用户名: 密码: 验证码:
IUGR幼鼠IR机制及营养干预作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:
     探讨IUGR幼鼠胰腺组织增殖和凋亡,肝脏mTOR/S6K通路及肝糖异生关键酶的变化与幼鼠IR发生的关系及营养干预的影响。
     方法:
     1.采用孕鼠妊娠期全程低蛋白饮食的方法建立IUGR幼鼠模型。通过控制每只母鼠喂哺幼鼠的个数,达到早期营养干预的目的。通过对3周龄和7周龄幼鼠进行腹腔葡萄糖耐量试验评价胰岛素敏感性。
     2.应用Real-time PCR方法测定IUGR幼鼠出生时,1周龄,3周龄和7周龄胰腺组织中增殖指标PDX-1和凋亡指标caspase-3,bax和bcl-2mRNA表达。
     3.应用Western Blot方法测定IUGR幼鼠出生时,3周龄和7周龄肝脏组织中mTOR/S6K,IRS-1以及p-IRS-1的蛋白表达。
     4.应用Real-time PCR方法测定IUGR幼鼠出生时,1周龄,3周龄和7周龄肝脏组织中PGC-1α和糖异生关键酶PEPCK和G-6-Pase mRNA表达。
     5.数据采用SPSS10.0软件进行统计学分析。
     结果:
     1.妊娠期全程低蛋白饮食母鼠(LPM)在妊娠第8天,第15天和分娩前体重均明显低于对照组(CM),P<0.01。并且LPM体重呈现先降低后升高的态势,而CM体重在整个妊娠期均呈增高趋势。
     2.宫内发育迟缓(IUGR)幼鼠出生时体重,鼻臀长和尾长都低于对照组(C),P<0.001。1周龄时,宫内发育迟缓未加强营养(IUGR-UN)幼鼠体重,鼻臀长和尾长均低于C,P<0.001。而宫内发育迟缓加强营养(IUGR-N)幼鼠体重已经达到对照组水平,P值为0.279;但是鼻臀长和尾长还落后于C,P值均为0.000。IUGR-N在体重,鼻臀长和尾长均高于IUGR-UN,P<0.01。3周龄时,IUGR-UN体重和鼻臀长明显低于C,P均值为0.000,但是尾长与C的差异没有统计学意义,P值为0.173。IUGR-N体重,鼻臀长和尾长不但超过了IUGR-UN,而且超过了C,P值均为0.000。7周龄时,IUGR-UN体重,鼻臀长和尾长都达到与C相同的水平,P>0.05;IUGR-N体重超过了C和IUGR-UN,P<0.05。但是鼻臀长和尾长在3组间是没有差异的,P>0.05。
     3.3周龄时,3组幼鼠的空腹血.糖(FPG)和腹腔糖耐量120分钟血糖(G120)水平没有差异,P>0.05。IUGR-N空腹胰岛素(PINS)120分钟胰岛素(I120)水平均明显高于C和IUGR-N,P<0.01。但是C和IUGR-UN胰岛素水平没有差异,P>0.05。7周龄时,IUGR-N FPG高于C,P值为0.022。IUGR-UN和IUGR-N G120均高于C组,P<0.05。但是这2组之间G120水平的差异没有统计学意义,P>0.05。IUGR-UN和IUGR-N PINS和I120均明显高于C,P<0.001。同时,IUGR-N FINS和I120水平亦明显高于IUGR-UN,P<0.001。
     4.3周龄时,IUGR-N HOMA-IR明显高于C组和IUGR-UN组,P值分别为0.000和0.000,IUGR-NU组和C组幼鼠的HOMA-IR差异没有统计学意义,P值为0.708。IUGR-N HOMA-IS明显低于C和IUGR-UN,P值分别为0.033和0.028,但C和IUGR-UN间差异没有统计学意义,P值为0.9111。IUGR-UN HOMA-β与C和IUGR-N的差异均存在统计学意义,P值分别为0.022和0.004。C组和IUGR-N组的差异没有统计学意义,P值为0.192。7周龄时,IUGR-UN和IUGR-N两组的HOMA-IR均明显高于C,P值均为0.000,这两组间的差异亦存在统计学意义,P值为0.006。IUGR-UN和IUGR-N两组幼鼠的HOMA-IS均明显低于C组,P值均为0.000,这两组间的差异也存在统计学意义,P值为0.016。IUGR-UN和IUGR-N两组HOMA-β均明显低于C组,P值均为0.000,这两组间的差异无统计学意义,P值为0.717。
     5.出生时,IUGR PDX-1mRNA表达低于C,P<0.05。1周龄时,3组幼鼠PDX-1mRNA表达差异无统计学意义,P>0.05。3周龄时,IUGR-N PDX-1mRNA表达高于C组和IUGR-UN,P<0.05。C和IUGR-UN间的差异没有统计学意义,P>0.05。7周龄时IUGR2组PDX-1mRNA表达明显高于C,P<0.05。IUGR-UN2组间的差异没有统计学意义,P<0.05。
     6.出生时,IUGR caspase-3mRNA表达高于C,P<0.05。1周龄时,3组幼鼠caspase-3mRNA表达的差异没有统计学意义,P>0.05。3周龄时,IUGR2组caspase-3mRNA表达均高于C,P<0.05。此2组间的差异没有统计学意义,P>0.05。7周龄时,IUGR2组caspase-3mRNA表达仍均高于C,P<0.05;IUGR-N caspase-3mRNA的表达明显高于IUGR-UN,P<0.05。
     7.出生时,IUGR bcl-2mRNA表达低于C,P<0.01。1周龄时,3组幼鼠bcl-2mRNA表达的差异没有统计学意义,P>0.05。3周龄时,IUGR2组bcl-2mRNA表达均低于C,P<0.05。此2组间的差异没有统计学意义。7周龄时IUGR2组bcl-2mRNA表达均低于C,P<0.05;此2组间的差异没有统计学意义。
     8.出生时,IUGR bax mRNA表达高于C,P<0.05。但是IUGR2组间bax mRNA表达的差异没有统计学意义。1周龄时,IUGR2组幼鼠bax mRNA表达均高于C组,P<0.05。IUGR-Nbax mRNA表达高于IUGR-UN,P值为0.000。3周龄时,IUGR2组bax mRNA表达均高于C,P值均为0.000。此2组间的差异没有统计学意义。7周龄时IUGR2组bax mRNA的表达均高于C组,P值分别为0.001和0.000;此2组间的差异没有统计学意义。
     9.出生时,IUGRmTOR, S6K, IRS-1和p-IRS-1ser636蛋白表达低于C,,P值分别为0.000,0.003,0.023和0.007。3周龄时,IUGR-UN和IUGR-N2组幼鼠mTOR和p-IRS-lser636的蛋白表达超过C组,P为0.000和0.000及0.019和0.012,虽然mTOR蛋白表达在IUGR2组间没有差异,P值为0.120;但是,p-IRS-1ser636蛋白表达在2组间有差异,P值为0.032。同时,S6K和IRS-1蛋白表达水平在3组间没有差异。7周龄时,IUGR mTOR,S6K和p-IRS-1ser636蛋白表达均超过C,P<0.05。但是IRS-1的蛋白表达在3组间没有差异。
     10.出生时,IUGR PGC-1α mRNA, G-6-Pase mRNA和PEPCK mRNA基因表达均高于C,P<0.05。1周龄时,3周龄时和7周龄时,IUGR PGC-la mRNA, G-6-Pase mRNA和PEPCK mRNA基因表达均高于C组,P<0.01。同时,IUGR-N PGC-la mRNA, G-6-Pase mRNA和PEPCK mRNA基因表达均高于IUGR-UN, P<0.01。
     结论:
     1.应用孕鼠妊娠期全程低蛋白饮食的方法,可以成功建立IUGR幼鼠的模型。
     2.对于IUGR幼鼠,哺乳期营养的加强只是加快了追赶生长的速度,加速了肥胖的出现,对于性成熟期前的最终身高没有影响。而且哺乳期营养的加强会造成性成熟期前IR的出现和血糖的异常。而正常喂养方式可以延缓IR和血糖异常的发生,但在性成熟期前,仍会造成糖代谢的异常。这或许是IUGR婴儿成年后2型糖尿病发病率增高的一个原因。因此对于具有IUGR病史的婴儿,应该尽早监测糖代谢的情况;阻止哺乳期婴儿快速体重增加或许可以预防儿童时期体重的增加。
     3.孕母妊娠期营养不良会使IUGR幼鼠出生时胰腺组织凋亡增加,增殖降低。营养干预影响IUGR幼鼠胰腺的凋亡和增殖。在生命早期,bax首先触发凋亡过程,哺乳期给予适当的营养干预,抑制凋亡基因的表达,增加胰腺增殖,是改善IUGR儿童今后糖代谢的关键。
     4.宫内不良的营养环境造成肝脏糖异生关键酶的表达增加,而这种增加持续至出生后,支持节俭表型假说,同时是IUGR幼鼠成年时期发生2型糖尿病的基础。
     5.营养是一柄双刃剑,通过激活mTOR/S6K途径,一方面促进机体的生长发育,另一方面造成IR,合理平衡营养,或许是改善IUGR婴儿成年预后的关键。对于mTOR/S6K途径的干预或许可以成为治疗IR的新靶点。
Objective:
     To study the relationships between insulin resistance of juvenile rats with intrauterine growth retardation and changes of pancreatic apoptosis and proliferation, mTOR/S6K pathway in the liver and key enzymes of gluconeogenesis and effect of nutritional intervention.
