacted on the contrary: TM TNF-αinhibited IRS-1 Ser270 phospharylation without effect on tyrosine phopharylation.
     3. Effect of fusion protein on insulin resistance in vivo
     ⑴Establish insulin resistance animal model: Four-week-old male wistar rats weighing between 110 and 130g were fed either with a normal diet (ND) or a high-fat and high-sucrose diet (HFS). Obesity associated insulin resistance was induced by HFS diet for 16 weeks, manifesting as increased body weight by 18%, significant hyperinsulinaemia but euglycemia maintenance and increased HOMAIR index by three times when compared with ND group.
     ⑵Treat animals: After 16 weeks, besides the ND group, the rats with HFS were randomly assigned into four treated groups: fusion protein treatment group, Fc protein treatment group, rabbit anti-TNF-αpolyclonal antibody treatment group and the HFS control group. The rats were given ingtravenoursly with 500μg TNFR1BP-Fc fusion protein or Fc protein or rabbit anti-TNF-αpolyclonal antibody in 0.1 ml normal saline (NS) or just 0.1ml NS treatmeny respectively, twice a week for four weeks.
     ⑶Inhibitory effect of the fusion protein on body weight, adiposity, serum triglyceride and total chcholesteraemia: At the end of 20 weeks, the mean body weight gain, the weight of epididymal fat pads and the level of serum triglyceride of HFS group was 2 times,1.8 and 3.7 times than that of ND group. Besides that histological analysis of adipose tissue also showed that adipocytes were larger in HFS group than in ND group. The treatment with fusion protein, but not Fc protein for 4 weeks resulted in a significantly decrease weight gain by 26%, fat pads by 30% and the triglyceride level by 50%。Similarity, histological analysis of adipose tissue also shows that adipocytes were larger in HFS fed rats than in control rats. In contrast, the treatment of fusion protein, but not Fc protein, diminished their larger diameter. All these measurements of anti-TNF-αantibody (HFS+Ab) group showed no difference between the ND group. Total cholesterol levels were unchanged in the plasma among the four group.
     ⑷The effect of the fusion protein on glucose metabolism and insulin sensitivity: The blood glucose leveles of all groups showed no difference among them and kept in normal limits, but the serum insulin, C-peptide and HOMAIR were about two folds of ND. The pancreatic islet was compensatory hypertrophy. The Insulin and glucose tolerance experiment explained that the sensitivity of insulin significantly impaired by HFS diet compared to ND group. After treatment with fusion protein, the parameter of insulin, C-peptide and HOMAIR nearly descended to 50%. The compensatory hypertrophy of pancreatic was released too. The Insulin and glucose tolerance experiment directly illustrated that the insulin sensitivity notably increased after post-treatment, but there was no effect of treatment with Fc. The insulin sensitivity of HFS+Ab group showed no difference between the ND group.
     ⑸The effect of the fusion protein on TNFαprotein level by ELISA: We detected the TNFαin serum and in the extract of tissue protein. In HFS group, the content of TNFαin muscle and adipose tissue, obviously upgraded. In fusion protein group, the content of TNFαin serum and adipose tissue downgraded to 50%, however, in muscle downgraded to 80%. In the HFS+Ab group, the content of TNFαin serum and adipose tissue downgraded to 60%, however, in muscle downgraded to 80%.There was no difference in liver tissue of all groups. The levels of TNFαin adipose tissue and muscle tissue were 2.5 and 1.4 folds than that of ND group.
     ⑹The effect of fusion protein on TNFαprotein in adipose and pancreatic tissue by immunity histochemistry: We compared the expression level of TNFαin adipose and pancreatic tissue by immunity histochemistry. Expression level of TNFαobviously increased compared to ND group and decreased after post-treatment by fusion protein.
     ⑺The effect of fusion protein on IRS-1 tyrosine phosphorylation: We tested tyrosine phosphrylation of IRS-1 by IP-Western blotting. The phosphorylation of IRS-1 was significantly inhibited in fat by 80%, muscle by 68% and liver by 32% for HFS-fed rats compared with that of ND-fed rats. Treatment with fusion protein led to a striking increase of IRS-1 phosphorylation by 54% in fat and by 50% in muscle. Treatment with antibody led to increase of IRS-1 phosphorylation by 75% in fat and by 59% in muscle.
