围产期食物限制对大鼠神经行为发育及空间学习与记忆能力的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的
     成年和老年食物限制(food restriction, FR)可以产生许多有益的生物学效应,包括延长了生命周期、延缓衰老和疾病的进程、提高学习和记忆能力及降低药物的毒性等。然而,围产期食物限制可导致胎儿宫内发育受限(intrauterine growth restriction, IUGR),主要表现为新生儿出生后体重降低、生长缓慢、学习成绩不良。实验动物研究表明,围产期食物限制或营养不良除导致体重降低、生长缓慢外,还表现为一些行为发育延迟及成年社会行为和空间学习能力下降等。由于围产期营养不良对人类的生命和健康有极大危害,因此研究其对大鼠出生后神经行为发育(发育转折点)及对认知能力的影响有利于进一步认识人类IUGR并为发展保护人类胎儿IUGR的策略提供有益的资料。本研究首先建立围产期50%食物限制(FR50) Wistar大鼠模型,之后分别观察围产期FR50对新生期子代雌雄大鼠神经行为发育(包括身体发育和神经反射)的影响。由于新生期神经行为发育的延迟或损伤往往与后期(包括幼年和成年)的认知行为下降有关,因此又研究了子代成年雄性大鼠空间学习与记忆能力、海马CA1突触可塑性及海马一氧化氮合酶(nitric oxide synthase, NOS)阳性神经元密度的变化,以揭示认知行为变化的可能机制。
     方法
     实验第一部分:围产期50%食物限制大鼠模型的建立。为了研究围产期营养不良对神经行为发育和认知能力的影响,需要有合适的动物模型。通常有蛋白限制法和食物限制法。鉴于前者在饲料制作方面困难,所以采用后者制作模型。根据食物限制的程度,通常又分为轻度(30%)、中度(50%)和重度(70%)食物限制法。本研究采用了中度食物限制法。健康成年(体重250-280 g)14周龄wistar雌鼠与雄鼠同笼(雌雄比2-3:1)。之后,每日早晨检查雌鼠阴道口或阴道涂片,发现阴道栓或阴道涂片中有大量精子存在记为大鼠妊娠第0天(embryonic day 0,E0),分笼喂养。将妊娠的雌鼠随机分为对照组(n=18)和模型组(n=16)。对照组自由摄食,并记录每日饲料消耗量。模型组从妊娠的E0到E6自由摄食,从妊娠的E7到仔鼠出生的21天(postnatal day 21, PD21)进行食物限制,食物限制量为对照组大鼠每日平均饲料消耗量的50%。两组孕鼠均予充足饮水。分别于妊娠的E1、E7、E14和E21测定孕鼠体重。大鼠自然分娩,仔鼠出生的当天记为PD0,分别于仔鼠出生的PD1、PD7、PD10、PD14和PD21称重。发现FR50新生仔鼠体重明显低于正常组新生鼠,且孕鼠E14和E21体重也明显低于正常组孕鼠。表明本实验方法模拟了围产期营养不良状态,成功复制出胎鼠IUGR,有助于下一步研究新生子代大鼠神经行为发育及围产期营养不良状态对认知能力的影响。
     实验第二部分:围产期食物限制对子代新生Wistar大鼠神经行为发育的影响。以肛殖距判断仔鼠的性别。典型的神经行为发育评估包括身体生长和神经反射两部分。身体生长包括开耳(PD2-PD5)、睁眼(PD14-PD17)、门齿生长(PD8-PD12)和毛发出现(PD11-PD14)。神经反射包括翻正反射(PD3-PD7)、负性趋地反射(PD6-PD10)和悬崖回避反射(PD4-PD9)。比较对照组和食物限制组新生仔鼠神经发育的变化。
     实验第三部分:围产期食物限制对子代成年雄性大鼠空间学习与记忆能力及海马CA1区长时程增强(long-term potentiation,LTP的影响。大鼠出生后70天(PD70).运用Morris水迷宫测试空间学习与记忆能力。Morris水迷宫测试包括定位航行实验(place navigation test)和空间探索(probe trial performance)两个部分。定位航行实验历时5天,每天分上下午两个时间段训练。。在每个时间段训练4次,分别从4个坐标象限的中点将大鼠面向池壁轻轻放入水中,记录大鼠在120 s找到平台的时间,即逃避潜伏期(escape latency)。若在120 s内找不到平台,则将大鼠引导到平台上并保持15 s。第6天撤除平台作空间探索实验,选与平台象限相对象限的中点为入水点将大鼠放入水中,记录120 s内大鼠跨越平台的次数及在目标象限的搜索时间百分比。之后,在海马CA1区在体记录场兴奋性突触后电位(fEPSP).单极记录电极垂直插入海马CA1区锥体细胞树突丛(前囟后3.4 mm,中线旁开2.5mm),双极刺激电极插入同侧海马Schaffer侧枝(前囟后4.2 mm,中线旁开3.5 mm)。调整记录电极及刺激电极深度,至记录到最大场兴奋性突触后电位(field excitatory postsynaptic potential, fEPSP)为止海马CA1区LTP区的记录参数为:以波宽0.2ms、频率0.033Hz的测试刺激,作用于大鼠海马CA3区Schaffer侧枝,诱导海马CA1区锥体细胞树突丛场兴奋性突触后电位(field excitatory postsynaptic potential, fEPSP)。以fEPSP幅度的50%电流强度作为最适刺激强度。记录30 min fEPSP后进行高频刺激。高频刺激由10串200 Hz的方波组成,串间隔2 s。之后,用相同的波宽(频率0.033Hz)记录120 min fEPSP。
     实验第四部分:围产期食物限制对子代成年雄性大鼠海马神经元型一氧化氮合酶(neuronal nitric oxide synthase, nNOS)阳性细胞密度的影响。由于空间学习与记忆能力的变化及海马CA1区LTP的诱导和维持需要兴奋性神经递质谷氨酸的释放。一氧化氮(nitric oxide, NO)作为一种逆向信使,促进突触前神经元谷氨酸的释放,从而易化了在突触后神经元上记录的LTP。因此,进一步用免疫组化方法观察海马三个亚区:nNOS阳性细胞的密度。以期从一氧化氮的角度研究围产期食物限制消弱了子代空间学习和记忆能力的机制。
     结果
     1)围产期食物限制导致新生雌雄大鼠身体生长延迟,包括开耳、门齿生长、毛发和睁眼延迟。
     2)围产期食物限制导致雌雄新生大鼠神经反射延迟,包括翻正反射和悬崖回避反射。另外,食物限制还导致雄性新生大鼠负性趋地反射延迟,但不影响雌性新生大鼠负性趋地反射。
     3)围产期食物限制导致成年雄性子代大鼠空间学习与记忆能力下降。
     4)围产期食物限制导致成年雄性子代大鼠海马CA1区突触可塑性降低。
     5)围产期食物限制降低了海马三个亚区(CA1区、CA3区和齿状回)nNOS阳性细胞的密度。
     结论
     1)围产期食物限制导致新生大鼠神经发育延迟;
     2)围产期食物限制对新生大鼠小脑的发育可能具有性别依赖性;
     3)围产期食物限制可能通过抑制NO的产生降低了海马突触可塑性,从而导致空间学习与记忆能力的降低。
Objective
     Food restriction can induce the beneficial effects. It extends life span, reduces and retards the incidence of several diseases, increases learning and memory ability and attenuates the toxicological effects of drugs. Howerver, perinatal food restriction induces intrauterine growth restriction (IUGR), with neonatal babies at decreased body weight, childhood stunting and poor school achievement Perinatal food restriction or malnutrition can also induce neurodevelopmental delay, abnormal social behaviors and impaired spatial learning and memory ability besides decreased body weight and childhood stunting. Because perinatal malnutrition exerts harmful effects on human life and health, we studied the effect of perinatal food restriction on neurobehavioral development (neurodevelopmental milestones) and congnition ability. Our studies help further to recognize human fetal IUGR and provide beneficial information on developing neuroprotective therapies of human fetal IUGR. To address this question, a model of perinatal 50% food restriction (FR50) was induced in Wistar rats. The effect of perinatal FR50 on neurobehavioral development was observed, including physical growth and neurological reflexes. Since impaired or delayed neurobehavioral development is usually related to later recognition behaviors in young and adults, we studied spatial learning and memory abilities of adult offspring rats. In addition, in order to explore the possible mechanism of impaired spatial learning and memory abilities, we also observed synaptic plasticity and the density of nitric oxide synthase neurons in the hippocampus.
     Methods