     Methods:
     1To establish the juvenile rats with intrauterine growth retardation model through low protein diet of whole pregnant process. To limit the number of juvenile rats in order to give early nutritional intervention. To evaluate insulin sensitivity of juvenile rats with intrauterine growth at3weeks and7weeks through intraperitoneal glucose tolerance test.
     2To measure PDX-1, caspase-3, bax and bcl-2mRNA expression of pancreatic tissue of IUGR juvenile rats by real time PCR at birthday,1week,3weeks and7weeks.
     3To measure mTOR/S6K, IRS-1and p-IRS-1protein expression of liver of IUGR juvenile rats by Western Blot at birth,3weeks and7weeks.
     4To measure PGC-1α, and PEPCK and G-6-Pase, which are the key enzymes of gluconeogeneis, mRNA expression of liver of IUGR juvenile rats by real time PCR at bithday,1week,3weeks and7weeks.
     5To statistical analyze the data with SPSS software10.0.
     Results:
     1The weights of mother rats with low protein diet of whole pregnant process(LPM) were lower than those of control mother(CM) at the8days,15days and before delivery during pregnance(p<0.01). The weights of LPM became lighter firstly and then heavier, hower the weights of CM became heavier and heavier during whole pregnance.
     2The birthweight, nose-hip length and tail length of intrauterine growth retardation (IUGR) juvenile rats were lower than those of control group (C)(p<0.001). At1week, the weight, nose-hip length and tail length of intrauterine growth retardation of un-nutrition (IUGR-UN) juvenile rats were lower than those of C (p<0.01). However, the weight of intrauterine growth retardation of nutrition (IUGR-N) juvenile rats was as same as that of C (the value of P was0.219). At the same time, nose-hip length and tail length of IUGR-N were lower than those of C(the values of P were0.000, respectively). The weight, nose-hip length and tail length of IUGR-N were higher than those of IUGR-UN(p<0.01). At3weeks, the weight and nose-hip length of IUGR-UN were lower than those of C(the values of P were0.000, respectively). The tail length of IUGR-UN was as same as that of C(the value of P was0.173). The weight, nose-hip length and tail length of IUGR-N were higher than those of both IUGR-UN and C(the value of P were0.000, respectively). At7weeks, the weight, nose-hip length and tail length of IUGR-UN were as same as those of C (P>0.05) and the weight of IUGR-N was heavier than that of C and IUGR-UN (P<0.05, respectively). However, there were no differences of nose-hip length and tail length between C, IUGR-UN and IUGR-N (P>0.05).
     3There were on differences of the levels of fast plasma glucose(FPG) and intraperitoneal glucose tolerance test120minute glucose(G120) between three groups at3weeks(P>0.05). At the same time, the concentrations of fast insulin(FINS) and intraperitoneal glucose tolerance test120minute insulin(I120) of IUGR-N were significantly higher than those of C and IUGR-UN(.P<0.01). However, there was no difference between C and IUGR-UN. At7weeks, the concentration of FPG in IUGR-N was higher than that of C, the value of P was0.022. The levels of G120in IUGR-UN and IUGR-N were all higher than that of C(P<0.05). There was no difference of G120between IUGR-UN and UGR-N. The levels of FINS and1120in IUGR-UN and IUGR-N were higher significantly than those of C. The concentrations of FINS and1120in IUGR-N were all higher than those of IUGR-UN.
     4At3threes, HOMA-IR of IUGR-N was significantly higher than that of C and IUGR-UN, the value of P were0.000and0.000, respectively. There was no difference between IUGR-UN and C, the value of P was0.708. HOMA-IS of IUGR-N was lower than that of C and IUGR-UN, the values of P were0.033and0.028, respectively. Hower there was no difference between C and IUGR-UN. There were differences of HOMA-β between IUGR-UN and C and between IUGR-UN and IUGR-N, the value of P were0.022and0.004, respectively. There was no difference between C and IUGR-N. At7weeks, HOMA-IR of IUGR-UN and IUGR-N were all higher than that of C, the value of P were all0.000. At same time, there was difference between IUGR-UN and IUGR-N, the value of P was0.006. HOMA-IS and HOMA-β of IUGR-UN and IUGR-N were all lower than those of C. The levels of HOMA-IS in IUGR-N was lower than that of IUGR-UN.
     5The level of PDX-1mRNA expression in IUGR pancreatic tissue was lower than that of C at birthday. There were no differences in PDX-1mRNA expression between three groups at1week. The level of PDX-1mRNA expression in IUGR-N was higher than that of C and IUGR-UN (P<0.05) at three weeks. There was no difference in PDX-1mRNA expression between C and IUGR-UN. The levels of caspase-3mRNA expression in IUGR were higher than that of C (P<0.05) at7weeks. There was no significant difference between IUGR-UN and IUGR-N.
     6The level of caspase-3mRNA expression in IUGR pancreatic tissue was higher than that of C at birthday. There were no differences in PDX-1mRNA expression between three groups at1week. The levels of PDX-1mRNA expression in IUGR-N and IUGR-UN were higher than that of C (P<0.05) at three weeks. There was no difference in caspase-3mRNA expression between IUGR-N and IUGR-UN. The level of caspase-3mRNA expression in IUGR were higher than that of C(P <0.05) at7weeks. The level of caspase-3mRNA expression in IUGR-N was higher than that of IUGR-UN(P<0.05).
     7The levels of bcl-2mRNA expression in IUGR pancreatic tissue was lower than that of C at birthday. There were no differences in bcl-2mRNA expression between three groups at1week. The levels of bcl-2mRNA expression in IUGR-N and IUGR-UN were lower than that of C (P<0.05) at three weeks. There was no difference in bcl-2mRNA expression between IUGR-N and IUGR-UN. The level of bcl-2mRNA expression in IUGR were lower than that of C(P<0.05) at7weeks. There was no significant difference between IUGR-UN and IUGR-N.
     8The levels of bax mRNA expression in IUGR pancreatic tissue was higher than that of C at birthday. The levels of bax mRNA expression in IUGR pancreatic tissue were higher than that of C at1week. The levels of bax mRNA expression in IUGR-N and IUGR-UN were higher than that of C (the value of P was0.000) at three weeks. There was no difference in bax mRNA expression between IUGR-N and IUGR-UN. The level of bax mRNA expression in IUGR were higher than that of C(P<0.05) at7weeks. There was no significant difference between IUGR-UN and IUGR-N.
     9The levels of mTOR, S6K, IRS-1and p-IRS-1ser636protein expressions in IUGR were lower than those of C at birthday. There were no differences in S6K and IRS-1protein expressions between C and IUGR at3weeks. The levels of mTOR and p-IRS-1ser636protein expressions in IUGR were higher than those of C at3weeks. The levels of mTOR, S6K and p-IRS-1ser636protein expressions in IUGR-UN and IUGR-N were higher than those of C at7weeks, but there was no difference in IRS-1protein expressions among these three groups.