     4. Cloning, expression and bioactivity appraisement of human sTNFRII and preparation its polyclonal antibody
     ⑴Construct and express the recombiant of human soluble TNF receptorII(sTNFRII) by E.coli: The extracellular domain cDNA of human TNFRII was obtained by RT-PCR from human monocytes. The gene fragment was inserted into pET-28a(+) and positive recombinant was identified by sequencing without mutation. Protein expression was induced by IPTG in E.coli. The expression product was isolated and purified by Ni-NTA agrose. Results showed that the highest expression was achieved after induction for 6 h with IPTG.. SDS-PAGE showed an extra protein band which was around 32kD in size, which occuppied 30% of the total protein in E.coli.
     ⑵Identify its biological activities: Western blotting showed that recombinant sTNFRII was able to bind anti-TNFRII antibody. Indirect immunofluorescene indicated that sTNFRⅡcould inhibit the binding of GFP-sTNF-αto TNFR on L929. TNF-αbioassay suggested that as low as 20μg/ml, sTNFRII completely inhibited the cytotoxicity induced by 100U/ml TNF-α.
     ⑶Prepare sTNFRII polyclonal antibody: we used recombinant sTNFRII as antigen to immune rabbit and gained polyclonal antibody. The titres of the sTNFRII specific ployclonal antibody obtained were 1:32000 and 1:16000.
     Conclusion:⑴Eukaryotic stable expresses and purifies the fusion protein TNFRIBP-Fc which it is bioactive in murine system;⑵In vitro we identify Fusion protein ameliorates insuin resistant of 3T3-L1 adipocyte induced by high glucose and sTNF-αthrough downregulation sTNF-αexpression level and inhibit their interaction between sTNF-αand TNFRI;⑶TM TNF-αand sTNF-αplay different role in insulin resistance:TM TNF-αcan improve insulin sensitivity but sTNF-αinduced insulin resistance;⑷TNFRIBP-Fc fusion protein improves obesity and insulin resistance induced by high fat and high sucrose diet and protected form the obesity-related reduction in the insulin recetor signaling in muscle and fat tissues, mainly responsible for peripheral glucose uptake with stimulation by insulin.
引文
1. Naser KA, Gruber A, Thomson GA. The emerging pandemic of obesity and diabetes: are we doing enough to prevent a disaster? Int J Clin Pract. 2006;60:1093-1097.
    2. Flegal KM, Carroll MD, Kuczmarski RJ, Johnson CL. Overweight and obesity in the United States: prevalence and trends, 1960-1994. Int J Obes Relat Metab Disord. 1998;22:39-47.
    3. Sjoholm A, Nystrom T. Inflammation and the etiology of type 2 diabetes. Diabetes Metab Res Rev. 2006;22:4-10.
    4. Lang CH, Dobrescu C, Bagby GJ. Tumor necrosis factor impairs insulin action on peripheral glucose disposal and hepatic glucose output. Endocrinology. 1992;130:43-52.
    5. Ruan H, Lodish HF. Insulin resistance in adipose tissue: direct and indirect effects of tumor necrosis factor-alpha. Cytokine Growth Factor Rev. 2003;14:447-455.
    6. Borst SE, Lee Y, Conover CF, Shek EW, Bagby GJ. Neutralization of tumor necrosis factor-alpha reverses insulin resistance in skeletal muscle but not adipose tissue. Am J Physiol Endocrinol Metab. 2004;287:E934-938.
    7. Cheung AT, Ree D, Kolls JK, Fuselier J, Coy DH, Bryer-Ash M. An in vivo model for elucidation of the mechanism of tumor necrosis factor-alpha (TNF-alpha)-induced insulin resistance: evidence for differential regulation of insulin signaling by TNF-alpha. Endocrinology. 1998;139:4928-4935.
    8. Skolnik EY, Marcusohn J. Inhibition of insulin receptor signaling by TNF: potential role in obesity and non-insulin-dependent diabetes mellitus. Cytokine Growth Factor Rev. 1996;7:161-173.