     The first part of the experiments:Establishment of animal model with perinatal FR50. To study the effect of perinatal malnutrition on neurobehavioral development and spatial learning and memory abilities, establishment of animal model with perinatal FR50 was necessary. Generally, there are two ways, one is to restrict protein content, the other is to restrict food content. Due to difficulties of rat chow preparation on protein restriction, we apply the way of restricting food content. According to the extent of food restriction, there are three ways:slight, mild and severe food restriction. In the present study, we used the mild food restriction. Two or three healthy female rats (body weight 250-280 g,14-week old) were paired with one male (2-3:1) until mating was confirmed by observation of a copulatory plug or the presence of sperm in a vaginal rinse. The day that mating was confirmed and recorded as embryonic day 0 (E0). Each dam was singly housed. The day of birth was identified as postnatal day 0 (PDO). Afterwards, The pregnant rats were assigned radomly into two groups:the control group (n=18) and the model group (n=16). In the control group, dams had free access to diet and water during gestation and lactation (average amount of food consumed by control was recorded). In the model group, dams received normal food intake from gestation day 0 to 6, and 50% of the daily food intake of control mothers from gestation day 7 until the postnatal day 21. Water were available ad libitum in both control and FR50 rats. The body weight of dams was measured on El, E7, E14 and E21. Dams delivered spontaneously and the day of delivery was designated as PDO. The body weight of offspring rats was measured on PD1, PD7. PD10, PD14 and PD21. The result showed that FR50 reduced the body weight of newborn pups and the body weight of FR50 dams during E14 and E21. The present model mimics perinatal malnutrition and models fetal IUGR. The present study helps to further explore neurobehavioral development and cognition ability.
     The second part of the experiments:Effects of perinatal food restriction on neurobehavioral development in newborn Wistar rats. Gender was distinquish between male and females according to the anogenital distance. Neurodevelopmental assessment typically includes analysis of physical growth and neurological reflexes. The parameters of physical growth include timing of pinna detachment (PD2-PD5), eye opening (PD14-PD17), teeth eruption (PD8-PD12) and hair growth (PD11-PD14). Neurological reflexes include righting reflexes (PD3-PD7), negative geotaxis (PD6-PD10) and cliff avoidance reflex (PD4-PD9). The changes of neurobehavior development were observed between control and FR50 offsprings.
     The third part of the experiments:Effects of perinatal food restriction on spatial learning and memory ability as well as long-term potentiation in hippocampal CA1 area in adult offspring rats. On PD70, spatial learning and memory ability of rats was tested in Morris water maze (MWM). MWM task consists of two phases, place navigation test and spatial probe performance. In place navigation test, the animals were subjected to two sessions (morning and afternoon) of four trials per day for 5 consecutive days. In each trial, rats were gently released into water, facing the wall of the tank, at one of the four starting points. The trial starting points were the middle of each quadrant edge. Rats were allowed to swim freely until they climbed on the platform. The swimming time to reach the platform (escape latency from start to goal) for each trial was recorded. If they failed to locate the platform within 120s, they were placed on it for 15 seconds. After the place navigation test, the platform was removed and the same rats were used in a spatial probe test to determine memory retention. Rats were released into water at the position opposite to the target quadrant and allowed to swim for 120s. The time percent in target quadrant and the number of platform location crossings were measured. Afterwards, the in vivo field excitatory postsynaptic potential (fEPSP) was observed in the hippocampal CA1 area. Adult wistar rats were implanted with a monopolar recording electrode in the stratum radiatum of the CA1 area (3.4 mm posterior to bregma and 2.5 mm lateral to the midline), and a bipolar stimulating electrode in the Schaffer collaterals of the dorsal hippocampus (4.2 mm posterior to bregma and 3.5 mm lateral to midline) via holes drilled through the skull. The optimal depth of the electrode in the stratum radiatum of the CA1 area of the dorsal hippocampus was determined by the maximal response to the Schaffer collateral pathway stimulus. The fEPSP slope was recorded at frequence of 0.033Hz (30 s interval) by delivering a single current pulse (0.2 ms in duration) to the Schaffer collateral pathway. The stimulus intensity was adjusted to give an fEPSP amplitude of 50% of maximum. Each experiment consisted of a baseline measurement taken for 30 min, followed by a further measurement of evoked responses (120 min) after high frequency stimulation (HFS) application. HFS consisted of ten trains of 20 stimuli at 200Hz with 2 s intertrain interval, with the same stimulation intensity (0.033Hz) used for the basline recordings. fEPSP slope was analysed by Clampfit 9.0 softwale.