     10The levels of PGC-1a mRNA, G-6-Pase mRNA and PEPCK mRNA expressions in IUGR were higher than those of C at birthday. The levels of PGC-1α mRNA, G-6-Pase mRNA and PEPCK mRNA expressions in IUGR were still higher than those of C at1week,3weeks and7weeks. The levels of PGC-la mRNA, G-6-Pase mRNA and PEPCK mRNA expressions in IUGR-N were higher than those of IUGR-UN at1week,3weeks and7weeks, too. Conclusions
     1We can establish intrauterine growth retardation juvenile rats model successfully through low protein diet during whole pregnant process.
     2For intrauterine growth juvenile rats, nutritional enhancement only accelerates speed of the catch-up growth and obesity appearance during the lactation. Nutritional enhancement will not influence the terminal height before sex maturity. However, nutritional enhancement during the lactation will lead to insulin resistance and abnormal blood glucose before sex maturity. Usual diet will postpone the insulin resistance and abnormal blood glucose, but at last, it will lead to abnormal glucose metabolism. Maybe it is the reason that leads to higher incidence of type2diabetes for IUGR infants. So the glucose metabolism condition should be monitored earlier.
     3The low protein diet during pregnance will lead to both apoptosis increase and proliferation decrease of pancreatic tissue for IUGR juvenile rat at birth. In the early life, bax triggers the apoptosis process firstly. It is the key point to improve glucose metabolism for children with IUGR in the future through suppressing apoptosis gene expression and increasing proliferation gene expression.
     4The malnutrition condition in intrauterine leads to key enzyme of gluconeogenesis expression increasing, which will last until after birth. This manifestation supports the thrift hypothesis, which is the basis that IUGR juvenile rats suffer from type2diabetes during adult period.
     5Nutrition has effect on IUGR juvenile rats by mTOR/S6K pathway with two sides. One side, it can promote growth and development, on the other side, it leads to insulin resistance. So, nutrition balance may be a key point to improve prognosis for IUGR infants. The mTOR/S6K pathway will be a new target for treatment of insulin resistance.
引文
[1]Barker DJP and Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales[J].Lancet,1986,327(5):1077-1081.
    [2]Hales CN, Barker DJ, Clark PM, et al. Fetal and infant growth and impaired glucose tolerance at age 64[J].BMJ,1991,303:1019-1022.
    [3]Barker DJ, Hales CN, Fall CH, et al. Type2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X):relation to reduced fetal growth[J]. Diabetologia,1993,36:62-67.
    [4]Barker DJ. Fetal origins of coronary heart disease[J]. BMJ,1995,311:171-174
    [5]Ravelli G, Stein ZA, and Susser MW. Obesity in young men after famine exposure in utero and early infancy[J]. N Engl J Med,1995,295:349-354.
    [6]Rich-Edwards JW, Colditz GA, Stampfer MJ, et al.Birthweight and the risk for type 2 diabetes mellitus in adult women[J]. Ann Intern Med,1999,130:278-284.
    [7]Valdez R, Athens MA, Thompson GH, et al. Birthweight and adult health outcomes in a biethnic population in the USA[J]. Diabetologia,1994,37:624-631.
    [8]Parker L, Lamont DW, Unwin N, et al. A lifecourse study of riskfor hyperinsulinaemia, dyslipidaemia and obesity (the central metabolic syndrome)at age 49-51 years[J]. Diabet Med,2003,20:406-415.
    [9]赵文华,陈春明,翟屹等.生命早期营养不良对成年后超重和肥胖患病危险影响的研究[J].中华流行病学杂志,2006,27(8)
    [10]Barker DJ, Eriksson JG, Forsen T, et al. Fetal origins of adult disease:strength of effects and biological basis[J]. Int J Epidemiol,2002,31:1235-1239.
    [11]Thamotharan M, Shin BC, Suddirikku DT, et al. GLUT4 expression and subcellular localization in the intrauterine growth-restricted adult rat female offspring[J]. Am J Physiol Endocrinol Metab,2005,288:E935-E947.
    [12]Jansson N, Pettersson J, Haafiz A, et al. Down-regulation of placental transport of amino acids precedes the development of intrauterine growth restriction in rats fed a low protein diet[J]. J Physiol,2006,576:935-946.
    [13]Vuguin P, Raab E, Liu B, et al. Hepatic insulin resistance precedes the development of diabetes in a model of intrauterine growth retardation[J]. Diabetes 2008,53:2617-2622.
    [14]Bassi JA, Rosso P, Moessinger AC, et al. Fetal growth retardation due to maternal tobacco smoke exposure in the rat[J]. Pediatr Res,1984,18:127-130.
    [15]Chen L, Nyomba BL. Effects of prenatal alcohol exposure on glucose tolerance in the rat offspring[J]. Metab,2009,52:454-462.
    [16]Barry JS, Davidsen ML, Limesand SW, et al. Developmental changes in ovine myocardial glucose transporters and insulin signaling following hyperthermia-induced intrauterine fetal growth restriction[J]. Exp Biol Med (Maywood),2006,231:566-575.
    [17]de R Sr, Painter RC, Roseboom TJ, et al. Glucose tolerance at age 58 and the decline of glucose tolerance in comparison with age 50 in people prenatally exposed to the Dutch famine[J]. Diabetologia,2006,49:637-643.
    [18]de R Sr, Painter RC, Phillips DI, et al. Impaired insulin secretion after prenatal exposure to the Dutch famine[J]. Diabetes Care 2006;29:1897-1901.
    [19]Painter RC, de R Sr, Bossuyt PM, et al. Blood pressure response to psychological stressors in adults after prenatal exposure to the Dutch famine[J].. J Hypertens,2009,24:1771-1778.
    [20]Stein AD, Zybert PA, van der Pal-de Bruin, et al. Exposure to famine during gestation, size at birth, and blood pressure at age 59 y:evidence from the Dutch Famine[J]. Eur J Epidemiol,2006,21:759-765.
    [21]Painter RC, de R Sr, Bossuyt PM, et al. Early onset of coronary artery disease after prenatal exposure to the Dutch famine[J]. Am J Clin Nutr,2009,84:322-327.
    [22]Stanner SA, Bulmer K, Andres C, et al. Does malnutrition in utero determine diabetes and coronary heart disease in adulthood? Results from the Leningrad siege study, a cross sectional study[J]. BMJ,1997,315:1342-1348.
    [23]王谦,应艳琴,宁琴等.宫内发育迟缓幼鼠胰岛素受体底物表达与胰岛素抵抗[J].中华内分泌代谢杂志,2007;23(4).
    [24]李瑶,辛颖.胰岛素受体底物2在宫内发育迟缓大鼠胰腺组织中的表达[J].中国中西医结合儿科学,2009,1(2)
    [25]Yan-ping Xu, Li Liang, Xiu-Min Wang. The levels of Pdx1/insulin, Cacnalc and Cacnald, and β-cell mass in a rat model of intrauterine undernutrition[J]. Journal of Maternal-Fetal and Neonatal Medicine,2010,24(3):437-443.
    [26]孙贺,韩萍,董玉梅等.不同剂量GLP-1对宫内发育迟缓幼鼠胰岛p细胞增殖和凋亡的影响.中华医学会第九次全国内分泌学学术会议论文汇编.
    [27]Khamzina L, Veilleux A, Marette A, er al. Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: possible involvement in obesity-linked insulin resistance[J]. Endocrinology,2009, 146(3):1473-1481.
    [28]Um SH, Frigerio F, Watanabe M, et al. Absence of S6K1 protects against age-and diet-induced obesity while enhancing insulin resistance[J]. Nature, 2004,431 (7007):200-205.
    [29]苑红,牛彦媚,刘彦辉等mTOR/S6K1信号通路在高脂饮食诱导小鼠胰岛素抵抗发生中的作用[J].中国糖尿病杂志,2009,12.