    9. Csehi SB, Mathieu S, Seifert U, et al. Tumor necrosis factor (TNF) interferes with insulin signaling through the p55 TNF receptor death domain. Biochem Biophys Res Commun. 2005;329:397-405.
    10. Feinstein R, Kanety H, Papa MZ, Lunenfeld B, Karasik A. Tumor necrosisfactor-alpha suppresses insulin-induced tyrosine phosphorylation of insulin receptor and its substrates. J Biol Chem. 1993;268:26055-26058.
    11. Tanti JF, Gremeaux T, van Obberghen E, Le Marchand-Brustel Y. Serine/threonine phosphorylation of insulin receptor substrate 1 modulates insulin receptor signaling. J Biol Chem. 1994;269:6051-6057.
    12. Chang I, Kim S, Kim JY, et al. Nuclear factor kappaB protects pancreatic beta-cells from tumor necrosis factor-alpha-mediated apoptosis. Diabetes. 2003;52:1169-1175.
    13. Lewis M, Tartaglia L, Lee A, et al. Cloning and Expression of cDNAs for Two Distinct Murine Tumor Necrosis Factor Receptors Demonstrate One Receptor is Species Specific. PNAS. 1991;88:2830-2834.
    14. Rohl M, Pasparakis M, Baudler S, et al. Conditional disruption of IkappaB kinase 2 fails to prevent obesity-induced insulin resistance. J Clin Invest. 2004;113:474-481.
    15. Rosenzweig T, Braiman L, Bak A, Alt A, Kuroki T, Sampson SR. Differential effects of tumor necrosis factor-alpha on protein kinase C isoforms alpha and delta mediate inhibition of insulin receptor signaling. Diabetes. 2002;51:1921-1930.
    16. de Alvaro C, Teruel T, Hernandez R, Lorenzo M. Tumor necrosis factor alpha produces insulin resistance in skeletal muscle by activation of inhibitor kappaB kinase in a p38 MAPK-dependent manner. J Biol Chem. 2004;279:17070-17078.
    17. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science. 1996;271:665-668.
    18. Schinner S, Scherbaum WA, Bornstein SR, Barthel A. Molecular mechanisms of insulin resistance. Diabet Med. 2005;22:674-682.
    19. Xu H, Sethi JK, Hotamisligil GS. Transmembrane tumor necrosis factor (TNF)-alpha inhibits adipocyte differentiation by selectively activating TNF receptor 1. J Biol Chem. Vol. 274; 1999:26287-26295.
    20. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection fromobesity-induced insulin resistance in mice lacking TNF-alpha function. Nature. 1997;389:610-614.
    21. Schreyer SA, Chua SC, Jr., LeBoeuf RC. Obesity and diabetes in TNF-alpha receptor- deficient mice. J Clin Invest. 1998;102:402-411.
    22. Lofgren P, van Harmelen V, Reynisdottir S, et al. Secretion of tumor necrosis factor-alpha shows a strong relationship to insulin-stimulated glucose transport in human adipose tissue. Diabetes. 2000;49:688-692.
    23. Hotamisligil GS, Spiegelman BM. Tumor necrosis factor alpha: a key component of the obesity-diabetes link. Diabetes. 1994;43:1271-1278.
    24. Hofmann C, Lorenz K, Braithwaite SS, et al. Altered gene expression for tumor necrosis factor-alpha and its receptors during drug and dietary modulation of insulin resistance. Endocrinology. 1994;134:264-270.
    25. Dominguez H, Storgaard H, Rask-Madsen C, et al. Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res. 2005;42:517-525.
    26. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259:87-91.
    27. Hotamisligil GS, Budavari A, Murray D, Spiegelman BM. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-alpha. J Clin Invest. 1994;94:1543-1549.
    28. Ofei F, Hurel S, Newkirk J, Sopwith M, Taylor R. Effects of an engineered human anti-TNF-alpha antibody (CDP571) on insulin sensitivity and glycemic control in patients with NIDDM. Diabetes. 1996;45:881-885.
