用户名: 密码: 验证码:
妊娠晚期接触细菌脂多糖对胎儿宫内生长发育和骨骼发育的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
细菌脂多糖(lipopolysaccharide, LPS),又称细菌内毒素(endotoxin, ET),是革兰氏阴性细菌细胞壁结构中的类脂多糖,广泛存在人和动物的消化道内。在肠道感染、饮酒等应激状态下,肠粘膜通透性增加,肠道LPS进入血液循环,引起血液LPS浓度迅速升高。动物实验研究已证实,妊娠早期接触LPS可产生显著的胚胎毒性,主要表现为胚胎吸收、流产和早产,但其作用机制目前尚不清楚。本课题在建立小鼠妊娠晚期接触低剂量LPS引起宫内胎儿死亡(IUFD)、生长发育迟缓(IUGR)和骨骼发育迟缓的动物模型基础上,进一步研究了活性氧(ROS)、一氧化氮(NO)和肿瘤坏死因子α(TNF-α)在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用,并深入探讨了抗氧化剂N-乙酰半胱氨酸(NAC)、抗坏血酸(AA)和褪黑素(MT)对LPS引起IUFD、IUGR和骨骼发育迟缓的影响。
     1. LPS诱发IUFD、IUGR和骨骼发育迟缓
     为研究LPS诱发IUFD、IUGR和骨骼发育迟缓的作用,LPS各剂量组孕鼠于妊娠第15-17天每天经腹腔注射一定剂量的LPS(25~75μg/kg, ip),对照组经腹腔注射等容量的生理盐水。妊娠第18天处死所有孕鼠,统计活胎、死胎和吸收胎数,称量活胎体重,测量胎鼠身长和尾长,并对胎鼠骨骼发育情况进行评价。结果显示:LPS各剂量组平均每窝死胎数显著高于对照组,并呈明显的剂量-效应关系。进一步观察发现,妊娠晚期接触LPS显著降低活胎平均体重、身长和尾长,并延缓胎鼠枕骨、胸骨、尾椎骨、前指骨和后掌(趾)骨骨化。上述研究结果表明:母鼠妊娠晚期接触低剂量LPS引起IUFD、IUGR和骨骼发育迟缓。
     2. TNF-α在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用
     为探讨TNF-α在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用,LPS处理组孕鼠于妊娠晚期经腹腔注射LPS(75μg/kg, ip);LPS+PTX处理组孕鼠于注射LPS前0.5 h给予TNF-α的合成抑制剂己酮可可碱(PTX, 100 mg/kg, ip),对照组经腹腔注射等容量的生理盐水或PTX。部分孕鼠给药第1天处死,测母肝、胎肝和胎盘的TNF-αmRNA及羊水和母血清TNF-α水平等。部分孕鼠边续给药3天,于妊娠第18天处死,记录活胎、死胎和吸收胎数,称活胎体重,测量胎鼠身长和尾长,并评价胎鼠骨骼发育情况。结果显示:LPS显著诱导母肝和胎盘组织TNF-αmRNA表达,增加母鼠血清和羊水TNF-α浓度;PTX预处理显著抑制TNF-α产生。进一步研究发现:单纯LPS处理组平均每窝死胎数显著高于对照组,PTX预处理预防LPS引起的IUFD;单纯LPS处理显著降低活胎体重、身长、尾长,延缓胎鼠枕骨、胸骨、尾椎骨、前指骨和后掌(趾)骨骨化,PTX预处理明显减轻LPS引起的生长发育和骨骼发育迟缓。这些研究结果提示:TNF-α至少部分参与LPS引起IUFD、IUGR和骨骼发育迟缓。
     3. ROS和NO在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用
     为探讨ROS和NO在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用,LPS处理组孕鼠于妊娠第15-17天每天注射LPS(75μg/kg, ip),LPS+PBN处理组孕鼠于注射LPS(75μg/kg, ip)前0.5 h和后3 h分别给予一定剂量的2-苯叔丁基硝酮(PBN, 100+50 mg/kg, ip);LPS+AG处理组孕鼠于注射LPS(75μg/kg, ip)前0.5 h和后3 h分别给予一定剂量的氨基胍(AG, 50+25 mg/kg, ip);对照组给予等容量的生理盐水或AG。部分孕鼠给药第1天处死,测母肝、胎肝和胎盘的MDA等。部分孕鼠边续给药3天,于妊娠第18天处死,记录活胎、死胎和吸收胎数,称量活胎体重,测量胎鼠身长和尾长,并对胎鼠骨骼发育情况进行评价。结果显示:AG处理对LPS引起IUFD、IUGR和骨骼发育迟缓几无影响,而PBN处理组平均每窝死胎数明显低于单纯LPS处理组,PBN处理显著减弱LPS降低胎鼠体重、身长和尾长,并显著抑制LPS引起枕骨、胸骨、尾椎骨、前指骨和后掌(趾)骨骨化不全。进一步研究发现,单纯LPS处理组与对照组比较,母鼠血清和羊水中NO水平无显著性差异(资料未列出);PBN显著对抗LPS引起的氧化应激,主要表现为:母肝、胎肝和胎盘组织MDA水平下降且GSH含量上升。这些研究结果提示:ROS在LPS引起IUFD、IUGR和骨骼发育迟缓中可能发挥重要作用,NO在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用不明显。
     4. NAC、AA和MT对LPS引起IUFD、IUGR和骨骼发育迟缓的影响
     NAC、AA和MT是重要的抗氧化剂,为探讨NAC、AA和MT对LPS引起IUFD、IUGR和骨骼发育迟缓的影响,孕鼠被分成两个实验组:实验1,LPS处理组孕鼠于妊娠第15-17天每天经腹腔注射LPS(75μg/kg, ip);LPS+NAC组在LPS处理前和/或处理后经腹腔注射NAC;LPS+AA组在LPS处理前和/或处理后经腹腔注射AA;LPS+MT组在LPS处理前和/或处理后经腹腔注射MT,对照组给予等容量的生理盐水或NAC、AA和MT。所有孕鼠于妊娠第18天处死,统计活胎、死胎和吸收胎数,称量活胎体重,测量活胎身长和尾长,并评价胎鼠骨骼发育情况。实验2,LPS处理组孕鼠于妊娠第15天经腹腔注射单剂量的LPS(75μg/kg),LPS+NAC、LPS+AA和LPS+MT组孕鼠于LPS处理前和/或处理后经腹腔分别注射注射NAC、AA和MT,对照组给予等容量的生理盐水或NAC、AA和MT。孕鼠于LPS处理后1.5 h或6 h处死,检测母肝、胎肝和胎盘组织丙二醛(MDA)和谷胱甘肽(GSH)水平,并测LPS+NAC组羊水和血清TNF-α含量。结果显示:LPS+NAC预处理组、LPS+AA预处理组、LPS+MT预+后处理组和后处理组宫内胎儿死亡数显著低于单纯LPS处理组;LPS+NAC后处理、LPS+AA后和预+后处理组平均每窝死胎数与单纯LPS组比较差异无显著性意义。此外,LPS+NAC预处理和后处理、LPS+AA各处理和LPS+MT各处理组均显著抑制LPS引起IUGR和骨骼发育迟缓。进一步研究发现:NAC预处理显著抑制LPS引起母肝、胎肝和胎盘组织脂质过氧化,NAC后处理显著抑制LPS引起母肝组织脂质过氧化,但对LPS引起的胎肝和胎盘组织脂质过氧化无明显抑制作用;NAC预处理显著抑制LPS引起血清和羊水TNF-α水平上升,而NAC后处理对LPS引起血清TNF-α水平上升无明显影响。AA预和后处理均显著抑制LPS引起母肝、胎肝和胎盘组织脂质过氧化,但AA预处理的作用强于后处理。MT预+后处理显著抑制LPS引起母肝和胎盘组织脂质过氧化,MT后处理显著抑制LPS引起胎盘组织脂质过氧化;但MT预+后处理和后处理对LPS降低母肝组织GSH含量均无明显影响。上述研究结果提示:NAC预处理预防LPS引起IUFD、IUGR和骨骼发育迟缓,NAC后处理对LPS引起IUFD无明显影响,且加重LPS引起早产;AA预处理通过抑制LPS引起的氧化应激,预防LPS引起IUFD、IUGR和骨骼发育迟缓,AA后和预+后处理对抗LPS引起IUGR和骨骼发育迟缓,但对LPS引起IUFD无保护作用;MT通过抑制LPS引起氧化应激,对抗LPS引起IUFD、IUGR和骨骼发育迟缓。
     根据上述研究结果,本课题可得出如下结论:(1)母鼠妊娠晚期接触LPS引起IUFD、IUGR和骨骼发育迟缓;(2)TNF-α部分参与了LPS引起IUFD、IUGR和骨骼发育迟缓;(3)ROS在LPS引起IUFD、IUGR和骨骼发育迟缓中发挥重要作用;NO在LPS引起IUFD、IUGR和骨骼发育迟缓中的作用不明显;(4)NAC预处理预防LPS引起IUFD、IUGR和骨骼发育迟缓,NAC后处理对LPS引起IUFD无明显影响,且加重LPS引起早产;AA预处理预防LPS引起IUFD、IUGR和骨骼发育迟缓,AA后和预+后处理对抗LPS引起IUGR和骨骼发育迟缓,但对LPS引起IUFD无保护作用;MT对LPS引起IUFD、IUGR和骨骼发育迟缓具有明显保护作用。