    [30]陈显,柳青,雷寒等.炎症应激加重CD36基因敲除小鼠胰岛素抵抗[J].第三军医大学学报,2010,32(9)
    [31]Moller DE. New drug targets for type 2 diabetes and the metabolic syndrome[J]. Nature,2010; 414:821-827.
    [32]赵惠中,萧建中,杨文英,等.肝脏胰岛素抵抗与肝糖输出调控基因表达的关系[J].中华肝脏病杂志,2006,14:45-48.
    [33]Goto M, Yoshioka T, Battelino T,er al. TNFalpha decreases gluconeogenesis in hepatocytes isolated from 10-day-old rats[J]. Pediatr Res,2010,49:552-557.
    [34]Miao J, Fang S, Bae Y, Kemper JK. Functionalinhibitory cross-talk between constitutive androstane receptor and hepatic nuclear factor-4 in hepatic lipid/glucose metabolism is mediated by competition for binding to the DR1 motif and to the common coactivators, GRIP-1 and PGC-lalpha[J].. JBiol Chem,2006,281: 14537-14546.
    [35]Yoon JC, Puigserver P, Chen G, et al. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1[J].. Nature 2001,413:131-138.
    [36]Burgess SC, Leone TC, Wende AR, et al. Diminished hepatic gluconeogenesis via defects in tricarboxylic acid cycle flux in peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1 alpha)-deficient mice[J]. J Biol Chem 2006,281:19000-19008.
    [37]Robert H, Nicolek M, Jennifer L, et al. Increased Hepatic Peroxisome Proliferator-Activated Receptor-Coactivator-1 Gene Expression in a Rat Model of Intrauterine Growth Retardation and Subsequent Insulin Resistance[J].. Endocrinology,2008,143(7):2486-2490.
    [38]Simmons RA, Templeton LJ, Gertz SJ. Intrauterine growth retardation leads to the development of type 2 diabetes in the rat[J]. Diabetes,2001,50(10):2279-86.
    [39]Shrader RE,Zeman FJ. Effect of maternal protein deprivation on morphological and enzymatic development of neonatal rat tissue[J]. J Nutr,1969,99:401-421.
    [40]Terry H,Mary ED. A rat model for the study of intrauterine growth retardation[J]. AmJ Obstet Gynecol,1988,158:1203.
    [41]Vogt E, Ng AK, Rote NS. A model for the antiphospholipid antibody syndrome:monoclonal antiphosphatidylserine antibody induces intrauterine growth restriction in mice[J]. Am J Obstet Gynecol,1996,174:700-707.
    [42]Rockwell LC,Keyes LE,Moore LG. Chronic hypoxia diminishes pregnancy associated DNA synthesis in guinea pig uteroplacental arteries[J]. Placenta, 2000,21:313-319.
    [43]Hohmann M and Kunzel W. Dihydroergotamine causes fetal growth retardation in guinea pigs[J]. Arch Gynecol Obstet,1992,251:187-192.
    [44]Bassan H,Trejo LL,Kariv N,et al. Experimental intrauterine growth retardation alters renal development [J]. Pediatr Nephrol,2000,15:192-195.
    [45]Guo W, Gregg JR, Fonkalsrud EW. Effect of maternal ethanol intake on fetal rabbit gastrointestinal development[J].J Pediatr Surg,1994,29:1030-1033.
    [46]张秀泉,严隽鸿,洪素英,等.被动吸烟对胎兔组织细胞周期改变的影响[J]..中华妇产科杂志,1993,28:720.
    [47]Greenwood PL, Slepetis RM, Hermanson JW, et al. Intrauterine growth retardation is associated with reduced cell cycle activity, but not myofibre number, in ovine fetal muscle[J]. Reprod Fertil Dev,1999,11:281-291.
    [48]Phillips ID,Anthony RV, Simonetta G, et al. Restriction of fetal growth has a differential impact on fetal prolactin and prolactin receptor mRNA expression[J]. J Neuro,2001,13:175-181.
    [49]Tarantal A F, Martha ML, Gargosky SE, et al. Effects of viral virulence on intrautering growth in SIV2infected fetal rhesus macaques (Macaca mulatta) [J]. J Acqu Imm Defi Syn Hum Retrovirol,1995,10:129-138.
    [50]Hein PR, Schatorje JS, Frencken HJ, et al. The effect of chronic oral methadone treatment on monkey chorionic gonadotropin, estradiol, dehydroepiandrosterone sulfate, progesterone, prolactin and cortisol levels during pregnancy in the cynomolgus monkey (Macaca fascicularis) [J]. Eur J Obstet Gynecol Reprod Biol,1991,38:145-150.
    [51]Kliegman RM. Alterations of fasting glucose and fat metabolism in intrauterine growth retarded newborn dogs[J]. Am J Physiol,1989,256:E380-385.
    [52]Wu G, Pond WG, Flynn SR, et al.Maternal dietary protein deficiency decreases nitric oxide synthase and ornithine decarboxylase activities in placenta and endometrium of pigs during early gestation[J]. J Nutr,1998,128:2395-2402.
    [53]Xu RJ,Mellor DJ,Birtles MJ,et al. Impact of intrauterine growth retardation on the gastrointestinal tract and the pancreas in newborn pigs[J]. J Pediatr Gastroenterol Nutr,1994,18:231-240.
    [54]Bauer R,Walter B,Zwiener U. Comparison between inulin clearance and endogenous creatinine clearance in newborn normal weight and growth restricted newborn piglets[J]. Exp Toxicolog Pathol,2008,52:367-372.
    [55]刘静波,姚英,于冰等.叶酸对初产母猪繁殖性能和宫内发育迟缓仔猪肾脏功能基因表达的影响[J].动物营养学报2010,22.
    [56]周根平,王怡,霍永久等.胎儿宫内发育迟缓对新生仔猪小肠组织发育的影响[J].动物营养学报,2006,18(1)
    [57]Stephanie R. Thorn,Timothy R. H. Regnault, Laura D. Brown, et al.Intrauterine growth restriction increases fetal hepatic gluconeogenic capacity and reduces messenger ribonucleic acid translation initiation and nutrient sensing in fetal liver and skeletal muscle endocrinology[J].2009,150(7):3021-3030.
    [58]Miles J. De Blasio, Kathryn L. Gatford, I. Caroline McMillen, et al Placental Restriction of Fetal Growth Increases Insulin Action, Growth, and Adiposity in the Young Lamb[J]. Endocrinology,2007,148(3):1350-1358.
    [59]Robinson JS, Kingston EJ, Jones CT,et al. Studies on experimental growth retardation in sheep:The effect of removal of a endometrial caruncles on fetal size and metabolism[J]. J Dev Physiol,1979,1:379-398.
    [60]Yasaman Shahkhalili, Julie Moulin, Irene Zbinden, et al.Comparison of two models of intrauterine growth restriction for early catch-up growth and later development of glucose intolerance and obesity in rats[J]. Am J Physiol Regul Integr Comp Physiol,2010,298:R141-R146.
    [61]Neitzke U, Harder T, Schellong K, et al. Intrauterine growth restriction in a rodent model and developmental programming of the metabolic syndrome:a critical appraisal of the experimental evidence[J].2008,29(3):246-54.
    [62]陈径,徐虹,沈茜等.宫内发育迟缓对大鼠血压及肾功能影响[J].中华肾脏病杂志,2006,11.
    [63]ROBERT H. LANE, NICOLE K. MACLENNAN, JENNIFER L. HSU, et al. Increased hepatic peroxisome proliferator-activated receptor-_ coactivator-1 gene expression in a rat model of intrauterine growth retardation and subsequent Insulin Resistance[J]. Endocrinology,2009,143:2486-2490.
    [64]Huang TT, Qiu XS, DU ML, et al.Expression of tumor necrosis factor alpha mRNA in adipose cell of intrauterine growth retarded rats and its relation to insulin resistance[J].Zhonghua Er Ke Za Zhi.,2005,43(1):39-43.
    [65]Desai M, Gayle DA, Jooby B, et al. Programmed obesity in intrauterine growth restricted newborns:modulation by newborn nutrition[J]. Am J Physiol Regul Integr Comp Physiol,2005,288:R91-R96.