    29. Hube F, Hauner H. The two tumor necrosis factor receptors mediate opposite effects on differentiation and glucose metabolism in human adipocytes in primary culture. Endocrinology. 2000;141:2582-2588.
    30. Xu H, Hotamisligil GS. Signaling pathways utilized by tumor necrosis factor receptor 1 in adipocytes to suppress differentiation. FEBS Lett. 2001;506:97-102.
    31. Tartaglia LA, Pennica D, Goeddel DV. Ligand passing: the 75-kDa tumor necrosis factor (TNF) receptor recruits TNF for signaling by the 55-kDa TNF receptor. J Biol Chem. 1993;268:18542-18548.
    32. Uysal KT, Wiesbrock SM, Hotamisligil GS. Functional analysis of tumor necrosis factor (TNF) receptors in TNF-alpha-mediated insulin resistance in genetic obesity. Endocrinology. 1998;139:4832-4838.
    33. Sethi JK, Xu H, Uysal KT, Wiesbrock SM, Scheja L, Hotamisligil GS. Characterisation of receptor-specific TNFalpha functions in adipocyte cell lines lacking type 1 and 2 TNF receptors. FEBS Lett. 2000;469:77-82.
    34. Hotamisligil GS, Arner P, Atkinson RL, Spiegelman BM. Differential regulation of the p80 tumor necrosis factor receptor in human obesity and insulin resistance. Diabetes. 1997;46:451-455.
    35. Bailey CJ. Treating insulin resistance in type 2 diabetes with metformin and thiazolidinediones. Diabetes Obes Metab. 2005;7:675-691.
    36. Grover JK, Vats V, Yadav SS. Pterocarpus marsupium extract (Vijayasar) prevented the alteration in metabolic patterns induced in the normal rat by feeding an adequate diet containing fructose as sole carbohydrate. Diabetes Obes Metab. 2005;7:414-420.
    37. Paquot N, Castillo MJ, Lefebvre PJ, Scheen AJ. No increased insulin sensitivity after a single intravenous administration of a recombinant human tumor necrosis factor receptor: Fc fusion protein in obese insulin-resistant patients. J Clin Endocrinol Metab. 2000;85:1316-1319.
    38. Takeshita S, Kawamura I, Yasuno T, et al. Amelioration of insulin resistance in diabetic ob/ob mice by a new type of orally active insulin-mimetic vanadyl complex: bis(1-oxy-2-pyridinethiolato)oxovanadium(IV) with VO(S(2)O(2)) coordination mode. J Inorg Biochem. 2001;85:179-186.
    39. del Aguila LF, Claffey KP, Kirwan JP. TNF-alpha impairs insulin signaling and insulin stimulation of glucose uptake in C2C12 muscle cells. Am J Physiol. 1999;276:E849-855.
    40. Mei J, Wang CN, O'Brien L, Brindley DN. Cell-permeable ceramides increasebasal glucose incorporation into triacylglycerols but decrease the stimulation by insulin in 3T3-L1 adipocytes. Int J Obes Relat Metab Disord. 2003;27:31-39.
    41. Nolte LA, Hansen PA, Chen MM, Schluter JM, Gulve EA, Holloszy JO. Short-term exposure to tumor necrosis factor-alpha does not affect insulin-stimulated glucose uptake in skeletal muscle. Diabetes. 1998;47:721-726.
    42. Hohmeier HE, Newgard CB. Cell lines derived from pancreatic islets. Mol Cell Endocrinol. 2004;228:121-128.
    43. Dickson LM, Lingohr MK, McCuaig J, et al. Differential activation of protein kinase B and p70(S6)K by glucose and insulin-like growth factor 1 in pancreatic beta-cells (INS-1). J Biol Chem. 2001;276:21110-21120.
    44. Parkash J, Chaudhry MA, Rhoten WB. Tumor necrosis factor-alpha-induced changes in insulin-producing beta-cells. Anat Rec A Discov Mol Cell Evol Biol. 2005;286:982-993.
    45. Kharroubi I, Ladriere L, Cardozo AK, Dogusan Z, Cnop M, Eizirik DL. Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-kappaB and endoplasmic reticulum stress. Endocrinology. 2004;145:5087-5096.