Lipopolysaccharide (LPS) or endotoxin (ET) is a toxic component of cell walls of gram-negative bacteria and is widely present in the digestive tracts of humans and animals. Gastrointestinal distress and alcohol drinking often increase permeability of LPS from gastrointestinal tract into blood. LPS has been associated with adverse developmental outcome, including embryonic resorption, abortion and preterm labor in animals. However, the exact mechanism of LPS-induced developmental toxicity remains unclear. In this study, we investigated the effect of prenatal exposure to lipopolysaccharide on intra-uterine fetal growth development and skeletal development in mice. We also explored the roles of reactive oxygen species (ROS), tumor necrosis factor-α(TNF-α) and nitric oxide (NO) on LPS-induced IUFD, IUGR and skeletal development retardation in mice, and the effect of N-acetylcysteine (NAC), ascorbic acid (AA) and melatonin (N-acetyl-5-methoxytryptamine, MT) on LPS induced IUFD, IUGR and skeletal development retardation.
     1. The effect of prenatal exposure to lipopolysaccharide on intra-uterine fetal growth development and skeletal development in mice
     To investigate the effects of LPS on IUFD, IUGR and skeletal development retardation. The timed pregnant mice were injected with different doses of LPS (25~75μg/kg, ip) on gestational day (gd) 15-17. The number of live fetuses, dead fetuses and resorption sites was counted on gd 18. Live fetuses in each litter were weighed. Crown-rump and tail lengths were examined and skeletal development was evaluated. Results showed that maternally administered LPS significantly increased fetal mortality in a dose-dependent manner. In addition, maternally administered LPS significantly reduced fetal weight and crown-rump and tail lengths of live fetuses and retarded skeletal ossification in caudal vertebrae, anterior and posterior phalanges, and supraoccipital bone in a dose-dependent manner. These results indicate that maternal LPS exposure at late gestational stages results in IUFD, IUGR and skeletal development retardation in mice.
     2. The role of TNF-αin LPS-induced IUFD, IUGR and skeletal development retardation in mice
     In this study, we investigated the role of TNF-αon LPS-induced IUFD, IUGR and skeletal development retardation in mice. All pregnant mice except controls accepted an intraperitoneal injection of LPS (75μg/kg, ip) daily on gestational late stage. In LPS+PTX group, the pregnant mice were injected with pentoxifylline (PTX, 100 mg/kg, ip) at 0.5 h before LPS treatment. The saline- and PTX- treated pregnant mice served as controls. Some pregnant mice were sacrificed on the day of injection, maternal liver and fetal liver and placenta were dissected for total RNA extraction, maternal serum and amniotic fluid were collected for measurement of TNF-αconcentration, and the left were injected the drug continuously and killed on gd 18. The number of live fetuses, dead fetuses and resorption sites were counted in each litter. Live fetuses were weighted, Crown-rump and tail lengths were measured, and the skeleton of live fetuses were evaluated in each litter. Results showed that pretreatment with PTX, an inhibitor of TNF-αsynthesis, was used to inhibit LPS-evoked TNF-αproduction, As expected, almost blocked LPS-induced IUFD. In addition, PTX significantly alleviated LPS-induced decreases in crown-rump and tail lengths and decrease in fetal weight, and reversed LPS-induced developmental retardation in Posterior phalanx, Caudal vertebrae and breastbone. PTX significantly attenuated LPS-induced lipid peroxidation in maternal liver, fetal liver and placenta. However, PTX had not effect on LPS-induced GSH depletion in maternal liver. Furthermore, PTX evidently mitigated LPS-induced increase in TNF-αconcentration in amniotic fluid and maternal serum. These results indicate that TNF-αis, at least in part, mediated in LPS-induced IUFD, IUGR and skeletal development retardation in mice.