    [66]Ken K L Ong, a Marion L Ahmed, Michael A Preece, et al. Association between postnatal catch-up growth and obesity in childhood:prospective cohort study[J]. BMJ,2000,320:967-971
    [67]MARY PYLIPOW, LOGAN G. SPECTOR, SUSAN E. PUUMALA, et al. Early postnatal weight gain, intellectual performance, and body mass index at 7 years of age in term infants with intrauterine growth restriction[J]. Pediatr,2009,154:201-6.
    [68]Krochik AG, Chaler EA, Maceiras M, et al. Presence of early risk markers of metabolic syndrome in prepubertal children with a history of intrauterine growth restriction[J]. Arch Argent Pediatr.,2010,108(1):10-6.
    [69]Limesand SW, Jensen J, Hutton JC, et al. Diminished β-cell replication contributes to reduced β-cell mass in fetal sheep with intrauterine growth restriction β[J]. Am J Physiol Regul Integr Comp Physiol,2005,288, R1297-R1305.
    [70]Limesand SW, Rozance PJ, Zerbe GO, et al. Attenuated insulin release and storage in fetal sheep pancreatic islets with intrauterine growth restriction[J]. Endocrinology,2006147:1488-1497.
    [71]De Blasio MJ, Gatford KL, McMillen IC, Robinson JS, Owens Jaet al. Placental restriction of fetal growth increases insulin action, growth, and adiposity in the young lamb[J]. Endocrinology,2007,148:1350-1358.
    [72]Kum Kum S. Bhasin, Atila van Nas, Lisa J. Martin, et al. Maternal low-protein diet or hypercholesterolemia reduces circulating essential amino acids and leads to intrauterine growth restriction[J].DIABETES,2009,58(3):559-566.
    [73]Arends MY, Wyllie AH. Apoptosis:mechanisms and roles n pathology[J]. Int Rev Exp Pathol,1991,32:223-254.
    [74]Pirot P, Cardozo AK, Eizirik DL.Mediators and mechanisms of pancreatic (3-cell death in type 1 diabetes[J]. Arq Bras Endocrinol Metabol,2008,52:156-165.
    [75]Gluckman, P.D. and Hanson, M.A. Livi ng with the past:evolution, development, and patterns of disease[J]. Science,2008,305,1733-1736.
    [76]Tsujimoto, Y., Finger, L.S., Yunis, J., et al.Cloning of the chromosome breakpoint of neoplastic B-cells with the t(14;18) chromosome translocation[J]. Science,1984,226:1097-1099.
    [77]Tracey H. Wagner, Anne M. Drewry, Sandra MacMillan, et al. Surviving sepsis: bcl-2 overexpression modulates splenocyte transcriptional responses in vivo[J]. Am J Physiol Regul Integr Comp Physiol,2007,292:R1751-R1759.
    [78]Thornberry NA, Bull HG, Calaycay JR, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes[J]. Nature.,1992,30:768-74.
    [79]Scatena R, Bottom P, Botta G, et al. The role of mitochondria in pharmaco toxicology:a reevaluation of an old, newly emerging topic[J]. Am J Physiol Cell Physiol,2007,293:C12-21.
    [80]Hacker G, Paschen SA. Therapeutic targets in the mitochondrial apoptotic pathway[J]. Expert Opin Ther Targets,2007,11:515-526.
    [81]Cryns V, Yuan J. Protease to die for[J]. Genes Dev,1998,12(11):1551-1570.
    [82]Sarah Ferber, Amir Halkin, Hofit Cohen, et al. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nature Medicine[J].2000,6(5):568-572.
    [83]Satsuki Miyazaki, Eiji Yamato, Jun-ichi Miyazaki. Regulated expression of pdx-1 promotes in vitro differentiation of insulin-producing cells from embryonic stem cells[J]. Diabetes,2008,53(4):1030-1037.
    [84]孙吉平,杨于嘉,王晓莉等.胰腺十二指肠同源框-1基因促进大鼠骨髓间充质干细胞分化为胰岛样细胞[J].中国科学c辑生命科学,2006,36(2):171-179.
    [85]Hideto Kojima, Takaaki Nakamura, Yukihiro Fujita. Combined expression of pancreatic duodenal homeobox 1 and islet factor 1 induces immature enterocytes to produce insulin[J]. Diabetes,2008,51(5):1398-1408.
    [86]NicoliniU, Hubinont C, Santolaya J, Fisk NM, and Rodeck CH. Effects of fetal intravenous glucose challenge in normal and growth-retarded fetuses[J]. Horm Metab Res,1990,22:426-430.
    [87]DahriS, Snoeck A, Reusens-Billen B,et al. Islet function in offspring of mothers on low-protein diet during gestation[J]. Diabetes,1991, Suppl 2:115-120. [88] BarkerDJP. Mother, Babies, and Health in Later Life. Edinburgh:Churchill Livingstone,1998.
    [89]Van AsscheFA, De Prins F, Aerts L, et al. The endocrine pancreas in small-for-dates infants[J]. Br J Obstet Gynaecol,1977,84:751-753.
    [90]GarofanoA, Czernichow P, and Breant B. Beta-cell mass and proliferation following late fetal and early postnatal malnutrition in the rat[J]. Diabetologia, 1998,41:1114-1120.
    [91]J. Petrik, B. Reusens, E. Arany, C, et al. A Low Protein Diet Alters the Balance of Islet Cell Replication and Apoptosis in the Fetal and Neonatal Rat and Is Associated with a Reduced Pancreatic Expression of Insulin-Like Growth Factor-II[J]. Endocrinology,1999,140:10 4861-4873.
    [92]Scaglia L, Cahill CJ, Finegood DT, et al. Apoptosis participates in the remodeling of the endocrine pancreas in the neonatal rat[J]. Endocrinology, 1997,138:1736-1741.
    [93]Bonner-Weir S:Perspective:postnatal pancreatic beta cell growth[J]. Endocrinology,2000,141:1926-1929.
    [94]Xu G, Stoffers DA, Habener JF,et al. Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats[J]. Diabetes,1999,48:2270-2276.
    [95]Stoffers DA, Kieffer TJ, Hussain MA, et al. Insulinotropic glucagon-like peptide 1 agonists stimulate expression of homeodomain protein IDX-1 and increase islet size in mouse pancreas[J]. Diabetes,2000,49:741-748.
    [96]A. Stoffers, Biva M. Desai, Diva D. DeLeon, et al. Neonatal Exendin-4 Prevents the Development of Diabetes in the Intrauterine Growth Retarded Rat[J]. Diabetes,2008,52:734-740.
    [97]Saltiel AR, Kahn CR.Insulin signaling and the regulation of glucose and lipid metabolism[J].Nature,2001,414(6865):799-806.
    [98]Tsuruzoe K, Emkey R, Kriauciunas KM, et al.Insulin receptor substrate-3(IRS-3)and IRS-4 impare IRS-1 and IRS-2 mediated signaling[J].Mol Cell Biol, 2001,21(1):26-38.
    [99]Woodgett JR, Cohen P.Multisite phosphorylation of glycogen synthase:molecular basis for the substrate specificity of glycogen synthase kinase-3 and casein kinase-II (glycogen synthase kinase-5) [J].Biochim Biophys Acta,1984,788(3): 339-347.
    [100]Zheng Y, Collins SL, Lutz MA, et al. A role for mammalian targetof rapamycin in regulating T cell activation versus anergy[J]. J Immunol,2007,178:2163-2170.
    [101]Peponi E, Drakos E, Reyes G, et al. Activation of mammalian target of rapamycin signaling promotes cell cycle progression and protects cells from apoptosis in mantle cell lymphoma[J]. Am J Pathol,2006,169:2171-2180.
    [102]Pene F, Claessens YE, Muller O, et al. Role of the phosphatidylinositol 3-kinase/Akt and mTOR/P70S6-kinase pathways in the proliferation and apoptosis in multiple myeloma[J]. Oncogene,2002; 21,6587-6597.
    [103]Wang X, Proud CG. The mTOR pathway in the control of protein synthesis[J]. Physiology (Bethesda),2006,21:362-369.