     The forth part of the experiments:Perinatal food restriction impaired spatial learning and memory behaviour and decreased the density of nitric oxide synthase neurons in the hippocampus of adult male rat offspring. Excitory neurotransmitters, such as glutamic acid, are necessary to learning and memory as well as consolidation and maintenance of LTP. Nitric oxide (NO) facilitates LTP in postsynaptic neuronal cells by promoting release of glutamic acid as a retrograde messenger. Therefore, the density of nitric oxide synthase neurons in the hippocampus of adult male rat offspring was studied using immunohistochemistry method. The aim is to study the possible mechanisms of impairment of spatial learning and memory ability by detecting the change of NO.
     Results
     1) Perinatal food restriction delayed physical growth, such as pinna detachment, hair growth, eruption of incisor teeth and eye opening.
     2) Perinatal food restriction delayed neurological reflexes in surface righting reflex and cliff avoidance reflex. In addition, it also exhibited a delay in achieving negative geotaxis response in male pups but not in female pups.
     3) Perinatal food restriction induced spatial learning and memory deficits in adult male rat offspring.
     4) Perinatal food restriction impaired synaptic plasticity in hippocampal CA1 area of adult male rat offspring.
     5) Perinatal food restriction decreased the density of nitric oxide synthase neurons in the hippocampus (CA1 area, CA3 area and dentate gyrus) of adult male rat offspring.
     Conclusion
     1) Perinatal food restriction delayed neurobehavior development in newborn rats.
     2) The effect of perinatal food restriction on the development of cerebellum is gender-dependent.
     3) Perinatal food restriction impaired synaptic plasticity in hippocampus by reducing the content of NO. Finally, it induced spatial learning and memory deficits.
引文
[1]Peterson BS, Anderson AW, Ehrenkranz R, et al. Regional brain volumes and their later neurodevelopmental correlates in term and preterm infants. Pediatrics.2003,111:939-48.
    [2]Seghier ML, Lazeyras F, Zimine S, et al. Combination of event related fMRI and diffusion tensor imaging in an infant with perinatal stroke. Neuroimage.2004,21:463-72.
    [3]Nelson CA, Monk CS. The use of event-related potentials in the study of cognitive development. In:Nelson CA, Luciana M, eds. Handbook in developmental cognitive neuroscience. Cambridge, MA:MIT Press,2001:125-36.
    [4]Davis EP, Townsend E, Gunnar M, et al. Effects of prenatal betamethasone exposure on regulation of stress physiology in healthy premature infants. Psychoneuroendocrinology. 2004,29:1028-36.
    [5]Armony-Sivan R, Eidelman A, Lanir A, et al. Iron status and neurobehavioral development of premature infants. J Perinatol.2004,24:757-62.
    [6]Freeman HE, Klein RE, Townsend JW, et al. Nutrition and cognitive development among rural Guatemalan children. Am J Public Health.1980,70:1277-85.
    [7]Simeon DT, Grantham-McGregor SM. Nutritional deficiencies and children's behaviour and mental development. Nutr Res Rev.1990,3:1-24.
    [8]Lucas A, Morley R, Cole TJ, et al. A randomised multicentre study of human milk versus formula and later development in preterm infants. Arch Dis Child.1994.70:F141-6.
    [9]Lucas A, Morley R. Breast milk and subsequent intelligence quotient in children born preterm. Lancet.1991,339(26):1-4.
    [10]Hoefer C, Hardy MC. Later development of breast fed and artificially fed infants. JAMA. 1929,92:615-9.
    [11]Gale CR, Martyn CN. Breast feeding, dummy use and adult intelligence. Lancet.1996,347: 1072-5.
    [12]Fierro-Beniter R, Casar R, Stanbury J et al. Long-term effects of correction of iodine deficiency on psychomotor development and intellectual development. In:Dunn J, Pretell E, Dara C, Viten F (Eds) Towards the Eradication of Endemic Goiter, Cretinism and Iodine Deficiency. Washington DC:Pan American Health Organization,1986.
    [13]Kar BR,Rao SL,Chandramouli BA.Cognitive development in children with chronic protein energy malnutrition. Behavioral and Brain Functions.2008,24,4:31.
    [14]Joos S, Pollitt E, Meuller W, et al. The Bacon Chow study:maternal nutritional supplementation and infant behavioral development. Child Dev.1983,54:669-76.
    [15]Grantham-McGregor SM, Powell CA, Walker SP, et al. Nutritional supplementation, psychosoaal stimulation, and mental development of stunted children, the Jamaican study Lancet.1991,338:1-5.
    [16]Lawrence PR, Pawel PB, Kimberly AV, et al. Placental angiogenesis in sheep models of compromised pregnancy. J Physiol.2005,565(1):43-58.
    [17]D Rice, S Barone Jr. Critical periods of vulnerability for the developing nervous system: evidence from humans and animal models. Environ Health Perspect.2000,108:511-533.
    [18]PJ Morgane, R Austin-LaFrance. J Bronzino, et al. Prenatal malnutrition and development of the brain. Neurosci Biobehav Rev.1993,17:91-128.
    [19]GeorgieffMK, Rao R. The role of nutrition in cognitive development. In:Nelson CA, Luciana M, eds. Handbook in developmental cognitive neuroscience. Cambridge, MA:MIT Press,2001:491-504.
    [20]Dobbing J. Vulnerable periods in the developing brain. In:Dobbing J, ed. Brain, behavior and iron in the infant diet. London, United Kingdom:Springer,1990,1-25.
    [21]Rao R, GeorgieffMK.Early nutrition and brain development. In:Nelson CA, ed. The effects of early adversity on neurobehavioral development. Minnesota Symposium on Child Psychology. Vol 31. Hillsdale. NJ:Erlbaum Associates,2000,1-30.
    [22]Kretchmer N, Beard JL. Carlson S. The role of nutrition in the development of normal cognition.Am J Clin Nutr.1996,63(suppl):997S-1001S.
    [23]Thompson RA, Nelson CA. Developmental science and the media:early brain development. Am Psychol 2001,56:5-15.
    [24]Winick M, Rosso P. The effect of severe early malnutrition on cellular growth of the human brain. Pediatr Res.1969:3:181-184.