     3. The roles of ROS and NO in LPS-induced IUFD, IUGR and skeletal development retardation in mice
     To investigated the role of Reactive oxygen species (ROS) and nitric oxide (NO) on LPS-induced IUFD, IUGR and skeletal development retardation in mice. In LPS-treated group, the pregnant mice were injected with LPS (75μg/kg, ip) daily on gestational late stage. In the LPS+PBN group, the pregnant mice were injected with 100 mg/kg of Alpha-phenyl-N-t-butylnitrone (PBN, ip) at 0.5 h before LPS (75μg/kg, ip) treatment, followed by additional dose (50 mg/kg) of PBN at 3 h after LPS treatment. In the LPS+AG group, the pregnant mice were injected with 50 mg/kg of aminoguanidine (AG, ip) at 0.5 h before LPS (75μg/kg, ip) treatment, followed by additional dose (25 mg/kg) of AG at 3 h after LPS treatment. the saline- and AG- treated pregnant mice served as controls. Some pregnant mice were sacrificed on the day of injection, maternal liver, fetal liver and placenta were dissected for measurement of MDA concentration and so on. The left were injected the drug continuously and killed on gd 18, For each litter, the number of live fetuses, dead fetuses and resorption sites were counted. Live fetuses in each litter were weighted. Crown-rump and tail lengths were measured. And the skeleton of all live fetuses in each litter were evaluated. Results showed that PBN, a free radical spin-trapping agent, almost blocked LPS-induced IUFD and revered LPS-induced intra-uterine skeletal development retardation. And abolished LPS-induced decrease in fetal weight and crown-rump and tail lengths. However, AG, an inhibitor of inducible nitric oxide synthase (iNOS) activity, which was used to inhibit LPS-evoked NO production, had no effect on LPS-induced IUFD, IUGR and skeletal development retardation in mice. Furthermore, PBN significantly attenuated LPS-induced lipid peroxidation in maternal liver, fetal liver and placenta, and GSH depletion in maternal liver and fetal liver. In addition, NO contents in serum and amniotic fluid were statistically indistinguishable between LPS-treated groups and control (data not shown). These results suggest that oxidative stress is, at least in part, mediated in LPS-induced IUFD, IUGR and skeletal development retardation; and LPS-induced IUFD, IUGR and skeletal development retardation in mice are independent of NO production.
     4. The effect of NAC, AA and MT on LPS-induced IUFD, IUGR and skeletal development retardation in mice
     NAC, AA and MT are potent antioxidants. To investigate the role of NAC, AA and MT on LPS-induced IUFD, IUGR and skeletal development retardation in ICR mice. The present study included two separate experiments. In experiment one, All pregnant mice except controls (either saline or NAC, AA, MT) received an intraperitoneal injection of LPS (75μg/kg, ip) on gd 15-17. In LPS+NAC, LPS+AA and LPS+MT groups, the pregnant mice were treated with NAC, AA or MT at before and / or after LPS. The number of live fetuses, dead fetuses and resorption sites were counted on gd 18. Live fetuses in each litter were weighed. Crown-rump and tail lengths were examined and skeletal development was evaluated. In experiment two, All pregnant mice except controls (either saline or NAC, AA, MT) received an intraperitoneal (75μg/kg) injection of LPS on gd 15. In LPS+NAC, LPS+AA and LPS+MT groups, the pregnant mice were treated with NAC, AA or MT at before and / or after LPS. All dams were sacrificed at 1.5 h or 6 h after LPS treatment. Maternal liver, fetal liver and placenta were dissected for GSH and MDA measurements, maternal serum and amniotic fluid were collected for TNF-αanalyses. Results showed that pretreatment with NAC, AA, and pre- plus post- treatments and post-treatment with MT significantly attenuated LPS-induced IUFD. However, post-treatment with NAC, both post- and pre- plus post- treatments with AA had less effect on LPS-induced IUFD. In addition, pre- and post- treatment with NAC, AA- and MT- treatments significantly alleviated LPS-induced IUGR, and reversed LPS-induced skeletal developmental retardation. Furthermore, pretreatment with NAC significantly attenuated LPS-induced elevation in TNF-a concentration in maternal serum and amniotic fluid and lipid peroxidation in maternal liver, fetal livers and placenta. By contrast to pretreatment, post-treatment with NAC had no effect on LPS-induced TNF-a production and lipid peroxidation; Pre- and post- treatmentwith AA significantly attenuated LPS-induced lipid peroxidation; pre- plus post- treatments with melatonin significantly attenuated LPS-induced lipid peroxidation in maternal liver and placenta. However, melatonin had no effect on LPS-induced GSH depletion. These results indicate that NAC had a dual effect on LPS-induced IUFD, IUGR and skeletal development. Pretreatment with NAC improves fetal survival and reverses LPS-induced IUGR and skeletal development retardation, whereas post-treatment with NAC aggravates LPS induced preterm labor; Pretreatment with AA protected against LPS-induced IUFD and reversed LPS-induced IUGR and skeletal development retardation, whereas post-treatment and pre- + post-treatment had less effect on LPS-induced IUFD. MT protects against LPS-induced IURD, IUGR and skeletal development retardation via counteracting LPS-induced oxidative stress.
     In summary, the present results allow us to reach the following conclusions. First, the timed pregnant mice were injected low dose of LPS on gestational day (gd) 15-17 resulted in intra-uterine fetal death (IUFD), intra-uterine growth retardation (IUGR) and skeletal development retardation in mice; second, TNF-αpartially contributes to LPS-induced IUFD, IUGR and skeletal development retardation in mice; third, LPS-induced IUFD, IUGR and skeletal development retardation in mice is mediated, at least in part, by reactive oxygen species; fourth, LPS-induced IURD, IUGR and skeletal development retardation in mice is independent of NO production; and fifth, NAC had a dual effect on LPS-induced IUFD, IUGR and skeletal development retardation, pretreatment with NAC improves fetal survival and reverses LPS-induced IUGR and skeletal development retardation, whereas post-treatment with NAC aggravates LPS induced preterm labor; Pretreatment with AA protected against LPS-induced IUFD and reversed LPS-induced IUGR and skeletal development retardation, whereas post-treatment and pre- + post-treatment had less effect on LPS-induced IUFD; and MT protects against LPS-induced IURD, IUGR and skeletal development retardation in mice.
引文
1. Seeds JW, Peng T. Impaired growth and risk of fetal death: is the tenth percentile the appropriate standard? Am J Obstet Gynecol, 1998; 178(4): 658-669.
    2. 刘建蒙, 李竹, 赵平, 等. 胎儿宫内生长迟缓与脑性瘫痪. 中华预防医学杂志, 2001; 35(6): 390-393.
    3. Yu VY, Upadhyay A. Neonatal management of the growth-restricted infant. Semin Fetal Neonatal Med, 2004; 9(5): 403-409.
    4. Tian JY, Cheng Q, Song XM, et al. Birth weight and risk of type 2 diabetes, abdominal obesity and hypertension among Chinese adults. Eur J Endocrinol, 2006; 155(4): 601-607.
    5. Jaquet D, Leger J, Levy-Marchal C, et al. Low birth weight: effect on insulin sensitivity and lipid metabolism. Horm Res, 2003; 59(1): 1-6.