    [104]Sarbassov D D, Guertin D A, Ali S M, et al. Phosphory-lation and regulation of Akt/PKB by the rictor-mTOR complex[J]. Science,2005,307(5712):1098-1101.
    [105]Sarbassov D.D., Ali S.M., Kim D.H., et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton[J]. Curr. Biol.2004,14,1296-1302.
    [106]Sarbassov D.D., Ali S.M., Sengupta S., et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB[J]. Mol. Cell,2006,22,159-168.
    [107]Wullschleger S., Loewith R. and Hall M.N. TOR signaling in growth and metabolism[J]. Cell,2006,124,471-484.
    [108]Cota D., Proulx K., Smith K.A., et al. Hypothalamic mTOR signaling regulates food intake[J].Science,2006,312,927-930.
    [109]Changhua Wang, Xuming Mao, Lixin Wang, et al.Adiponectin Sensitizes Insulin Signaling by Reducing p70 S6 Kinase-mediated Serine Phosphorylation of IRS-1[J]. Biological Chemistry,2007,282(11) 7991-7996.
    [110]Tzatsos, A., and Kandror, K. V. Nutrients suppress phosphatidylinositol 3-kinase/Akt signaling via raptor-dependent mTOR-mediated insulin receptor substrate 1 phosphorylation[J]. Mol. Cell. Biol,2006 26,63-76.
    [11]Ueno M., Carvalheira J. B., Tambascia R. C., et al. Regulation of insulin signalling by hyperinsulinaemia:role of IRS-1/2 serine phosphorylation and the mTOR/p70 S6K pathway[J]. Diabetologia,2005,48,506-518.
    [112]Tremblay, F., Gagnon, A., Veilleux, A., et al. Activation of the mammalian target of rapamycin pathway acutely inhibits insulin signaling to Akt and glucose transport in 3T3-L1 and human adipocytes[J].Endocrinology,2005,146,1328-1337.
    [113]Berg C. E., Lavan B. E., and Rondinone C. M. Biochem. Rapamycin partially prevents insulin resistance induced by chronic insulin treatment[J]. Biophys. Res. Commun,2009,293,1021-1027.
    [114]Um S. H., D'Alessio, D., and Thomas G. Nutrient overload, insulin resistance, and ribosomal protein S6 kinase 1, S6K1[J]. Cell Metab,2006,3, 393-402.
    [115]Um S. H., Frigerio F., Watanabe M., et al. Normalizing a hyperactive mTOR initiates muscle growth during obesity [J]. Nature,2004,431,200-205.
    [116]Liang H, Balas B, Tantiwong P, et al.Whole body overexpression of PGC-1 alpha has opposite efffects on hepatic and muscle insulin sensitivity[J]. Am J Physiol Endocrinol Metab,2009,296:E945-E954.
    [117]Leone TC, Lehman JJ, Finck Bn, et al. PGC-1 alpha deficiency causes multi-system energy metabolic derangements:Muscle dysfunction, abnormal weight control and hepatic steatosis[J]. Plos Biol,2005,3:e101.
    [118]Goto M, Yoshioka T, Battelino T,et al. TNFalpha decreases gluconeogenesis in hepatocytes isolated from 10-day-old rats[J]. Pediatr Res,2001,49:552-557.
    [119]Ropelle ER, Pauli JR, Cintra DE, et al. Acute exercise modulaes the Foxol/PGC-1 alpha pathway in the liver of diet-induced obesity rats[J]. J Physiol. 2009,587:2069-2076.
    [120]Li X, Monks B,Ge Q, et al. Akt/PKB regulates hepatic metabolism by directly inhibiting PGC-1 alpha transcripton coactivator[J]. Nature,2007,447:1012-1016.
    [121]Stephanie R. Thorn, Timothy R. H. Regnault, Laura D. Brown, et al. Intrauterine growth restriction increases fetal hepatic gluconeogenic capacity and reduces messenger ribonucleic acid translation initiation and nutrient sensing in fetal liver and skeletal muscle[J]. Endocrinology,2009,150(7):3021-3030.
    [122]Simmons RA, Templeton LJ, Gertz SJ. Intrauterine growth retardation leads to the development of type 2 diabetes in the rat[J]. Diabetes,2001,50:2279-2286.
    [123]Khamzina L, Veilleux A, Bergeron S, et al. Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: possible involvement in obesity-linked insulin resistance[J]. Endocrinology,2005, 146:1473-81.
    [124]Korsheninnikova E, van der Zon GC, Voshol PJ, et al. Sustained activation of the mammalian target of rapamycin nutrient sensing pathway is associated with hepatic insulin resistance, but not with steatosis, in mice[J]. Diabetologia,2006,49: 3049-57.
    [125]Tremblay F, Jacques H and Marette A. Modulation of insulin action by dietary proteins and amino acids:role of the mammalian target of rapamycin nutrient sensing pathway[J]. Curr Opin Clin Nutr Metab Care,2005,8:457-62.
    [126]Tremblay F and Marette A. Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway. A negative feedback mechanism leading to insulin resistance in skeletal muscle cells[J]. J Biol Chem,2001,276:38052-60.
    [127]Tremblay F, Krebs M, Dombrowski L, et al. Overactivation of S6 kinase 1 as a cause of human insulin resistance during increased amino acid availability [J]. Diabetes,2005,54:2674-84.
    [128]Tremblay F, Gagnon A, Veilleux A, et al. Activation of the mammalian target of rapamycin pathway acutely inhibits insulin signaling to Akt and glucose transport in 3T3-L1 and human adipocytes[J]. Endocrinology,2005,146:1328-37.
    [129]Chen Xinlong,Xia Zhaofan, Ben Duofeng, et al.mTOR partly mediates insulin resistance by phosphorylation of insulin receptor substrate-1 on serine307 residues after burn[J].Burns,2011,37(1):86-93.
    1 Barker DJ, Hales CN, Fall CH, et al. Type 2 (non-insulin-dependent) diabetes mellitus, hypertensionand hyperlipidaemia (syndrome X):relation to reduced fetal growth[J]. Diabetologia 1993; 36:62-67.
    2 Barker DJ. Fetal origins of coronary heart disease. BMJ 1995;311:171-174.
    3 Hales CN, Barker DJ, Clark PM, et al. Fetal and infant growth and impaired glucose toleranceat age 64[J]. BMJ 1991; 303:1019-1022.
    4 Singhal A, Cole TJ, Lucas A. Early nutrition in preterm infants and later blood pressure:two cohorts after randomised trials[J]. Lancet 2001; 357:413-419.
    5 Singhal A, Fewtrell M, Cole TJ, et al. Low nutrient intake and early growth for later insulin resistance in adolescents born preterm[J]. Lancet 2003; 361:1089-1097.
    6 Singhal A, Cole TJ, Fewtrell M, et al. Breastmilk feedingand lipoprotein profile in of a prospective randomised study[J]. Lancet 2004; 363:1571-1578.
    7 Cianfarani S,Germani D, Branca F. Low birthweight and adult insulin resistance: the "catch-up growth" hypothesis[J].Arch Dis Child Fetal Neonatal Ed 1999; 81: F71-F73.
    8 Gluckman PD, Hanson MA. Living with the past:evolution, development, and patterns of disease[J]. Science 2004; 305:1733-1736.
    9 Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus:the thrifty phenotype hypothesis[J]. Diabetologial992; 35:595-601.
    10 Hales CN, Barker DJ. The thrifty phenotype hypothesis[J]. BrMed Bull 2001; 60: 5-20.
    11 Lucas A. Programming by early nutrition:an experimental approach[J]. J Nutr 1998; 128:401S-406S.
    12 Singhal A, Lucas A. Early origins of cardiovascular disease:is there a unifying hypothesis? [J] Lancet 2004; 363:1642-1645.
    13 Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet 1986; 1:1077-1081.
    14 Curhan GC, Willett WC. Rimm EB, et al. Birth weight and adult hypertension, diabetes mellitus, and obesity in US men. Circulation 1996; 94:3246-3250.
    15 Lithell HO, McKeigue PM, Berglund L, et al. Relation of size at birth to non-insulin dependent diabetes and insulin concentrations in men aged 50-60 years[J]. BMJ 1996; 312:406-410.