    [25]Winick M. Nobel A. Cellular responses in rats during malnutrition at various ages. J Nutr. 1966.89:300-6.
    [26]Levitsky DA. Strupp BJ. Malnutrition and the brain:changing concepts, changing concerns. J Nutr.1995.125:2212S-20S
    [27]Gressens P, Muaku SM, Besse L, et al. Maternal protein restriction early in rat pregnancy alters brain development in the progeny. Brain Res Dev Brain Res.1997,20;103(1):21-35.
    [28]Bonatto F. Polydoro M. Andrades ME, et al. Effects of maternal protein malnutrition on oxidative markers in the young rat cortex and cerebellum. Neuroscience Letters.2006, 406(3):281-4.
    [29]Barja-Fidalgo C, Souza EP, Silva SV, et al. Impairment of inflammatory response in adult rats submitted to maternal undernutrition during early lactation:role of insulin and glucocorticoid. Inflamm Res.2003,52(11):470-6.
    [30]DeMaeyer E, Adiels-Tegman M. The prevalence of anaemia in the world. World Health StatQ.1985,38:302-16.
    [31]Chockalingam UM, Murphy E, Ophoven JC, et al. Cord transferring and ferritin levels in newborn infants at risk for prenatal uteroplacental insufficiency and chronic hypoxia. J Pediatr.1987,111:283-6.
    [32]Georgieff MK, Landon MB, MillsMM, et al. Abnormal iron distribution in infants of diabetic mothers:spectrum and maternal antecedents. J Pediatr.1990,117:455-61.
    [33]Georgieff MK, Petry CE, Wobken JD, et al. Liver and brain iron deficiency in newborn infants with bilateral renal agenesis (Potter's syndrome). Pediatr Pathol.1996,16:509-19.
    [34]Petry CD, Eaton MA, Wobken JD, et al. Iron deficiency of liver, heart, and brain in newborn infants of diabetic mothers. J Pediatr.1992,121:109-14.
    [35]Rao R, Georgieff MK. Perinatal aspects of iron metabolism. Acta Pediatr Suppl.2002, 91:124-9.
    [36]Sandstead HH. Zinc:essentiality for brain development and function. Nutr Rev.1985, 43:129-37.
    [37]Duncan J, Hurley L. Thymidine kinase and DNA polymerase activity in normal and zinc deficient developing rat embryos. Proc Soc Exp Biol Med.1978,159:39-43.
    [38]McNall A, Etherton T, Fosmire G. The impaired growth induced by zinc deficiency in rats is associated with decreased expression of hepatic insulin-like growth factor Ⅰ and growth hormone receptor genes. J Nutr.1995,125:874-9.
    [39]Frederickson C, Danscher G. Zinc-containing neurons in hippocampus and related CNS structures. Prog Brain Res.1990,83:71-84.
    [40]Hesse G. Chronic zinc deficiency alters neuronal function of hippocampal mossy fibers. Science.1979,205:1005-7.
    [41]Golub MS, Takeuchi PT, Keen CL. et al. Modulation of behavioral performance of prepubertal monkeys by moderate dietary zinc deprivation. Am J Clin Nutr.1994,60: 238-43.
    [42]Merialdi M, Caulfield LE, Zavaleta N, et al. Randomized controlled trial of prenatal zinc supplementation and the development of fetal heart rate. Am J Obstet Gynecol.2004,190: 1106-12.
    [43]Prohaska JR, Gybina AA. Rat brain iron concentration is lower following perinatal copper deficiency. J Neurochem.2005,93:698-705.
    [44]West EC, Prohaska JR. Cu, Zn-superoxide dismutase is lower and copper chaperone CCS is higher in erythrocytes of copper-deficient rats and mice. Exp Biol Med (Maywood) 2004,229:756-64.
    [45]Gybina AA, Prohaska JR. Increased rat brain cytochrome c correlates with degree of perinatal copper deficiency rather than apoptosis. J Nutr.2003,133:3361-8.
    [46]Prohaska JR, Brokate B. Dietary copper deficiency alters protein levels of rat dopamine beta-monooxygenase and tyrosine monooxygenase. Exp Biol Med (Maywood).2001, 226:199-207.
    [47]Penland JG, Prohaska JR. Abnormal motor function persists following recovery from perinatal copper deficiency in rats. J Nutr.2004,134:1984-8.
    [48]San Giovanni JP, Parra-Cabrera S, Colditz GA, et al. Meta-analysis of dietary essential fatty acids and long-chain polyunsaturated fatty acids as they relate to visual resolution acuity in healthy preterm infants. Pediatrics.2000,105:1292-8.
    [49]O'Connor DL, Hall R, Adamkin D, et al. Growth and development in preterm infants fed long-chain polyunsaturated fatty acids:a prospective, randomized controlled trial. Pediatrics 2001,108:359-71.
    [50]Auestad N, Halter R, Hall RT, et al. Growth and development in term infants fed long-chain polyunsaturated fatty acids:a double-masked, randomized parallel, prospective, multivariate study. Pediatrics.2001,108:372-81.
    [51]Aghini-Lombardi FA, Pinchera A, Antonangeli T, et al. Mild iodine deficiency during fetal /neonatal life and neuropsychological impairment in Tuscany. J Endocrinol Invest.1995, 18:57-72.
    [52]Mitchell JH, Nicol F, Beckett GJ, Arthur JR. Selenoprotein expression and brain development in preweanling selenium and iodine deficient rats. J Mol Endocrinol.1998, 20:203-10.
    [53]Ferguson SA, Berry KJ, Hansen DK, et al. Behavioral effects of prenatal folate deficiency in mice. Birth Defects Res A Clin Mol Teratol.2005,4:249-52.
    [54]Zeisel SH. Niculescu MD. Perinatal choline influences brain structure and function. Nutr Rev. 2006,64:197-203.
    [55]Chen Z, Schwahn BC, Wu Q, et al. Postnatal cerebellar defects in mice deficient in methylenetetrahydrofolate reductase. Int J Dev Neurosci.2005.23(5):465-74.
    [56]林良明.刘玉琳,张新利.米杰,曹兰华.中国低出生体重儿抽样调卉结果.中华预防医学杂志.2002.36:149-153.
    [57]Martin Gronert MS, Ozanne SE. Experimental IUGR and later diabetes.J Intern Med, 2007.261(5):437-52.