    46. Kim HS, Kim S, Lee MS. IFN-gamma sensitizes MIN6N8 insulinoma cells to TNF-alpha-induced apoptosis by inhibiting NF-kappaB-mediated XIAP upregulation. Biochem Biophys Res Commun. 2005;336:847-853.
    47. Marshall S, Bacote V, Traxinger RR. Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. J Biol Chem. 1991;266:4706-4712.
    48. Hribal ML, Perego L, Lovari S, et al. Chronic hyperglycemia impairs insulin secretion by affecting insulin receptor expression, splicing, and signaling in RIN beta cell line and human islets of Langerhans. Faseb J. 2003;17:1340-1342.
    49. Sykiotis GP, Papavassiliou AG. Serine phosphorylation of insulin receptor substrate-1: a novel target for the reversal of insulin resistance. Mol Endocrinol. 2001;15:1864-1869.
    50. Kanety H, Feinstein R, Papa MZ, Hemi R, Karasik A. Tumor necrosis factor alpha-induced phosphorylation of insulin receptor substrate-1 (IRS-1). Possible mechanism for suppression of insulin-stimulated tyrosine phosphorylation of IRS-1. J Biol Chem. 1995;270:23780-23784.
    51. Kroder G, Bossenmaier B, Kellerer M, et al. Tumor necrosis factor-alpha- and hyperglycemia-induced insulin resistance. Evidence for different mechanisms and different effects on insulin signaling. J Clin Invest. 1996;97:1471-1477.
    52. Liu LS, Spelleken M, Rohrig K, Hauner H, Eckel J. Tumor necrosis factor-alpha acutely inhibits insulin signaling in human adipocytes: implication of the p80 tumor necrosis factor receptor. Diabetes. 1998;47:515-522.
    53. Wrede CE, Dickson LM, Lingohr MK, Briaud I, Rhodes CJ. Fatty acid and phorbol ester-mediated interference of mitogenic signaling via novel protein kinase C isoforms in pancreatic beta-cells (INS-1). J Mol Endocrinol. 2003;30:271-286.
    54. Morimoto Y, Nishikawa K, Ohashi M. KB-R7785, a novel matrix metalloproteinase inhibitor, exerts its antidiabetic effect by inhibiting tumor necrosis factor-alpha production. Life Sci. 1997;61:795-803.
    55. Voros G, Maquoi E, Collen D, Lijnen HR. Influence of membrane-bound tumor necrosis factor (TNF)-alpha on obesity and glucose metabolism. J Thromb Haemost. 2004;2:507-513.
    56. Kirchner S, Holler E, Haffner S, Andreesen R, Eissner G. Effect of different tumor necrosis factor (TNF) reactive agents on reverse signaling of membrane integrated TNF in monocytes. Cytokine. 2004;28:67-74.
    57. Peraldi P, Hotamisligil GS, Buurman WA, White MF, Spiegelman BM. Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase. J Biol Chem. 1996;271:13018-13022.
    58. Warzocha K, Bienvenu J, Coiffier B, Salles G. Mechanisms of action of the tumor necrosis factor and lymphotoxin ligand-receptor system. Eur Cytokine Netw. 1995;6:83-96.
    59. Youngren JF, Paik J, Barnard RJ. Impaired insulin-receptor autophosphorylation is an early defect in fat-fed, insulin-resistant rats. J Appl Physiol. 2001;91:2240-2247.
    60. Yu Y, Ohmori K, Chen Y, et al. Effects of pravastatin on progression of glucose intolerance and cardiovascular remodeling in a type II diabetes model. J Am Coll Cardiol. 2004;44:904-913.
    61. van den Brandt J, Kovacs P, Kloting I. Metabolic features in disease-resistant as well as in spontaneously hypertensive rats and newly established obese Wistar Ottawa Karlsburg inbred rats. Int J Obes Relat Metab Disord. 2000;24:1618-1622.
    62. Zhao C, Wang P, Xiao X, et al. Gene therapy with human tissue kallikrein reduces hypertension and hyperinsulinemia in fructose-induced hypertensive rats. Hypertension. 2003;42:1026-1033.