    16 Valdez R, Athens MA, Thompson GH, et al. Birthweight and adult health outcomes in a biethnic population in the USA. Diabetologia 1994; 37:624-631.
    17 Cook JT, Levy JC, Page RC,et al. Association of low birth weight with beta cell function in the adult first degree relatives of non-insulin dependent diabetic subjects[J]. BMJ 1993; 306:302-306.
    18 Khan N, Couper JJ. Low-birth-weight infants show earlier onset of IDDM[J]. Diabetes Care 1994; 17:653-656.
    19 Persson B, Pschera H, Binder C,et al. Decreased beta-cell function in women with previous small for gestational age infants[J]. Horm Metab Res 1993; 25: 170-174.
    20 Huxley RR, Shi ell AW, Law CM. The role of size at birth and postnatal catch-up growth in determining systolic blood pressure:a systematic review of the literature[J]. J Hypertens 2000; 18:815-831.
    21 Law CM, Shiell AW. Is blood pressure inversely related to birth weight? The strength of evidence from a systematic review of the literature[J]. J Hypertens 1996; 14:935-941.
    22 Tappy L. Adiposity in children born small for gestational age[J]. Int J Obes 2006; 30:S36-S40.
    23 Roseboom T, de Rooij S, Painter R. The Dutch famine and its long-term consequences for adult health[J]. Early Hum Dev 2006; 82:485-491.
    24 de Rooij SR, Painter RC, Phillips DI, et al. Impaired insulin secretion after prenatal exposure to the Dutch famine[J].Diabetes Care 2006; 29:1897-1901.
    25 Yajnik CS, Deshpande SS, Jackson AA, et al. Vitamin B12 and folate concentrations during pregnancy and insulin resistance in the offspring:the Pune Maternal Nutrition Study[J].Diabetologia 2008; 51:29-38.
    26 Desai M, Crowther NJ, Ozanne SE, et al.Adult glucose and lipid metabolism may be programmedduring fetal life[J]. Biochem Soc Trans 1995; 23:331-335.
    27 Lauren L, Jarvelin MR, Elliott P. Relationship between birthweight and blood lipid concentrations in later life:evidence from the existing literature[J]. Int J Epidemiol 2003; 32:862-876.
    28 Martyn CN, Meade TW, Stirling Y, et al. Plasma concentrations of fibrinogen and factor Ⅶ in adult life and their relation to intra-uterine growth[J]. Br J Haematol 1995; 89:142-146.
    29 McKeigue PM, Lithell HO, Leon DA. Glucose tolerance and resistance to insulin-stimulated glucose uptake in men aged 70 years in relation to size at birth[J]. Diabetologia 1998; 41:1133-1138.
    30 Woods LL. Fetal origins of adult hypertension:a renal mechanism? [J]Curr Opin Nephrol Hypertens 2000; 9:419-425.
    31 Barker DJP. Mothers, babies and diseases in later life. London:BMJ Publishing Group 1994.
    32 Lindsay RS, Lindsay RM, Edwards CR, et al. Inhibition of 11-beta-hydroxysteroid dehydrogenase in pregnant rats and the programming of blood pressure in the offspring[J].Hypertension 1996; 27:1200-1204.
    33 Benediktsson R, Lindsay RS, Noble J, et al. Glucocorticoid exposure in utero:new model for adult hypertension [J]. Lancet 1993; 341:339-341.
    34 Dahri S, Snoeck A, Reusens-Billen B, et al. Islet function in offspring of mothers on low-protein diet during gestation[J]. Diabetes 1991; 40 Suppl 2:115-120
    35 Holemans K, Gerber R, Meurrens K, et al. Maternal food restriction in the second half of pregnancy affects vascular function but not blood pressure of rat female offspring[J]. Br J Nutr 1999; 81:73-79.
    36 Woodall SM, Johnston BM, Breier BH, et al. Chronic maternal undernutrition in the rat leads to delayed postnatal growth and elevated blood pressure of offspring[J]. Pediatr Res 1996; 40:438-443.
    37 Langley-Evans SC. Critical differences between two low protein diet protocols in the programming of hypertension in the rat. Int J Food Sci Nutr 2000; 51:11-17.
    38 Bloomfield FH, Oliver MH, Giannoulias CD, et al. Brief undernutrition in late-gestation sheep programs the hypothalamic-pituitary-adrenal axis in adult offspring[J]. Endocrinology 2003; 144:2933-2940.
    39 Oliver MH, Breier BH, Gluckman PD, et al. Birth weight rather than maternal nutrition influences glucose tolerance, blood pressure, and IGF-I levels in sheep[J]. Pediatr Res2002; 52:516-524.
    40 Christou H, Serdy S, Mantzoros CS. Leptin in relation to growth and developmental processes in the fetus[J]. Semin Reprod Med 2002; 20:123-130.
    41 El-Haddad MA, Desai M, Gayle D, Ross MG. In utero development of fetal thirst and appetite:potential for programming[J]. J Soc Gynecol Investig 2004; 11: 123-130.
    42 Roberts TJ, Nijland MJ, Caston-Balderrama A, et al. Central leptin stimulates ingestive behavior and urine flow in the near term ovine fetus. Horm Metab Res 2001; 33:144-150.
    43 McMillen IC, Muhlhausler BS, Duffield JA, et al. Prenatal programming of postnatal obesity:fetal nutrition and the regulation of leptin synthesis and secretion before birth[J].Proc Nutr Soc 2004; 63:405-412.
    44 Cheverud JM, Pletscher LS, Vaughn TT, et al. Differential response to dietary fat in large (LG/J) and small (SM/J) inbred mouse strains. Physiol Genomics 1999; 1: 33-39.
    45 Snoeck A, Remacle C, Reusens B,et al. Effect of a low protein diet during pregnancy on the fetal rat endocrine pancreas[J].Biol Neonate 1990; 57:107-118.
    46 Bertram CE, Hanson MA. Animal models and programming of the metabolic syndrom[J]e. Br Med Bull 2001; 60:103-121.
    47 Yajnik CS. Early life origins of insulin resistance and type 2 diabetes in India and other Asian countries. J Nutr 2004; 134:205-210.
    48 Singhal A, Farooqi IS, O'Rahilly S, et al. Early nutrition and leptin concentrations in later life[J].Am J Clin Nutr 2002; 75:993-999.
    49 Cianfarani S, Germani D, Rossi P,et al. Intrauterine growth retardation:evidence for the activation of the insulinlike growth factor (IGF)-related growth-promoting machinery and the presence of a cation-independent IGF binding protein-3 proteolytic activity by two months of life[J]. Pediatr Res 1998; 44:374-380.
    50 Leger J, Noel M, Limal JM, et al. Growth factors and intrauterine growth retardation. Ⅱ. Serum growth hormone, insulin-like growth factor (IGF) I, and IGF-binding protein 3 levels in children with intrauterine growth retardation compared with normal control subjects:prospective study from birth to two years of age. Study Group of IUGR[J]. Pediatr Res 1996; 40:101-107.
    51 Gluckman PD, Hanson MA. The developmental origins of the metabolic syndrome[J]. Trends Endocrinol Metab 2004; 15:183-187.
    52 Colle E, Schiff D, Andrew G, et al. Insulin responses during catch-up growth of infants who were small for gestational age[J]. Pediatrics 1976; 57:363-371.
    53 Finken MJJ, Keijzer-Veen MG, Van Mont, et al. Early catch up growth in weight of very preterm low birth weight infants is associated with high level LDL-cholesterol and apo-B at age 19. Pediatr Res 2003; 53:32A.
    54 Metcalfe NB, Monaghan P. Compensation for a bad start:grow now, pay later? [J] Trends Ecol Evol 2001; 16:254-260.
    55 Karlberg J, Albertsson-Wikland K. Growth in full-term small for gestational-age infants:from birth to final height[J]. PediatrRes 1995; 38:733-739.
    56 Eriksson JG, Forsen T, Tuomilehto J, et al. Catch-up growth in childhood and death from coronary heart disease:longitudinal study[J]. BMJ 1999; 318:427-431.