    [58]Ravelli AC, van Der Meulen JH, Osmond C, et al. Obesity at the age of 50 y in men and women exposed to famine prenatally. Am J Clin Nutr,1999,70(5):811-6.
    [59]Stein AD, Kahn HS, Rundle A, et al. Anthropometric measures in middle age after exposure to famine during gestation:evidence from the Dutch famine. Am J Clin Nutr,2007, 85(3):869-76.
    [60]Hart N. Famine, maternal nutrition and infant mortality:a re-examination of the Dutch hunger winter. Popul Stud (Camb).1993,47(1):27-46.
    [61]赵文华,杨正雄,翟屹,等.生命早期营养不良成年后超重和肥胖患病危险影响的研究.中华流行病学杂志,2006,27(8):647-650.
    [62]Yang Z, Zhao W, Zhang X, et al. Impact of famine during pregnancy and infancy on health in adulthood. Obes Rev.2008,9 (Suppl 1):95-9.
    [63]Kind KL, Clifton PM, Grant PA, et al. Effect of maternal feed restriction during pregnancy on glucose tolerance in the adult guinea pig. Am J Physiol Regul Integr Comp Physiol. 2003,284(1):R140-52.
    [64]Garofano A, Czernichow P, Breant B. Beta-cell mass and proliferation following late fetal and early postnatal malnutrition in the rat. Diabetologia.1998,41 (9):1114-20.
    [65]Petrik J, Reusens B, Arany E, 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. Endocrinology.1999, 140(10):4861-73.
    [66]Park KS, Kim SK, Kim MS, et al. Fetal and early postnatal protein malnutrition cause long-term changes in rat liver and muscle mitochondria. J Nutr.2003,133 (10):3085-90.
    [67]Lee HK, Song JH, Shin CS, et al. Decreased mitochondrial DNA content in peripheral blood precedes the development of non-insulin-dependent diabetes mellitus. Diabetes Res Clin Pract.1998,42(3):161-7.
    [68]Desai M, Byrne CD, Meeran K, et al. Regulation of hepatic enzymes and insulin levels in offspring of rat dams fed a reduced-protein diet. Am J Physiol.1997,273 (4 Pt 1):G899-904.
    [69]Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus:the thrifty phenotype hypothesis. Diabetologia.1992,35:595-601.
    [70]Gluckman PD, Hans onMA. Living with the past:evolution, development and patterns of disease. Science.2004,305:1773-1776.
    [71]Gluckman PD, Hans on MA. The developmental origins of the metabolic syndrome. Trends Endocrin Met.2004,15:183-187.
    [72]Gluckman PD, Hanson MA. Maternal constraint of fetal growth and its consequences. Semin Fetal Neonat Med.2004,9:419-425.
    [73]Gluckman PD, Cutfield W, Hofman P, et al. The fetal, neonatal and infant environments the long term consequences for disease risk. Early Hum Dev.2005,81:51-59.
    [74]Le Douarin NM. Embryonic neural chimaeras in the study of brain development. Trends Neurosci.1993,16(2):64-72.
    [75]Gressens P, Richelme C, Kadhim HJ, et al. The genninative zone produces the most cortical astrocytes after neuronal migration in the developing mammalian brain. Bio Neonate.1992, 61(1):4-24.
    [76]Brown NA, Goulding EH, Fabro S. Ethanol embryotoxicity:direct effects on mammalian embryos in vitro. Science.1979,206(4418):573-5.
    [77]Gofflot F, Nassogne MC, Etzion T, et al. In vitro neuroteratogenicity of valproic acid and 4-en-VPA. Neurotoxicol Teratol.1995,17(4):425-35.
    [78]Zeng Hui, Shu Wei-qun, Zhao Qing, et al. Reproductive and neurobehavioral outcome of drinking purified water under magnesium deficiency in the rat's diet. Food Chem Toxicol. 2008,46(5):1495-502.
    [79]Carney EW, Zablotny CL, Marty MS, et al. The effects of feed restriction during in utero and postnatal development in rats. Toxicol Sci.2004,82(1):237-249.
    [80]O'Rahilly R, Muller F. Significant features in the early development of the human brain. Ann Anat.2008,190:105-118.
    [81]Herlenius E, Lagercrantz H. Development of neurotransmitter systems during critical period. Exp Nerol.2004,190:S8-S21.
    [82]Casper RC. Nutrients, neurodevelopment and mood. Curr Psychiatry Res.2004,6:625-629.
    [83]Cannon M, Clarke MC. Risk for schizophrenia-broadening the concepts, pushing back the boundaries. Schizophr Res.2005,79:5-13.
    [84]Langley R, Rice F, van den Bree MB, et al. Maternal smoking during pregnancy as an environmental risk factor for attention defi cit hyperactivity disorder behaviour. A review. Minerva Pediatr.2005,57:359-371.
    [85]Kinney DK, Munir KM, Crowley DJ, et al. Prenatal stress and risk for autism. Neurosci Biobehav Res.2008,32:1519-1523.
    [86]EPA. Health Effects Test Guidelines, OPPTS 870.6300, Developmental Neurotoxicity Study, EPA.1998,712-C-98-239.
    [87]OECD. Guideline for the testing of chemicals, Draft proposal for a new guideline 426, Developmental Neurotoxicity Study,1995.
    [88]Fox WM. Reflex ontogeny and behavioral development of the mouse. Anim Behav.1965, 13:234-241.
    [89]Petruzzi S. Fiore M. Dell'Omo G. et al. Medium and longterm behavioral eff ects in mice of gestational exposure to ozone. Neurotoxicol Teratol.1995.17:463-470.
    [90]Rayburn WF,Christenen HD,Gonzales GL. A placebo-controlled comparison between butamethasone and dexamethasone for fetal maturation:differences in neurobehavioral development of mice off spring. Am J Obstet Gynecol.1997,176:842-851.
    [91]Slikker W,Chang LW. Hanbook of developmental neurotoxicology. Academic Press,San Diego,USA,1998,1-748.
    [92]Dubovicky M, Ujhazy E, Kovacovsky P, et al. Effect of long-term administration of stobadine on exploratory behaviour and on striatal levels of dopamine and serotonin in rats and their off spring. J Appl Toxicol 1997a.17:63-70.
    [93]Dubovicky M, Tokarev D, Skultetyova I, et al. Changes of exploratory behaviour and its habituation in rats neonatally treated with monosodium glutamate. Pharmacol Biochem Behav,1997b,6:565-569.