    63. Yoshioka S, Uemura K, Tamaya N, et al. Dietary fat-induced increase in blood pressure and insulin resistance in rats. J Hypertens. 2000;18:1857-1864.
    64. Sandu O, Song K, Cai W, Zheng F, Uribarri J, Vlassara H. Insulin resistance and type 2 diabetes in high-fat-fed mice are linked to high glycotoxin intake. Diabetes. 2005;54:2314-2319.
    65. Bullo M, Garcia-Lorda P, Megias I, Salas-Salvado J. Systemic inflammation, adipose tissue tumor necrosis factor, and leptin expression. Obes Res. 2003;11:525-531.
    1. Mokdad AH, Serdula MK, Dietz WH, Bowman BA, Marks JS, Koplan JP. The spread of the obesity epidemic in the United States, 1991-1998. Jama. 1999;282:1519-1522.
    2. Naser KA, Gruber A, Thomson GA. The emerging pandemic of obesity and diabetes: are we doing enough to prevent a disaster? Int J Clin Pract. 2006;60:1093-1097.
    3. Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM. Prevalence of overweight and obesity in the United States, 1999-2004. Jama. 2006;295:1549-1555.
    4. Flegal KM, Carroll MD, Kuczmarski RJ, Johnson CL. Overweight and obesity in the United States: prevalence and trends, 1960-1994. Int J Obes Relat Metab Disord. 1998;22:39-47.
    5. Troiano RP, Flegal KM, Kuczmarski RJ, Campbell SM, Johnson CL. Overweight prevalence and trends for children and adolescents. The National Health andNutrition Examination Surveys, 1963 to 1991. Arch Pediatr Adolesc Med. 1995;149:1085-1091.
    6. Warne JP. Tumour necrosis factor alpha: a key regulator of adipose tissue mass. J Endocrinol. 2003;177:351-355.
    7. Sjoholm A, Nystrom T. Inflammation and the etiology of type 2 diabetes. Diabetes Metab Res Rev. 2006;22:4-10.
    8. Lang CH, Dobrescu C, Bagby GJ. Tumor necrosis factor impairs insulin action on peripheral glucose disposal and hepatic glucose output. Endocrinology. 1992;130:43-52.
    9. Marette A. Molecular mechanisms of inflammation in obesity-linked insulin resistance. Int J Obes Relat Metab Disord. 2003;27 Suppl 3:S46-48.
    10. Schinner S, Scherbaum WA, Bornstein SR, Barthel A. Molecular mechanisms of insulin resistance. Diabet Med. 2005;22:674-682.
    11. Hotamisligil GS, Spiegelman BM. Tumor necrosis factor alpha: a key component of the obesity-diabetes link. Diabetes. 1994;43:1271-1278.
    12. Skolnik EY, Marcusohn J. Inhibition of insulin receptor signaling by TNF: potential role in obesity and non-insulin-dependent diabetes mellitus. Cytokine Growth Factor Rev. 1996;7:161-173.
    13. Miles PD, Romeo OM, Higo K, Cohen A, Rafaat K, Olefsky JM. TNF-alpha-induced insulin resistance in vivo and its prevention by troglitazone. Diabetes. 1997;46:1678-1683.
    14. Dandona P, Weinstock R, Thusu K, Abdel-Rahman E, Aljada A, Wadden T. Tumor necrosis factor-alpha in sera of obese patients: fall with weight loss. J Clin Endocrinol Metab. 1998;83:2907-2910.
    15. Lofgren P, van Harmelen V, Reynisdottir S, et al. Secretion of tumor necrosis factor-alpha shows a strong relationship to insulin-stimulated glucose transport in human adipose tissue. Diabetes. 2000;49:688-692.
    16. Xu H, Hirosumi J, Uysal KT, Guler AD, Hotamisligil GS. Exclusive action of transmembrane TNF alpha in adipose tissue leads to reduced adipose mass and local but not systemic insulin resistance. Endocrinology. 2002;143:1502-1511.
    17. Plomgaard P, Bouzakri K, Krogh-Madsen R, Mittendorfer B, Zierath JR, Pedersen BK. Tumor necrosis factor-alpha induces skeletal muscle insulin resistance in healthy human subjects via inhibition of Akt substrate 160 phosphorylation. Diabetes. 2005;54:2939-2945.