    57 Fewtrell MS, Doherty C, Cole TJ, et al. Effects of size at birth, gestational age and early growth in preterm infants on glucose and insulin concentrations at 9-12 years[J]. Diabetologia 2000; 43:714-717.
    58 Parsons TJ, Power C, Manor O. Fetal and early life growth and body mass index from birth to early adulthood in 1958 British cohort:longitudinal study[J]. BMJ 2001; 323:1331-1335.
    59 Hokken-Koelega AC, De Ridder MA, Lemmen RJ, et al. Children born small for gestational age:do they catch up? [J] Pediatr Res 1995; 38:267-271.
    60 Lucas A. Programming by early nutrition in men.The Childhood Environment andAdult Disease. CIBA Foundation Symposium 156 Chichester:Whiley, UK,1991: 55-83.
    61 Bhargava SK, Sachdev HS, Fall CH, et al. Relation of serial changes in childhood body-mass index to impaired glucose tolerance in young adulthood[J]. N Engl J Med 2004; 350:865-875.
    62 Hales CN, Desai M, Ozanne SE,et al. Fishing in the stream of diabetes:from measuring insulin to the control of fetal organogenesis[J]. Biochem Soc Trans 1996; 24:341-350.
    63 Williams JP, Tanner JM, Hughes PC. Catch-up growth in male rats after growth retardation during the suckling period[J]. Pediatr Res 1974; 8:149-156.
    64 Plagemann A, Harder T, Rake A, et al. Perinatal elevation of hypothalamic insulin, acquired malformation of hypothalamic galaninergic neurons, and syndrome x-like alterations in adulthood of neonatally overfed rats. Brain Res 1999; 836:146-155.
    65 Plagemann A, Heidrich I, Gotz F, et al. Obesity and enhanced diabetes and cardiovascular risk in adult rats due to early postnatal overfeeding[J]. Exp Clin Endocrinol 1992; 99:154-158.
    66 Hahn P. Effect of litter size on plasma cholesterol and insulin and some liver and adipose tissue enzymes in adult rodents[J]. J Nutr 1984; 114:1231-1244.
    67 Morken NH, Kallen K, Hagberg H,et al. Preterm birth in Sweden 1973-2001:rate, subgroups, and effect of changing patterns in multiple births, maternal age, and smoking[J].Acta Obstet Gynecol Scand 2005; 84:558-565.
    68 Branum AM, Schoendorf KC. Changing patterns of low birthweight and preterm birth in the United States,1981-98[J]. Paediatr Perinat Epidemiol 2002; 16:8-15.
    69 Edwards CR, Benediktsson R, Lindsay RS, et al. Dysfunction of placental glucocorticoid barrier:link between fetal environment and adult hypertension? [J] Lancet 1993; 341:355-357.
    70 Bertram CE, Hanson MA. Prenatal programming of postnatal endocrine responses by glucocorticoid[J]s. Reproduction 2002; 124:459-467.
    71 Campbell AL, Murphy BE. The maternal-fetal cortisol gradient during pregnancy and at delivery[J]. J Clin Endocrinol Metab 1977; 45:435-440.
    72 Murphy BE, Clark SJ, Donald IR, et al. Conversion of maternal cortisol to cortisone during placental transfer to the human fetus[J]. Am J Obstet Gynecol 1974; 118:538-541.
    73 Bernal AL, Flint AP, Anderson AB,et al.11 beta-Hydroxyteroid dehydrogenase activity (E.C.1.1.1.146) in human placenta and decidua[J]. J Steroid Biochem 1980; 13:1081-1087
    74 Crowley P. Prophylactic corticosteroids for preterm birth. Cochrane Database Syst Rev 2000; CD000065.
    75 Seckl JR, Meaney MJ. Glucocorticoid programming. Ann NY Acad Sci 2004; 1032:63-84.
    76 Lindsay RS, Lindsay RM, Waddell BJ, et al. Prenatal glucocorticoid exposure leads to offspring hyperglycaemia in the rat:studies with the 11 beta-hydroxysteroid dehydrogenase inhibitor carbenoxolone[J]. Diabetologia 1996; 39:1299-1305.
    77 Langley-Evans SC. Intrauterine programming of hypertension by glucocorticoids[J]. Life Sci 1997; 60:1213-1221.
    78 Langley-Evans SC. Hypertension induced by foetal exposure to a maternal low-protein diet, in the rat, is prevented by pharmacological blockade of maternal glucocorticoid synthesis[J]. J Hypertens 1997; 15:537-544.
    79 Doyle LW, Ford GW, Davis NM, et al. Antenatal corticosteroid therapy and blood pressure at 14 years of age in preterm children. Clin Sci (Lond) 2000; 98:137-142.
    80 Aghajafari F, Murphy K, Willan A, et al. Multiple courses of antenatal corticosteroids:a systematic review and meta-analysis[J]. Am J Obstet Gynecol 2001; 185:1073-1080.
    81 Dalziel SR, Walker NK, Parag V,et al. Cardiovascular risk factors after antenatal exposure to betamethasone:30-year follow-up of a randomised controlled trial[J]. Lancet 2005; 365:1856-1862.
    82 Dessens AB, Haas HS, Koppe JG. Twenty-year follow-up of antenatal corticosteroid treatment[J]. Pediatrics 2000; 105:E77.
    83 Phillips DI, Walker BR, Reynolds RM, et al. Low birth weight predicts elevated plasma cortisol concentr ations in adults from 3 populations[J]. Hypertension 2000; 35:1301-1306.
    84 Levitt NS, Lambert EV, Woods D, et al. Impaired glucose tolerance and elevated blood pressure in low birth weight, nonobese, young south african adults:early programming of cortisol axis[J]. J Clin Endocrinol Metab 2000; 85:4611-4618.
    85 Wyrwoll CS, Mark PJ, Waddell BJ. Developmental programming of renal glucocorticoid sensitivity and the renin-angiotensin system[J]. Hypertension 2007; 50: 579-584.
    86. Waterland RA, Jirtle RL. Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases[J]. Nutrition 2004;20:63-68.
    87. Jaenisch R, Bird A. Epigenetic regulation of gene expression:how the genome integrates intrinsic and environmental signals[J]. Nat Genet 2003;33:245-254.
    88. Fu Q, McKnight RA, Yu X, et al. Growth retardation alters the epigenetic characteristics of hepatic dual specificity phosphatase 5[J]. FASEB J 2006;20:2127-2129.
    89. Lillycrop KA, Phillips ES, Jackson AA, et al. Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring[J]. J Nutr 2005;135:1382-1386.
    90. Thamotharan M, Garg M, Oak S, et al. Transgenerational Inheritance of the Insulin Resistant Phenotype in Embryo-Transferred Intra-Uterine Growth Restricted Adult Female Rat Offspring[J]. Am J Physiol Endocrinol Metab.2007.
    91. Burdge GC, Slater-Jefferies J, Torrens C, et al. Dietary protein restriction of pregnant rats in the generation induces altered methylation of hepatic gene promoters in the adult male offspring in the F1 and F2 generations [J]. Br J Nutr 2007;97:435-439.
    92 Armitage JA, Taylor PD, Poston L. Experimental models of developmental programming:consequences of exposure toan energy rich diet during development[J]. J Physiol 2005; 565:3-8.
    93 Langley-Evans SC. Intrauterine programming of hypertension in the rat:nutrient interactions[J]. Comp Biochem Physiol A Physiol 1996; 114:327-333.
    94 Fetita LS, Sobngwi E, Serradas P, et al. Consequences of fetal exposure to maternal diabetes in offspring[J]. J Clin Endocrinol Metab 2006; 91:3718-3724.
    95 Krishnaveni GV, Veena SR, Hill JC, et al. Intrauterinc exposure to maternal diabetes is associated with higher adiposity and insulin resistance and clustering of cardiovascular risk markers in Indian children[J]. Diabetes Care 2010; 33:402-404.
    96 Harder T, Rodekamp E, Schellong K, et al. Birth weight and subsequent risk of type 2 diabetes:a meta-analysis[J]. Am J Epidemiol 2007; 165:849-857.

© 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