    [94]Dubovicky M, Skultetyova I, Jezova D. Neonatal stress alters habituation of exploratory behavior in adult male but not female rats. Pharmacol Biochem Behav.1999b,64:681-686.
    [95]Dubovicky M, Ujhazy E, Kovacovsky P, et al. Evaluation of long-term administration of the antioxidant stobadine on exploratory behavior in rats of both genders. J Appl Toxicol 1999a, 19:431-436.
    [96]Dubovicky M,Jezova D. Eff ect of chronic emotional stress on habituation processes in open field in adult rats. Ann NY Acad Sci.2004,1018:199-206.
    [97]Dobbing J, Sands J. Comparative aspects of the brain growth spurt. Early Human Dev.1979, 3:79-83.
    [98]West JR. Use of pups in a cup model to study brain development. J Nutr.1993,123:382-385.
    [99]Accuf KD, Vorhees CV. Neurobehavioral Teratology. In:Introduction to Neurobehavioral Toxicology:Food and Environment, Niesink RJM, Jaspers RMA,Kornet LMW,van Ree JM, Tilson HA (eds). CRC Press, New York,1998,27-69.
    [100]Lanfumey L, Mongeau R, Cohen-Salmon C, et al. Corticosteroid-serotonin interaction in the neurobiological mechanisms of stress-related disorders. Neurosci Biobehav Rev.2008. 32:1174-1184.
    [101]Barone S, Stanton ME, Mundy WR. Neurotoxic effects of neonatal triethyltin (TET) exposure are exacerbated with aging. Neurobiol Aging.1995,16:723-735.
    [102]Weitzdoerfer R, Gerstl N, Hoeger H, et al. Long-term sequele of perinatal asphyxia in aging rats. Cell Mol Life Sci.2002,59:519-526.
    [103]Barlow BK, Cory-Slechta DA, Richfi eld EK, et al. The gestational environment and Parkinson's disease:evidence for neurodevelopmental origins of neurodegenetative disorders. Reprod Toxicol.2007,23:457-570.
    [104]Sayer AA, Cooper C, Barker DJ. Is lifespan determined in utero? Arch Dis Child Fetal Neonatal.1997,77:162-164.
    [105]Bezek S, Mach M, Ujhazy E, et al. Nongenomic memory of foetal history in chronic diseases development. Neuroendocrinol Lett.2008,29:620-626.
    [106]SM Gabriel, JR Roncancio. NS Ruiz. Growth hormone pulsatility and the endocrine milieu during sexual maturation in male and female rats. Neuroendocrinology.1992,56:619-625.
    [107]McEwen BS. Lieberburg I. Chaptal C, et al. Aromatization:important for sexual differentiation of the neonatal rat brain. Horm Behav.1977,9:249-263.
    [108]Selmanoff MK, Brodkin LD, Weiner RI, et al. Aromatization and 5alpha-reduction of androgens in discrete hypothalamic and limbic regions of the male and female rat. Endocrinology.1977.101:841-848.
    [109]Jones HE. Ruscio MA, Keyser LA, et al. Prenatal stress alters the size of the rostral amerior commissure in rats. Brain Res Bull.1997.42:341-346.
    [110]Gorski RA, Gordon JH, Shryne JE, et al. Evidence for a morphological sex difference within the medial preoptic area of the rat brain. Brain Res.1978,148:333-346.
    [111]刘镇伟.实用膜片钳技术.军事医学科学出版社.1-6.
    [112]李振彪,吴希如,姜玉武.发育中的海马神经元离子通道功能变化.实用儿科临床杂志.2006,21(11):694-695.
    [113]Baso AC, Goulart FC, Teodorov E, et al. Effects of maternal exposure to picrotoxin during lactation on physical and reflex development, square crossing and sexual behavior of rat offspring. Pharmacol. Biochem. Behav.2003,75:733-740.
    [114]de Castro VL, Chiorato SH, Pinto NF. Relevance of developmental testing of exposure to methamidophos during gestation to its toxicology evaluation. Toxicol Lett.2000,118: 93-102.
    [115]Mesquita AR, Pego JM, Avielle TS, et al. Neurodevelopment milestones abnormalities in rats exposed to stress in early life. Neuroscience.2007,147:1022-1033.
    [116]Koizumi M, Nishimura N, Enami T, et al. Comparative toxicity study of 3-aminophenol in newborn and young rats.J Toxicol Sci.2002,27:411-421.
    [117]Coluccia A, Belfiore D, Bizzoca A, et al. Gestational all-trans retinoic acid treatment in the rats:neurofunctional changes and cerebellar phenotype.Neurotoxicol. Teratol.2008,30: 395-403.
    [118]Taylor Dj, Howie PW. Fetal growth achievement and neurodevelopmental disability. Br J Obstet Gynaecol.1989,96:789-794.
    [119]Chang SM, Walker SP, Grantham-McGregor S, et al. Early childhood stunting and later behaviour and school achievement. J Child Psycho! Psychiatry.2002,43:775-783.
    [120]Almeida SS, Tonkiss J, Galler JR. Malnutrition and reactivity to drugs acting in the central nervous system. Neurosci Biobehav Rev.1996,20:389-402.
    [121]Morgane PJ. Mokler DJ, Galler JR. Effects of prenatal protein malnutrition on the hippocampal formation. Neurosci Biobehav Rev.2002,26:471-483.
    [122]Gallagher EA. Newman JP, Green LR, et al. The effect of low protein diet in pregnancy on the development of brain metabolism in rat offspring, J Physiol.2005,568:553-558.
    [123]Secher T, Novitskaia V, Berezin V, et al. A neural cell adhesion molecule-derived fibroblast growth factor receptor agonist,the fgl-peptide, promotes early postnatal sensorimotor development and enhances social memory retention. Neuroscience.2006, 141:1289-1299.
    [124]Jordan TC. Howells KF. Effects of early undernutrition on individual cerebellar lobes in male and female rats. Brain Res.1978.157:202-205.
    [125]Chan SY. Andrews MH. Lingas R. et al. Maternal nutrient deprivation induces sex-specific changes in thyroid hormone receptor and deiodinase expression in the fetal guinea pig brain. J Physiol.2005.566:467-480.
    [126]Kilby MD. Gittoes N_, McCabe C, et al. Expression of thyroid receptor isofoms in the human fetal central nervous system and the effects of intrauterine growth restriction. Clin Endocrinol (Oxf).2000.53:469-477.
