葛根素干预血管性痴呆模型大鼠海马组织低氧诱导因子1α明显下降
葛根素对大鼠颅脑损伤的保护作用
葛根素对大鼠颅脑损伤的保护作用李国亮;邸方;杨亚东【期刊名称】《中国临床神经外科杂志》【年(卷),期】2016(21)8【摘要】目的探讨葛根素对大鼠颅脑损伤(TBI)的保护作用及其机制。
方法将45只SD大鼠随机分为假手术组、模型组、低剂量葛根素组、中剂量葛根素组和高剂量葛根素组,每组9只。
采用Feeney氏自由落体法制备TBI大鼠模型。
低、中、高剂量葛根素组腹腔注射葛根素,剂量分别为10、25、50 mg/kg。
造模后1、3、7 d采用改良神经功能缺损评分(m NSS)评价神经功能。
造模后7 d,干湿法测定脑组织含水量;ELISA法检测脑组织丙二醛(MDA)、超氧化物歧化酶(SOD)、谷胱甘肽(GSH)、过氧化氢酶(CAT)、核因子κB(NF-κB)、细胞间黏附分子-1(ICAM-1)、白介素-6(IL-6)、肿瘤坏死因子-α(TNF-α)、caspase-3水平;免疫印迹法检测Bax、Bcl-2的表达。
结果葛根素能显著降低TBI大鼠m NSS(P<0.05),显著减轻脑组织水肿(P<0.05),显著降低脑组织MDA、SOD、GSH、CAT、NF-κB、ICAM-1、IL-6、TNF-α、caspase-3水平(P<0.05),显著下调Bax表达而上调Bcl-2表达(P<0.05)。
结论葛根素可通过减轻颅脑水肿、抑制氧化应激及炎性反应以及调节Bax/Bcl-2表达从而发挥神经保护作用。
【总页数】4页(P479-482)【关键词】颅脑损伤;氧化应激;炎症反应;细胞凋亡;大鼠;葛根素【作者】李国亮;邸方;杨亚东【作者单位】武汉市东西湖区人民医院神经外科;黄冈市中心医院重症医学科【正文语种】中文【中图分类】R651.15【相关文献】1.葛根素对离体大鼠缺血/复灌心脏的保护作用及其作用机制 [J], 潘红阳;高琴;姚慧;夏强2.移植转染大鼠骨髓基质干细胞对颅脑损伤大鼠的保护作用 [J], 张震宇;温二生;薛进华;鲁友明;曾艳;蒋秋华3.葛根素对肝硬化大鼠肝脏缺血再灌注损伤保护作用及其保护机制的研究 [J], 孙鹏;李五生;张楠;周小娜;刘金龙4.葛根素脂质微球注射液、葛根素注射液药动学及对局灶性脑缺血再灌注大鼠的神经保护作用的比较研究 [J], 汪春燕;陈绪龙;汪新娌;徐希强;周爱鲜;梁新丽5.葛根素磺酸钠与葛根素对大鼠急性心肌缺血的保护作用的比较研究 [J], 袁叶飞;胡祥宇;欧贤红因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素在正常和_淀粉样蛋白所致痴呆模型大鼠体内药代动力学比较_姚莹
医
参照 Giovannelli 的方法进行[5]: SD 雄性大鼠体重( 180
药
± 20) g,腹腔注射 3% 戊巴比妥钠 30mg·kg - 1 麻醉后,将头
部固定在脑立体定位仪上,剪去颅顶部毛,酒精消毒后,行
学 正中切开,分离皮下肌肉组织,用棉球搽净渗血。参照大鼠 刊 脑立体图谱,以前囟为基点,后移 2. 8mm,左移 0. 5mm,定
( 1. 浙江医学高等专科学校药学系,浙江 杭州 310053; 2. 浙江中医药大学药学院药理教研室,浙江 杭州 310053)
摘 要:目的: 探讨葛根素在正常和 β - 淀粉样蛋白所致痴呆模型大鼠( AD 模型大鼠) 体内的动力学过程。