    6. Engelbregt MJ, van Weissenbruch MM, Lips P. Body composition and bone measurements in intra-uterine growth retarded and early postnatally undernourished male and female rats at the age of 6 months: comparison with puberty. Bone, 2004; 34(1): 180-186.
    7. 陶芳标, 许进, 邓国支, 等. 低出生体重儿死亡率与脑性瘫痪患病率的研究. 安徽医科大学学报, 2003; 38(5): 374-376.
    8. 郑树国, 邓辉, 鲍月琴, 等. 低出生体重儿和早产儿牙釉质发育缺陷及其易感因素的临床分析研究. 北京医科大学学报, 1999; 31(1): 59-61.
    9. Doyle LW, Faber B, Callanan C, et al. Bronchopulmonary dysplasia in very low birth weight subjects and lung function in late adolescence. Pediatrics, 2006; 118(1): 108-13.
    10. 毛淑芳, 李文华, 尹长恒. 胎儿宫内生长发育迟缓的远期不良影响. 承德医学院学报, 2001; 18(1): 66-68.
    11. Valero De Bernabe J, Soriano T, Albaladejo R, et al. Risk factors for low birth weight: a review. Eur J Obstet Gynecol Reprod Biol, 2004; 116(1): 3-15.
    12. Kimura KA, Reynolds JN, Brien JF. Ethanol neurobehavioral teratogenesis and the role of the hippocampal glutamate-N-methyl-D-aspartate receptor-nitric oxide synthase system. Neurotoxicol Teratol, 2000; 22(5): 607-616.
    13. Kalter H. Teratology in the 20th century: environmental causes of congenital malformations in humans and how they were established. Neurotoxicol Teratol, 2003; 25(2): 131-282.
    14. Parman T, Wiley MJ, Wells PG. Free radical-mediated oxidative DNA damage in the mechanism of thalidomide teratogenicity. Nat Med, 1999; 5(5): 582-585.
    15. Bernstein IM, Mongeon JA, Badger GJ, et al. Maternal smoking and its association with birth weight. Obstet Gynecol, 2005; 106(5 Pt 1): 986-991.
    16. Jensen DM, Damm P, Moelsted-Pedersen L, et al. Outcomes in Type 1 Diabetic Pregnancies:A nationwide, population-based study. Diabetes Care, 2004; 27(12): 2819-2823.
    17. Penney GC, Mair G, Pearson DW. Scottish Diabetes in Pregnancy Group. Outcomes of pregnancies in women with type 1 diabetes in Scotland: a national population-based study. BJOG, 2003; 110(3): 315-318.
    18. Vasileiadis GT, Roukema HW, Romano W, et al. Intrauterine herpes simplex infection. Am J Perinatol, 2003; 20(2): 55-58.
    19. Jacob AI, Goldberg PK, Bloom N, et al. Endotoxin and bacteria in portal blood. Gastroenterology, 1977; 72(6): 1268-1270.
    20. Lambert GP. Role of gastrointestinal permeability in exertional heatstroke. Exerc Sport Sci Rev, 2004; 32(4): 185-190.
    21. Zhou Z, Wang L, Song Z, et al. A critical involvement of oxidative stress in acutealcohol-induced hepatic TNF-alpha production. Am J Pathol, 2003; 163(3): 1137-1146.
    22. Chen YH, Wang JP, Wang H, et al. Lipopolysaccharide treatment downregulates the expression of the pregnane X receptor, cyp3a11 and mdr1a genes in mouse placenta. Toxicology, 2005; 211(3): 242-252.
    23. Xu DX, Chen YH, Wang JP, et al. Perinatal lipopolysaccharide exposure downregulates pregnane X receptor and Cyp3a11 expression in fetal mouse liver. Toxicol Sci, 2005; 87(1): 38-45.
    24. Collins JG, Smith MA, Arnold RR. Effects of Escherichia coli and Porphyromonas gingivalis lipopolysaccharide on pregnancy outcome in the golden hamster. Infect. Immun, 1994; 62(10): 4652-6455.
    25. Deb K, Chaturvedi MM, Jaiswal YK. A 'minimum dose' of lipopolysaccharide required for implantation failure: assessment of its effect on the maternal reproductive organs and interleukin-1alpha expression in the mouse. Reproduction, 2004; 128(1): 87-97.
    26. Romero R, Espinoza J, Mazor M. Can endometrial infection/inflammation explain implantation failure, spontaneous abortion, and preterm birth after in vitro fertilization? Fertil Steril, 2004; 82(4): 799-804.
    27. Vazquez-Torres A, Vallance BA, Bergman MA, et al. Toll-like receptor 4 dependence of innate and adaptive immunity to Salmonella: importance of the Kupffer cell network. J Immunol, 2004; 172(10): 6202-6208.
    28. Wang H, Hirsch E. Bacterially-induced preterm labor and regulation of prostaglandin metablizing enzyme expression in mice: The role of toll-like receptor 4. Biol Reprod, 2003; 69(6): 1957-1963.
    29. Kobayashi H, Yoshida R, Kanada Y, et al. Suppression of lipopolysaccharide- induced cytokine production of gingival fibroblasts by a soybean, Kunitz trypsininhibitor. J Periodontal Res, 2005; 40(6): 461-468.
    30. Covert MW, Leung TH, Gaston JE, et al. Achieving stability of lipopolysaccharide- induced NF-kappaB activation. Science, 2005; 309(5742): 1854-1857.
    31. Park HS, Jung HY, Park EY, et al. Cutting edge: direct interaction of TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced production of reactive oxygen species and activation of NF-kappa B. J Immunol, 2004; 173(6): 3589-3593.
    32. Rhee SH, Hwang D. Murine TOLL-like receptor 4 confers lipopolysaccharide responsiveness as determined by activation of NF kappa B and expression of the inducible cyclooxygenase. J Biol Chem, 2000; 275(44): 34035-34040.
    33. Liu Y, Dong W, Chen L, et al. BCL10 mediates lipopolysaccharide/toll-like receptor-4 signaling through interaction with Pellino2. J Biol Chem, 2004; 279(36): 37436-37444.
    34. Tsan MF, Clark RN, Goyert SM, et al. Induction of TNF-alpha and MnSOD by endotoxin: role of membrane CD14 and Toll-like receptor-4. Am J Physiol Cell Physiol, 2001; 280(6): C1422-1430.
    35. Silver RM, Edwin SS, Trautman MS, et al. Bacterial lipopolysaccharide-mediated fetal death. Production of a newly recognized form of inducible cyclooxygenase (COX-2) in murine decidua in response to lipopolysaccharide. J Clin Invest, 1995; 95(2): 725-731.
    36. Gross G, Imamura T, Vogt SK, et al. Inhibition of cyclooxygenase-2 prevents inflammation-mediated preterm labor in the mouse. Am J Physiol Regul Integr Comp Physiol, 2000; 278(6): R1415-1423.