    [127]Hintz SR. Kendrick DE. Vohr B, et al. Gender differences in neurodevelopmental outcomes among extremely preterm, extremely-low-birthweight infants. Acta Paediatr.2006, 95:1239-1248.
    [128]Messerschmidt A, Fuiko R, Prayer D, et al. Disrupted cerebellar development in preterm infants is associated with impaired neurodevelopmental outcome. Eur J Pediatr.2008,167: 1141-1147.
    [129]Nguon K, Ladd B and Sajdel-Sulkowska EM. Exposure to altered gravity during specific developmental periods differentially affects growth, development, the cerebellum and motor functions in male and female rats. Adv. Space Res.2006,38:1138-1147.
    [130]Sousa N. Almeida OF, Wotjak CT. A hitchhiker's guide to behavioral analysis in laboratory rodents. Genes Brain Behav,2006,5:5-24.
    [131]Spear, LP. Neurobehavioral assessment during the early postnatal period. Neurotoxico Teratol.1990.12:489-495.
    [132]Gayoso MJ, Primo C, Al-Majdalawi A. et al. Brain lesions and water-maze learning deficits after systemic administration of kainic acid to adult rats. Brain Research.1994,653:92-100.
    [133]Hass U, Lund S P, Simonsen L. et al. Effects of prenatal exposure to xylene on postnatal development and behavior in rats. Neurotoxicology and teratology.1995,17(3):341-349.
    [134]Hass U, Lund S P, Hougaard KS, et al. Developmental neurotoxicity after toluene inhalation exposure in rats. Neurotoxicology and Teratology.1999,21(4),349-357.
    [135]Hougaard KS, Hass U, Lund SP, et al. Effects of prenatal exposure to toluene on postnatal development and behavior in rats. Neurotoxicology And Teratology.1999,21(3):241-50.
    [136]Tilson, HA, Cabe, PA. and Spencer, PS. Behavioural procedures for the assessment of neurotoxicity. In:Experimental and Clinical Neurotoxicity. Eds. Spencer PS and Schaumburg HH, Williams & Wilkins, Baltimore,1980,p.758.
    [137]Pryor GT, Uyeno ET, Tilson HA, et al. Assessment of chemicals using a battery of neurobehavioural tests:A comparative study. Neurobehav Toxicol Teratol.1983,5:91.
    [138]Kanit L, Yilmaz O, Taskiran D, et al. Intersession interval affects performance in the Morris Water Maze. International Journal Of Neuroscience,1998,96:197-204.
    [139]Kikusui T. Tonohiro T, Kaneko T. Simultaneous evaluation of spatial working memory and motivation by the allocentric place discrimination task in the water maze in rats. Journal Of Veterinary Medical Science.1999.61 (6):673-81.
    [140]Cimadevilla JM, Gonzalez-Pardo H, Lopez L, et al. Sex-related differences in spatial learning during the early postnatal development of the rat. Behavioural Processes.1999, 46/2:159-171.
    [141]Hort J, Brozek G. Komarek V, et al. Interstrain differences in cognitive functions in rats in relation to status epilepticus. Behavioural Brain Research.2000,112 (1-2):77-83.
    [142]Bedi KS. Spatial learning ability of rats undernourished during early postnatal life. Physiology and Behavior.1992,51:1001-1007.
    [143]Shukitt-Hale B, Mouzakis G, Joseph JA. Psychomotor and spatial memory performance in aging male Fischer 344 rats. Experimental Gerontology.1998,33 (6):615-24.
    [144]Healy SD, Braham SR, Braithwaite VA. Spatial working memory in rats:no differences between the sexes. Proceedings Of The Royal Society Of London. Series B:Biological Sciences.1999,266 (1435):2303-8.
    [145]Hunter AJ, Roberts FF. The effects of pirenzepine on spatial learning in the Morris water maze. Pharmacol Biochem Behav.1988,30:519-523.
    [146]Panakhova E, Buresova O and Bures J. The effect of hypothermia on the rat's spatial memory in the water tank test. Behav Neural Bio.1984,42,191-196.
    [147]Daniel JM, Roberts SL, Dohanich GP. Effects of ovarian hormones and environment on radial maze and water maze performance of female rats. Physiology And Behavior.1999, 66(1):11-20.
    [148]Tees RC. The influences of sex, rearing environment, and neonatal choline dietary supplementation on spatial and nonspatial learning and memory in adult rats. Developmental Psychobiology.1999,35(4):328-42.
    [149]Robinson L, Bridge H, Riedel G. Visual discrimination learning in the water maze:a novel test for visual acuity. Behavioural Brain Research,2001,119(1):77-84.
    [150]Ruth Morley, Alan Lucas. Nutrition and cognitive development. British Medical Bulletin. 1997.53(1):123-134.
    [151]Wang L, Xu RJ. The effects of perinatal protein malnutrition on spatial learning and memory behaviour and brain-derived neurotrophic factor concentration in the brain tissue in young rats. Asia Pac J Clin Nutr.2007,16 (Suppl 1):467-72.
    [152]Andrade JP, Paula-Barbosa MM. Protein malnutrition alters the cholinergic and GABAergic systems of the hippocampal formation of the adult rat:an immunocytochemical study. Neurosci Lett.1996,211(3):211-215.
    [153]Partadiredja G. Simpson E. Bedi M. The effects of pre-weaning under nutrition on the expression levels of the radical deactivating enzymes in the mouse brain. Nutr Neurosci. 2005,8(3):183-193.
    [154]Chen JC, Tonkiss JR., Galler JR. et al. Prenatal malnutrition enhances serotonin release from the hippocampus.J Nutr.1992; 122 (Ⅱ):2138-2143.
    [155]Fontana L.The scientific basis of caloric restriction leading to longer life. Curr Opin Gastroenterol.2009.25(2):144-150.
    [156]Bonorden MJ. Rogozina OP. Kluczny CM. el al. Intermittent calorie restriction delays prostate tumor detection and increases survival time in TRAMP mice. Nutr Cancer.2009. 61(2):265-275.
    [157]Duan W. and Mattson MP. Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson's disease.J Neurosci Res.1999.57(2):195-206.
    [158]Yu ZF. Mattson MP. Dietary restriction and 2-deoxyglucose administration reduce focal ischemic brain damage and improve behavioral outcome:Evidence for a preconditioning mechanism.J Neurosci Res.1999.57(6):830-839.
    [159]Ingram. DK, Weindruch R, Spangler EL.et al. Dietary restriction benefits learning and motor performance of aged mice. J Gerontol,1987,42(1):78-81.