    18. Machann J, Haring H, Schick F, Stumvoll M. Intramyocellular lipids and insulin resistance. Diabetes Obes Metab. 2004;6:239-248.
    19. del Aguila LF, Claffey KP, Kirwan JP. TNF-alpha impairs insulin signaling and insulin stimulation of glucose uptake in C2C12 muscle cells. Am J Physiol. 1999;276:E849-855.
    20. Tessier JP, Thurner B, Jungling E, Luckhoff A, Fischer Y. Impairment of glucose metabolism in hearts from rats treated with endotoxin. Cardiovasc Res. 2003;60:119-130.
    21. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259:87-91.
    22. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature. 1997;389:610-614.
    23. Ruan H, Lodish HF. Insulin resistance in adipose tissue: direct and indirect effects of tumor necrosis factor-alpha. Cytokine Growth Factor Rev. 2003;14:447-455.
    24. Chen L, Xie Z, Teng Y, et al. Highly efficient selection of the stable clones expressing antibody-IL-2 fusion protein by a dicistronic expression vector containing a mutant neo gene. J Immunol Methods. 2004;295:49-56.
    25. Patiag D, Gray S, Idris I, Donnelly R. Effects of tumour necrosis factor-alpha and inhibition of protein kinase C on glucose uptake in L6 myoblasts. Clin Sci (Lond). 2000;99:303-307.
    26. Gao Z, Zuberi A, Quon MJ, Dong Z, Ye J. Aspirin inhibits serine phosphorylation of insulin receptor substrate 1 in tumor necrosis factor-treated cells through targeting multiple serine kinases. J Biol Chem.2003;278:24944-24950.
    27. Feinstein R, Kanety H, Papa MZ, Lunenfeld B, Karasik A. Tumor necrosis factor-alpha suppresses insulin-induced tyrosine phosphorylation of insulin receptor and its substrates. J Biol Chem. 1993;268:26055-26058.
    28. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science. 1996;271:665-668.
    29. Rosenzweig T, Braiman L, Bak A, Alt A, Kuroki T, Sampson SR. Differential effects of tumor necrosis factor-alpha on protein kinase C isoforms alpha and delta mediate inhibition of insulin receptor signaling. Diabetes. 2002;51:1921-1930.
    30. Mei J, Wang CN, O'Brien L, Brindley DN. Cell-permeable ceramides increase basal glucose incorporation into triacylglycerols but decrease the stimulation by insulin in 3T3-L1 adipocytes. Int J Obes Relat Metab Disord. 2003;27:31-39.
    31. Peraldi P, Hotamisligil GS, Buurman WA, White MF, Spiegelman BM. Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase. J Biol Chem. 1996;271:13018-13022.
    32. Ruan H, Hacohen N, Golub TR, Van Parijs L, Lodish HF. Tumor necrosis factor-alpha suppresses adipocyte-specific genes and activates expression of preadipocyte genes in 3T3-L1 adipocytes: nuclear factor-kappaB activation by TNF-alpha is obligatory. Diabetes. 2002;51:1319-1336.
    33. Xu H, Sethi JK, Hotamisligil GS. Transmembrane tumor necrosis factor (TNF)-alpha inhibits adipocyte differentiation by selectively activating TNF receptor 1. J Biol Chem. Vol. 274; 1999:26287-26295.
    34. Winder WW, Hardie DG. AMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes. Am J Physiol. 1999;277:E1-10.
    35. Moller DE, Berger JP. Role of PPARs in the regulation of obesity-related insulin sensitivity and inflammation. Int J Obes Relat Metab Disord. 2003;27 Suppl 3:S17-21.
    36. Shepherd PR, Kahn BB. Glucose transporters and insulin action--implications for insulin resistance and diabetes mellitus. N Engl J Med. 1999;341:248-257.
    37. Stephens JM, Lee J, Pilch PF. 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;272:971-976.
    38. Asensio C, Muzzin P, Rohner-Jeanrenaud F. Role of glucocorticoids in the physiopathology of excessive fat deposition and insulin resistance. Int J Obes Relat Metab Disord. 2004;28 Suppl 4:S45-52.