    37. Ogando DG, Paz D, Cella M, et al. The fundamental role of increased production of nitric oxide in lipopolysaccharide-induced embryonic resorption in mice.Reproduction, 2003; 125(1): 95-110.
    38. Leazer TM, Barbee B, Ebron-McCoy M, et al. Role of the maternal acute phase response and tumor necrosis factor alpha in the developmental toxicity of lipopolysaccharide in the CD-1 mouse. Reprod Toxicol, 2002; 16(2): 173-179.
    39. Frost RA, Nystrom GJ, Lang CH. Tumor necrosis factor-alpha decreases insulin-like growth factor-I messenger ribonucleic acid expression in C2C12 myoblasts via a Jun N-terminal kinase pathway. Endocrinology, 2003; 144(5): 1770-1779.
    40. Priego T, Ibanez de Caceres I, Martin AI, et al. NO plays a role in LPS-induced decreases in circulating IGF-I and IGFBP-3 and their gene expression in the liver. Am J Physiol Endocrinol Metab, 2004; 286(1): E50-56.
    41. Randhawa R, Cohen P. The role of the insulin-like growth factor system in prenatal growth. Mol Genet Metab, 2005; 86(1-2): 84-90.
    42. Abuzzahab MJ, Schneider A, Goddard A, et al. IGF-I receptor mutations resulting in intrauterine and postnatal growth retardation. N Engl J Med, 2003; 349(23): 2211-2222.
    43. Buhimschi IA, Buhimschi CS, Weiner CP. Protective effect of N-acetylcysteine against fetal death and preterm labor induced by maternal inflammation. Am J Obstet Gynecol, 2003; 188(1): 203-208.
    44. Victor VM, Rocha M, De la Fuente M. N-acetylcysteine protects mice from lethal endotoxemia by regulating the redox state of immune cells. Free Radic Res, 2003; 37(9): 919-29.
    45. Xu DX, Wei W, Sun MF, et al. Melatonin attenuates lipopolysaccharide-induced down-regulation of pregnane X receptor and its target gene CYP3A in mouse liver. J Pineal Res, 2005; 38(1): 27-34.
    46. Banfi C, Sironi L, De Simoni G, et al. Pentoxifylline prevents spontaneous brainischemia in stroke-prone rats. J Pharmacol Exp Ther, 2004; 310(3): 890-895..
    47. Ehlers S, Mielke ME, Blankenstein T, et al. Kinetic analysis of cytokine gene expression in the livers of naive and immune mice infected with Listeria monocytogenes. The immediate early phase in innate resistance and acquired immunity. J Immunol, 1992; 149(9): 3016-3022.
    48. Murray LJ, Lee R, Martens C. In vivo cytokine gene expression in T cell subsets of the autoimmune MRL/Mp-lpr/lpr mouse. Eur J Immunol, 1990; 20(1): 163-170.
    49. Tracey WR, Tse J, Carter G. Lipopolysaccharide-induced changes in plasma nitrite and nitrate concentrations in rats and mice: pharmacological evaluation of nitric oxide synthase inhibitors. J Pharmacol Exp Ther, 1995; 272(3): 1011-1015.
    50. Grisham MB, Johnson GG, Lancaster JR Jr. Quantitation of nitrate and nitrite in extracellular fluids. Methods Enzymol, 1996; 268: 237-246.
    51. Griffith OW. Determination of glutathione and glutathione disulfide using glutathione reductase and 2-vinylpyridine. Anal Biochem, 1980; 106(1): 207-212.
    52. Lowry OH, Rosebrough NJ, Farr AL, et al. Protein measurement with the Folin phenol reagent. J Biol Chem, 1951; 193(1): 265-275.
    53. Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxidation in animal tissues by thiobarbituric acid reaction. Anal Biochem, 1979; 95(2): 351-358.
    54. Gayle DA, Beloosesky R, Desai M, et al. Maternal LPS induces cytokines in the amniotic fluid and corticotropin releasing hormone in the fetal rat brain. Am J Physiol Regul Integr Comp Physiol, 2004; 286(6): R1024-1029.
    55. Cheng YJ, Liu MY, Wu TP, et al. Regulation of tumor necrosis factor-alpha in glioma cells by lead and lipopolysaccharide: involvement of common signaling pathway. Toxicol Lett, 2004; 152(2): 127-137.
    56. Silver RM, Lohner WS, Daynes RA, et al. Lipopolysaccharide-induced fetal death:the role of tumor-necrosis factor alpha. Biol Reprod, 1994; 50(5): 1108-1112.
    57. Bell MJ, Hallenbeck JM, Gallo V. Determining the fetal inflammatory response in an experimental model of intrauterine inflammation in rats. Pediatr Res, 2004; 56(4): 541-546.
    58. Gendron RL, Nestel FP, Lapp WS, et al. Lipopolysaccharide-induced fetal resorption in mice is associated with the intrauterine production of tumour necrosis factor-alpha. J Reprod Fertil, 1990; 90(2): 395-402.
    59. Liu YP, Lin HI, Tzeng SF. Tumor necrosis factor-alpha and interleukin-18 modulate neuronal cell fate in embryonic neural progenitor culture. Brain Res, 2005; 1054(2): 152-158.
    60. Robin MA, Demeilliers C, Sutton A, et al. Alcohol increases tumor necrosis factor alpha and decreases nuclear factor-kappab to activate hepatic apoptosis in genetically obese mice. Hepatology, 2005; 42(6): 1280-1290.
    61. Beshay E, Croze F, Prud'homme GJ. The phosphodiesterase inhibitors pentoxifylline and rolipram suppress macrophage activation and nitric oxide production in vitro and in vivo. Clin Immunol, 2001; 98(2): 272-279.
    62. Loftis LL, Meals EA, English BK. Differential effects of pentoxifylline and interleukin-10 on production of tumor necrosis factor and inducible nitric oxide synthase by murine macrophages. J Infect Dis, 1997; 175(4): 1008-1011.
    63. Kwiecien S, Brzozowski T, Konturek PC, et al. Gastroprotection by pentoxyfilline against stress-induced gastric damage. Role of lipid peroxidation, antioxidizing enzymes and proinflammatory cytokines. J Physiol Pharmacol, 2004; 55(2): 337-55.
    64. Arias-Diaz J, Vara E, Torres-Melero J, et al. Local production of oxygen free radicals and nitric oxide in rat diaphragm during sepsis: effects of pentoxifylline and somatostatin. Eur J Surg, 1997; 163(8): 619-625.
    65. Hecht M, Muller M, Lohmann-Matthes ML, et al. In vitro and in vivo effects of pentoxifylline on macrophages and lymphocytes derived from autoimmune MRL-lpr/lpr mice. J Leukoc Biol, 1995; 57(2): 242-249.