    [160]Stewart J, Mitchell J, Kalant N. The effects of life-long food restriction on spatial memory in young and aged Fischer 344 rats measured in the eight-arm radial and the Morris water mazes. Neurobiol Aging.1989,10(6):669-675.
    [161]Govic A, Kent S, Levay EA, et al. Testosterone, social and sexual behavior of perinatally and lifelong calorie restricted offspring. Physiol Behav,2008.94(3):516-522.
    [162]Ranade SC. Rose A, Rao M, et al. Different types of nutritional deficiencies affect different domains of spatial memory function checked in a radial arm maze. Neuroscience. 2008,152(4):859-866.
    [163]Okada T. Yamada N, Tsuzuki K. et al. Long-term otentiation in the hippocampal CA1 area and dentate gyrus plays different roles in spatial learning. Eur J Neurosci.2003. 17(2):341-349.
    [164]Remondes M, Schuman EM. Role for a cortical input to hippocampal area CA1 in the consolidation of a long-term memory. Nature.2004,431(7009):699-703.
    [165]Bliss TV, Collingridge GL. A synaptic model of memory:long-term potentiation in the hippocampus. Nature.1993,361 (6407):31-39.
    [166]Morgane PJ, Austin-LaFrance R, Bronzino J, et al. Prenatal malnutrition and development of the brain. Neurosci Biobehav Rev.1993,17(1),91-128.
    [167]Morgane PJ, Mokler DJ, Galler JR. Effects of prenatal protein malnutrition on the hippocampal formation. Neurosci Biobehav Rev.2002,26(4),471-483.
    [168]Palmer RMJ, Ferrige AG, Mocada S. Nitric oxide release accounts f or the biological activity of endothelium relaxing factor. Nature.1987,327:524-530.
    [169]Moncada S, Palmer RMJ, Higgs EA. Nitric oxide:physiology, pathophy siology a nd pharmacology.Pharmacol Rev.1991,43:109-112.
    [170]刘辉,陈俊抛,田时雨,等.神经元型一氧化氮合酶在学习记忆过程中的变化和作用.中国神经免疫学和神经病学杂志,2000,7(2):116-122.
    [171]Chen J, Zhang S, Zuo P, et al. Memory-related changes of nitric oxide synthase activity and nitrite level in rat brain. Neuroreport.1997,8(7):1771-1774.
    [172]张世仪,陈纪君,左萍萍,等.学习过程脑内一氧化氮变化的研究(简报).中国医学科学院学报.1997.19(1):17.
    [173]Halaris A, Piletz J. Agmatine:Metabolic pathway and spectrum of activity in brain. CNS Drugs.2007,21:885-900.
    [174]Arteni NS. Lavinsky D, Rodrigues AL, et al. Agmatine facilitates memory of an inhibitory avoidance task in adult rats. Neurobiol Learn Mem.2002,78:465-469.
    [175]Liu P, Rushaidhi M, Collie ND, et al. Behavioural effects of intracerebroventricular microinfusion of agmatine in adult rats. Behav Neurosci.2008.122:557-569.
    [176]Liu P, Chary S, Devaraj R, et al. Effects of aging on agmatine levels in memory-associated brain structures. Hippocampus.2008,18(9):853-6.
    [177]Liu P, Collie ND, Chary S, et al. Spatial learning results in elevated agmatine levels in the rat brain. Hippocampus.2008,18(11):1094-8.
    [178]Yamada K, Noda Y, Nakayama S, et al. Role of nitric oxide in learning and memory and in monoamine metabolism in the rat brain. Br J Pharmacol.1995,115(5):852-858.
    [179]Garthwaite J. Neural nitric oxide signaling. Trends Neurosci.1997,18(2):51-52.
    [180]Holscher C. Nitric oxide,the enigmatic neuronal messenger:its role in synaptic plasticity. Trends Neurosci.1997,20(7):298-303.
    [181]Zhou M, Small SA, Kandel ER, et al. Nitric oxide and carbon monoxide produce activity-dependent long-term synaptic enhancement in hippocampus. Science.1993,260 (5116):1949-1950.
    [182]Ward L, Mason SE, Abraham WC. Effects of the NMDA antagonists CPP and MK-801 on radial arm maze performance in rats. Pharmacol Biochem Behav.1990,35(4):785-790.
    [183]陆汉新,王革平,戴君勇,等.Mg2+对小鼠学习记忆的影响及其与海马CA3区和大脑皮层突触界面结构的相关性.神经科学,1996,3(3):109-113.
    [184]Molina JA, Jimenez JFJ, Orti PM, et al. The role of nitric oxide in neurodegeneraion. Potential for pharmacologicl intervention. Drugs Aging,1998,12(4):251-259.
    [185]张均田.我国神经药理学十年研究进展.中国药理学通报,1997.13(2):97-103.
    [186]Arancio O. Kiebler M, Lee CJ. et al. Nitric oxide acts directly in the presynaptic neuron to produce long-term potentiation in cultured hippocampal neurons. Cell.1996,87(6): 1025-1035.
    [187]Haley JE, Schaible E, Pavlidis P, et al. Basal and apical synap ses of CA1 pyramidal cells employ different LTP induction mechanisms. Learn Mem.1996,3(4):289-295.
    [188]Brenman JE, Chao DS, Gee SH. et al. Interaction of nitric oxide synthase with the postsynaptic density protein PSD-95 and alphal-syntrophin mediated by PDZ domains. Cell.1996,84(5):757-767.
    [189]Christopherson KS, Hillier BJ, Lim WA, et al. PSD-95 assembles a ternary complex with the N-methyl-D-aspartic acid receptor and a bivalent neuronal NO synthase PDZ domain. J Biol Chem.1999,274(39):27467-27473.
    [190]Sancesario G, Morello M, Reiner A, et al. Nitrergic neurons make synapses on dual-input dendritic spines of neurons in the cerebral cortex and the striatum of the rat:implication for a postsynaptic action of nitric oxide. Neuroscience.2000,99(4):627-642.
    [191]ValtschanoffJG. Weinberg RJ. Laminar organization of the NMDA receptor complex within the postsynaptic density.J Neurosci.2001,21(4):1211-1217.
    [192]Lu YF. Kandel ER. Hawkins RD. Nitric oxide signaling contributes to late-phase LTP and CREB phosphorylation in the hippocampus.J Neurosci.1999,19(23):10250-10261.