    39. Bjorntorp P, Holm G, Rosmond R. Hypothalamic arousal, insulin resistance and Type 2 diabetes mellitus. Diabet Med. 1999;16:373-383.
    40. Ruan H, Miles PD, Ladd CM, et al. Profiling gene transcription in vivo reveals adipose tissue as an immediate target of tumor necrosis factor-alpha: implications for insulin resistance. Diabetes. 2002;51:3176-3188.
    41. Shibasaki M, Takahashi K, Itou T, et al. Alterations of insulin sensitivity by the implantation of 3T3-L1 cells in nude mice. A role for TNF-alpha? Diabetologia. 2002;45:518-526.
    42. Shojima N, Sakoda H, Ogihara T, et al. Humoral regulation of resistin expression in 3T3-L1 and mouse adipose cells. Diabetes. 2002;51:1737-1744.
    43. Hube F, Hauner H. The two tumor necrosis factor receptors mediate opposite effects on differentiation and glucose metabolism in human adipocytes in primary culture. Endocrinology. 2000;141:2582-2588.
    44. Sethi JK, Xu H, Uysal KT, Wiesbrock SM, Scheja L, Hotamisligil GS. Characterisation of receptor-specific TNFalpha functions in adipocyte cell lines lacking type 1 and 2 TNF receptors. FEBS Lett. 2000;469:77-82.
    45. Uysal KT, Wiesbrock SM, Hotamisligil GS. Functional analysis of tumor necrosis factor (TNF) receptors in TNF-alpha-mediated insulin resistance in genetic obesity. Endocrinology. 1998;139:4832-4838.
    46. Hotamisligil GS, Arner P, Atkinson RL, Spiegelman BM. Differential regulation of the p80 tumor necrosis factor receptor in human obesity and insulin resistance.Diabetes. 1997;46:451-455.
    47. Tartaglia LA, Pennica D, Goeddel DV. Ligand passing: the 75-kDa tumor necrosis factor (TNF) receptor recruits TNF for signaling by the 55-kDa TNF receptor. J Biol Chem. 1993;268:18542-18548.
    48. Nakai Y, Hamagaki S, Takagi R, Taniguchi A, Kurimoto F. Plasma concentrations of tumor necrosis factor-alpha (TNF-alpha) and soluble TNF receptors in patients with bulimia nervosa. Clin Endocrinol (Oxf). 2000;53:383-388.
    49. Liu LS, Spelleken M, Rohrig K, Hauner H, Eckel J. Tumor necrosis factor-alpha acutely inhibits insulin signaling in human adipocytes: implication of the p80 tumor necrosis factor receptor. Diabetes. 1998;47:515-522.
    50. Schreyer SA, Chua SC, Jr., LeBoeuf RC. Obesity and diabetes in TNF-alpha receptor- deficient mice. J Clin Invest. 1998;102:402-411.
    51. Hotamisligil GS, Budavari A, Murray D, Spiegelman BM. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-alpha. J Clin Invest. 1994;94:1543-1549.
    52. Paquot N, Castillo MJ, Lefebvre PJ, Scheen AJ. No increased insulin sensitivity after a single intravenous administration of a recombinant human tumor necrosis factor receptor: Fc fusion protein in obese insulin-resistant patients. J Clin Endocrinol Metab. 2000;85:1316-1319.
    53. Dominguez H, Storgaard H, Rask-Madsen C, et al. Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res. 2005;42:517-525.
    54. Ofei F, Hurel S, Newkirk J, Sopwith M, Taylor R. Effects of an engineered human anti-TNF-alpha antibody (CDP571) on insulin sensitivity and glycemic control in patients with NIDDM. Diabetes. 1996;45:881-885.
    55. Bailey CJ. Treating insulin resistance in type 2 diabetes with metformin and thiazolidinediones. Diabetes Obes Metab. 2005;7:675-691.
    56. Peraldi P, Spiegelman B. TNF-alpha and insulin resistance: summary and future prospects. Mol Cell Biochem. 1998;182:169-175.

© 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