    66. Kohmura Y, Kirikae T, Kirikae F, et al. Lipopolysaccharide (LPS)-induced intra-uterine fetal death (IUFD) in mice is principally due to maternal cause but not fetal sensitivity to LPS. Microbiol Immunol, 2000; 44(11): 897–904.
    67. Casado M, Velasco M, Bosca L. Absence of NO synthase type II expression in fetal liver from pregnant rats under septic shock conditions. Hepatology, 1997; 25(6): 1406-1411.
    68. Bautista AP, Meszaros K, Bojta J, et al. Superoxide anion generation in the liver during the early stage of endotoxemia in rats. J Leukoc Biol, 1990; 48(2): 123-128.
    69. Ejima K, Koji T, Nanri H, et al. Expression of thioredoxin and thioredoxin reductase in placentae of pregnant mice exposed to lipopolysaccharide. Placenta, 1999; 20(7): 561-566.
    70. Tanaka N,Tajima S,Ishibashi A. Immunohistochemical detection of lipid peroxidation products,protein-bound acrolein and 4-hydroxynonenal protein adducts, in actinic elastosis of photodamaged skin. Arch Dermatol Res, 2001; 293(7): 363-367.
    71. Xu DX, Shen HM, Zhu QX, et al. The associations among semen quality, oxidative DNA damage in human spermatozoa and concentrations of cadmium, lead and selenium in seminal plasma. Mutat Res, 2003; 534(1-2): 155-163.
    72. Parman T, Wiley MJ, Wells PG. Free radical-mediated oxidative DNA damage in the mechanism of thalidomide teratogenicity. Nat Med, 1999; 5(5): 582-585.
    73. Winn LM, Wells PG. Evidence for embryonic prostaglandin H synthase-catalyzed bioactivation and reactive oxygen species-mediated oxidation of cellular macromolecules in phenytoin and benzo[a]pyrene teratogenesis. Free Radic BiolMed, 1997; 22(4): 607-621.
    74. Wells PG, Kim PM, Laposa RR, et al. Oxidative damage in chemical teratogenesis. Mutat Res, 1997; 396(1-2): 65-78.
    75. Kotch LE, Chen SY, Sulik KK. Ethanol-induced teratogenesis: free radical damage as a possible mechanism. Teratology, 1995; 52(3): 128-136.
    76. Lee JH, Park JW. Protective role of alpha-phenyl-N-t-butylnitrone against ionizing radiation in U937 cells and mice. Cancer Res, 2003; 63(20): 6885-6893.
    77. Ogando DG, Paz D, Cella M, et al. The fundamental role of increased production of nitric oxide in lipopolysaccharide-induced embryonic resorption in mice. Reproduction, 2003; 125(1): 95-110.
    78. Athanassakis I, Aifantis I, Ranella A, et al. Inhibition of Nitric Oxide production rescues LPS-induced fetal abortion in mice. Nitric Oxide, 1999; 3(3): 216-224.
    79. Nehru B, Kanwar SS. N-acetylcysteine exposure on lead-induced lipid peroxidative damage and oxidative defense system in brain regions of rats. Biol Trace Elem Res, 2004; 101(3): 257-264.
    80. Aruoma OI, Halliwell B, Hoey BM, et al. The antioxidant action of N-acetylcysteine: its reaction with hydrogen peroxide, hydroxyl radical, superoxide, and hypochlorous acid. Free Radic Biol Med, 1989; 6(6): 593-597.
    81. Song M, Kellum JA, Kaldas H, et al. Evidence that glutathione depletion is a mechanism responsible for the anti-inflammatory effects of ethyl pyruvate in cultured lipopolysaccharide-stimulated RAW 264.7 cells. J Pharmacol Exp Ther, 2004; 308(1): 307-316.
    82. Neuschwander-Tetri BA, Bellezzo JM, Britton RS, et al. Thiol regulation of endotoxin-induced release of tumour necrosis factor alpha from isolated rat Kupffer cells. Biochem J, 1996; 320(Pt3): 1005-1010.
    83. Hadzic T, Li L, Cheng N, et al. The role of low molecular weight thiols in T lymphocyte proliferation and IL-2 secretion. J Immunol, 2005; 175(12): 7965-7972.
    84. Xu DX, Chen YH, Wang H, et al. Tumor necrosis factor alpha partially contributes to lipopolysaccharide-induced intra-uterine fetal growth restriction and skeletal development retardation in mice. Toxicol Lett, 2006; 163(1): 20-29.
    85. Peristeris P, Clark BD, Gatti S, et al. N-acetylcysteine and glutathione as inhibitors of tumor necrosis factor production. Cell Immunol, 1992; 140(2): 390-399.
    86. Victor VM, De la Fuente M. N-acetylcysteine improves in vitro the function of macrophages from mice with endotoxin-induced oxidative stress. Free Radic Res, 2002; 36(1): 33-45.
    87. Sagrista ML, Garcia AE, Africa De Madariaga M, et al. Antioxidant and pro-oxidant effect of the thiolic compounds N-acetyl-L-cysteine and glutathione against free radical-induced lipid peroxidation. Free Radic Res, 2002; 36(3): 329-340.
    88. Shen H, Yang C, Liu J, et al. Dual role of glutathione in selenite-induced oxidative stress and apoptosis in human hepatoma cells. Free Radic Biol Med, 2000; 28(7): 1115-1124.
    89. Oikawa S, Yamada K, Yamashita N, et al. N-acetylcysteine, a cancer chemopreventive agent, causes oxidative damage to cellular and isolated DNA. Carcinogenesis, 1999; 20(8): 1485-1490.
    90. Sprong RC, Winkelhuyzen-Janssen AM, Aarsman CJ, et al. Low-dose N-acetylcysteine protects rats against endotoxin-mediated oxidative stress, but high-dose increases mortality. Am J Respir Crit Care Med, 1998; 157(4 Pt 1): 1283-1293.
    91. De la Fuente M, Victor VM. Ascorbic acid and N-acetylcysteine improve in vitrothe function of lymphocytes from mice with endotoxin-induced oxidative stress. Free Radic Res, 2001; 35(1): 73-84.
    92. Peng Y, Kwok KH, Yang PH, et al. Ascorbic acid inhibits ROS production, NF-kappa B activation and prevents ethanol-induced growth retardation and microencephaly. Neuropharmacology, 2005; 48(3): 426-434.
    93. Sen Gupta R, Sen Gupta E, Dhakal BK, et al. Vitamin C and vitamin E protect the rat testes from cadmium-induced reactive oxygen species. Mol Cells, 2004; 17(1): 132-139.
    94. Paolini M, Pozzetti L, Pedulli GF, et al. The nature of prooxidant activity of vitamin C. Life Sci, 1999; 64(23): PL 273-278.
    95. Harreus U, Baumeister P, Zieger S, et al. The influence of high doses of vitamin C and zinc on oxidative DNA damage. Anticancer Res, 2005; 25(5): 3197-3201.
    96. Li SD, Su YD, Li M, et al. Hemin-mediated Hemolysis in Erythrocytes: Effects of Ascorbic Acid and Glutathione. Acta Biochim Biophys Sin (Shanghai), 2006; 38(1): 63-69.
    97. Chen Q, Wei W. Effects and mechanisms of melatonin on inflammatory and immune responses of adjuvant arthritis rats. Int Immunopharmacol, 2002; 2(10): 1443-1449.
    98. Wei W, Shen YX, Dai M, et al. Effects and mechanisms of melatonin on immune responses in mice of different months. Acta Pharmacol Sin, 2003; 24(7): 719-723.
    99. Okatani Y, Okamoto K, Hayashi K, et al. Maternal-fetal transfer of melatonin in pregnant women near term. J Pineal Res, 1998; 25(3): 129-134.
    100. Allegra M, Reiter RJ, Tan DX, et al. The chemistry of melatonin's interaction with reactive species. J Pineal Res, 2003; 34(1): 1-10.
    101. Matuszak Z, Reszka K, Chignell CF. Reaction of melatonin and related indoleswith hydroxyl radicals: EPR and spin trapping investigations. Free Radic Biol Med, 1997; 23(3): 367-372.
    102. Tan DX, Reiter RJ, Manchester LC, et al. Chemical and physical properties and potential mechanisms: melatonin as a broad spectrum antioxidant and free radical scavenger. Curr Top Med Chem, 2002; 2(2): 181-197.
    103. Rodriguez C, Mayo JC, Sainz RM, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res, 2004; 36(1): 1-9.
    104. Shen YX, Xu SY, Wei W, et al. The protective effects of melatonin from oxidative damage induced by amyloid beta-peptide 25-35 in middle-aged rats. J Pineal Res, 2002; 32(3): 85-89.
    105. Tan DX, Manchester LC, Reiter RJ, et al. A novel melatonin metabolite, cyclic 3-hydroxymelatonin: a biomarker of in vivo hydroxyl radical generation. Biochem Biophys Res Commun, 1998; 253(3): 614-620.
    106. Zhang H, Squadrito GL, Uppu R, et al. Reaction of peroxynitrite with melatonin: A mechanistic study. Chem Res Toxicol, 1999; 12(6): 526-534.
    107. El-Sokkary GH, Omar HM, Hassanein AF, et al. Melatonin reduces oxidative damage and increases survival of mice infected with Schistosoma mansoni. Free Radic Biol Med, 2002; 32(4): 319-332.
    108. Blanchard B, Pompon D, Ducrocq C. Nitrosation of melatonin by nitric oxide and peroxynitrite. J Pineal Res, 2000; 29(3): 184-192.
    109. Tan DX, Manchester LC, Reiter RJ, et al. Significance of melatonin in antioxidative defense system: reactions and products. Biol Signals Recept, 2000; 9(3-4): 137-159.
    110. Rodriguez C, Mayo JC, Sainz RM, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res, 2004; 36(1): 1-9.
    111. Liu F, Ng TB. Effect of pineal indoles on activities of the antioxidant defense enzymes superoxide dismutase, catalase, and glutathione reductase, and levels of reduced and oxidized glutathione in rat tissues. Biochem Cell Biol, 2000; 78(4): 447-453.
    112. Jaworek J, Leja-Szpak A, Bonior J, et al. Protective effect of melatonin and its precursor L-tryptophan on acute pancreatitis induced by caerulein overstimulation or ischemia/reperfusion. J Pineal Res, 2003; 34(1): 40-52.
    113. Crespo E, Macias M, Pozo D, et al. Melatonin inhibits expression of the inducible NO synthase II in liver and lung and prevents endotoxemia in lipopolysaccharide-induced multiple organ dysfunction syndrome in rats. FASEB J, 1999; 13(12): 1537-1546.
    1. Jirillo E, Caccavo D, Magrone T, et al. The role of the liver in the response to LPS: experimental and clinical findings. J Endotoxin Res, 2002; 8(5): 319-327.
    2. 芦惠, 王丽杰. 新生鼠肠损伤时肝脏黄嘌呤氧化酶的变化. 世界华人消化杂志, 2004; 12(10): 2486-2487.
    3. Tuin A, Huizinga-Van der Vlag A, van Loenen-Weemaes AM, et al. On the role and fate of LPS-dephosphorylating activity in the rat liver. Am J Physiol Gastrointest Liver Physiol, 2006; 290(2): G377-85.
    4. Cheng YJ, Yang BC, Liu MY. Lead increases lipopolysaccharide-induced liver-injury through tumor necrosis factor-alpha overexpression by monocytes/macrophages: role of protein kinase C and P42/44 mitogen-activated protein kinase. Environ Health Perspect, 2006; 114(4): 507-13.
    5. Lu Y, Cederbaum AI. Enhancement by pyrazole of lipopolysaccharide-induced liver injury in mice: role of cytochrome P450 2E1 and 2A5. Hepatology, 2006; 44(1): 263-74.
    6. Kitoh Y, Ohmori M, Araki N, et al. Dosing-time-dependent differences in lipopolysaccharide-induced liver injury in rats. Chronobiol Int, 2005; 22(6): 987-996.
    7. 徐正婕, 范建高, 王国良, 等. 小剂量内毒素导致非酒精性脂肪性肝炎大鼠急性肝功能衰竭. 中华肝脏病杂志, 2004; 12(4): 244.
    8. Fujimoto M, Uemura M, Nakatani Y, et al. Plasma endotoxin and serum cytokine levels in patients with alcoholic hepatitis: relation to severity of liver disturbance. Alcohol Clin Exp Res, 2000; 24(4): 48S-54S.
    9. Hansen J, Cherwitz DL, Allen JI. The role of tumor necrosis factor-alpha in acture endotoxin-induced hepatotoxicity in ethanol-fed rats. Hepatology, 1994; 20(2):461-474.
    10. Adachi Y, Moore LE, Bradford BU, et al. Antibiotics prevent liver in rats following long-term exposure to ethanol. Gastroenterology, 1995; 108(1): 218-224.
    11. Park EJ, Jeon CH, Ko G, et al. Protective effect of curcumin in rat liver injury induced by carbon tetrachloride. J Pharm Pharmacol, 2000; 52(4): 437-440.
    12. Tsukamoto H, Towner SJ, Ciofalo LM, et al. Ethanol-induced liver fibrosis in rats fed high fat diet. Hepatology, 1986; 6(5): 814-822.
    13. Nanji, AA, Khettry U, Sadrzadeh SM. Lactobacillus feeding reduces endotoxemia and severity of experimental alcoholic liver (disease). Proc Soc Exp Biol Med, 1994; 205(3): 243-247.
    14. Czaja MJ, Xu J, Ju Y, et al. Lipopolysaccharide-neutralizing antibody reduces hepatocyte injury from acute hepatotoxin administration. Hepatology, 1994; 19(5): 1282-1289.
    15. Romero R, Espinoza J, Mazor M. Can endometrial infection/inflammation explain implantation failure, spontaneous abortion, and preterm birth after in vitro fertilization? Fertil Steril, 2004; 82(4): 799-804.
    16. Deb K, Chaturvedi MM, Jaiswal YK. A ‘minimum dose' of lipopolysaccharide required for implantation failure: assessment of its effect on the maternal reproductive organs and interleukin-1alpha expression in the mouse. Reproduction, 2004; 128(1): 87-97.
    17. Elovitz MA, Wang Z, Chien EK, et al. A new model for inflammation-induced preterm birth: the role of platelet-activating factor and Toll-like receptor-4. Am J Pathol, 2003; 163(5): 2103-2111.
    18. McLauchlin J. Human listeriosis in Britain, 1967-85, a summary of 722 cases. 1. Listeriosis during pregnancy and in the newborn. Epidemiol Infect, 1990; 104(2):181-189.
    19. Li RY, Tsutsui Y. Growth retardation and microcephaly induced in mice by placental infection with murine cytomegalovirus. Teratology, 2000; 62(2): 79-85.
    20. Atreya CD, Mohan KV, Kulkarni S. Rubella virus and birth defects: molecular insights into the viral teratogenesis at the cellular level. Birth Defects Res A Clin Mol Teratol, 2004; 70(7): 431-437.
    21. Lanning JC, Hilbelink DR, Chen LT. Teratogenic effects of endotoxin on the golden hamster. Teratog Carcinog Mutagen, 1983; 3(2): 145-149.
    22. Chua JS, Rofe AM, Coyle P. Dietary zinc supplementation ameliorates LPS-induced teratogenicity in mice. Pediatr Res, 2006; 59(3): 355-358.
    23. Paige DM, Augustyn M, Adih WK, et al. Bacterial vaginosis and preterm birth: a comprehensive review of the literature. J Nurse Midwifery, 1998; 43(2): 83–89.
    24. Dasanayake AP. Poor periodontal health of the pregnant woman as a risk factor for low birth weight. Ann Periodontol, 1998; 3(1): 206-212.
    25. Offenbacher S, Katz V, Fertik G, et al. Periodontal infection as a possible risk factor for preterm low birth weight. J Periodontol, 1996; 67(10): 1103-1113.
    26. Xu DX, Chen YH, Zhao L, et al. Reactive oxygen species are involved in lipopolysaccharide-induced intrauterine growth restriction and skeletal development retardation in mice. Am J Obstet Gynecol, 2006; 195(6): 1707-1714.
    27. Rivera DL, Olister SM, Liu X, et al. Interleukin-10 attenuates experimental fetal growth restriction and demise. FASEB J. 1998; 12(2): 189-197.
    28. Uchida T, Kataoka M, Higuchi Y, et al. Involvement of CD14 in lipopolysaccharide-induced liver injury in mice pretreated with Propionibacterium acnes. Pathobiology, 2004; 71(5): 246-252.
    29. Peng Y, Gong JP, Liu CA, et al. Expression of toll-like receptor 4 and MD-2 gene and protein in Kupffer cells after ischemia-reperfusion in rat liver graft. World JGastroenterol, 2004; 10(19): 2890-2893.
    30. Wang H, Hirsch E. Bacterially-induced preterm labor and regulation of prostaglandin metablizing enzyme expression in mice: The role of toll-like receptor 4. Biol Reprod, 2003; 69(6): 1957-1963.
    31. Brass DM, Savov JD, Whitehead GS, et al. LPS binding protein is important in the airway response to inhaled endotoxin. J Allergy Clin Immunol, 2004; 114(3): 586-92.
    32. Lee JW, Paape MJ, Zhao X. Recombinant bovine soluble CD14 reduces severity of experimental Escherichia coli mastitis in mice. Vet Res, 2003; 34(3): 307-316.
    33. Miyake K. Innate recognition of lipopolysaccharide by Toll-like receptor4 / MD-2 and RP105 / MD-1. J Endotoxin Res, 2000; 6(5): 389-391.
    34. Rallabhandi P, Bell J, Boukhvalova MS, et al. Analysis of TLR4 polymorphic variants: new insights into TLR4/MD-2/CD14 stoichiometry, structure, and signaling. J Immunol, 2006; 177(1): 322-32.
    35. Shishodia S,Koul D,Aggarwal BB. Cyclooxygenas(eCOX)-2 inhibitor celecoxib abrogates TNF-induced NF-kappa B activation through inhibition of activation of I kappa B alpha kinase and Akt in human non-small cell lung carcinoma:correlation with suppression of COX-2 synthesis. J Immunol, 2004, 173(3): 2011-2022.
    36. Wheeler MD, Yamashina S, Froh M, et al. Adenoviral gene delivery can inactivate Kupffer cells: role of oxidants in NF-kappaB activation and cytokine production. J Leukoc Biol, 2001, 69(4): 622-630.
    37. Bautista AP, Meszaros K, Bojta J, et al. Superoxide anion generation in the liver during the early stage of endotoxemia in rats. J Leukoc Biol, 1990; 48(2): 123-128.
    38. Park HS, Jung HY, Park EY, et al. Cutting edge: direct interaction of TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced production of reactive oxygen species and activation of NF-kappa B. J Immunol,2004; 173(6): 3589-3593.
    39. Rhee SH, Hwang D. Murine TOLL-like receptor 4 confers lipopolysaccharide responsiveness as determined by activation of NF kappa B and expression of the inducible cyclooxygenase. J Biol Chem, 2000; 275(44): 34035-34040.
    40. Tsan MF, Clark RN, Goyert SM, et al. Induction of TNF-alpha and MnSOD by endotoxin: role of membrane CD14 and Toll-like receptor-4. Am J Physiol Cell Physiol, 2001; 280(6): C1422-1430.
    41. Liu Y, Dong W, Chen L, et al. BCL10 mediates lipopolysaccharide/toll-like receptor-4 signaling through interaction with Pellino2. J Biol Chem, 2004; 279(36): 37436-37444.
    42. Gross G, Imamura T, Vogt SK, et al. Inhibition of cyclooxygenase-2 prevents inflammation-mediated preterm labor in the mouse. Am J Physiol Regul Integr Comp Physiol, 2000; 278(6): R1415-1423.
    43. Kim SH, Kim J, Sharma RP. Inhibition of p38 and ERK MAP kinases blocks endotoxin-induced nitric oxide production and differentially modulates cytokine expression. Pharmacol Res, 2004; 49(5): 433-439.
    44. Zingarelli B, Hake PW, Denenberg A, et al. Sesquiterpene lactone parthenolide, an inhibitor of IkappaB kinase complex and nuclear factor-kappaB, exerts beneficial effects in myocardial reperfusion injury. Shock, 2002; 17(2): 127-134.

© 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