方法: AD 模型组大鼠基底核定位注射 Aβ,7 天后灌胃 500mg / kg 葛根素注射液,正常组灌胃等剂量葛根素注射
氯酸溶液 200 μL,旋涡混合 2min,4000r / min 离心 15min,取
上清液进样 20 μL 测定。 2. 3 方法学考察
2. 3. 1 线性范围及检测限 分别取 7 份空白血浆各 200
μL,加入不同浓度梯度的葛根素对照品稀释液各 200 μL, 配成每 mL 血 浆 含 葛 根 素 为 0. 50、1. 0、5. 0、10. 0、20. 0、
测得结果依次分别为 83. 16% ,74. 84% ,79. 03% 。葛根素
平均萃取回收率为( 79. 01 ± 4. 16) % 。
2. 3. 3 精密度 分别取 9 份空白血浆各 200 μL,加入 3 种
不同浓度的葛根素对照品稀释液,制成浓度为 5. 0、10. 0、 50. 0 μg·mL - 1 的血浆样品各 3 份,按 2. 2 项下方法处理
葛根素注射液对血管性痴呆小鼠神经元的保护作用
葛根素注射液对血管性痴呆小鼠神经元的保护作用作者:田华刘富刘冰华高音来源:《科技创新导报》 2014年第4期田华1 刘富2 刘冰华2 高音2(1齐齐哈尔医学院药理教研室黑龙江齐齐哈尔 161006;2齐齐哈尔医学院解剖教研室黑龙江齐齐哈尔 161006)摘要:目的:旨在探讨葛根素注射液对血管性痴呆小鼠皮层、海马神经元的保护作用。
方法:通过采用改良的双侧颈总动脉持久性结扎法制作血管性痴呆模型,从形态学上观察葛根素对血管性痴呆小鼠皮层、海马神经元的保护作用。
结果:(1)跳台试验结果提示,葛根素注射液组小鼠的反应时间明显缩短,潜伏期显著延长,错误次数减少。
(2)HE染色结果显示,与假手术组相比,模型组小鼠皮层、海马神经元细胞排列紊乱,数目减少,周围有小胶质细胞和星形胶质细胞增生,形态异常,许多细胞出现体积缩小,出现核溶解、固缩、破裂等。
与模型组相比,葛根素注射液组小鼠海马、皮层神经元细胞形态异常、损伤的情况明显减轻。
结论:葛根素注射液对血管性痴呆小鼠皮层、海马神经元发挥一定的神经保护作用。
关键词:葛根素血管性痴呆皮层神经元中图分类号:R285 文献标识码:A 文章编号:1674-098X(2014)02(a)-0221-02血管性痴呆(vascular dementia,VD)是继阿尔茨海默病之后第二位最常见的痴呆症,是指大脑功能衰退,特别是与智能有关的功能全面衰退,致其发生的主要原因是脑血流量减少,引起脑组织生化代谢障碍[1]。
葛根素(Puerarin,Pur)化学名为4,7一二羟基-8-D葡萄糖基异黄酮,近年来被广泛用于心脑血管疾病等急性期的治疗,包括心绞痛、心肌梗塞,心律失常、脑缺血等[2]。
本实验从行为学、形态学角度,观察葛根素注射液对血管性痴呆小鼠皮层、海马神经元的保护作用。
1 材料和方法1.1 血管性痴呆模型建立成年昆明小鼠,18~22g,雌雄不限,购自齐齐哈尔医学院动物研究中心。
采用双侧颈总动脉持久性结扎法,并对该方法进行改良。
葛根素对缺血性脑卒中模型大鼠神经损伤的防护作用
葛根素对缺血性脑卒中模型大鼠神经损伤的防护作用于心洋;张贺齐;刘云平;尚淑玲【期刊名称】《中国药业》【年(卷),期】2022(31)4【摘要】目的探讨葛根素对缺血性脑卒中模型大鼠神经损伤的防护作用。
方法将90只雄性SD大鼠随机分为空白对照组(等体积生理盐水)、假手术组(等体积生理盐水)、模型组(等体积生理盐水)、尼莫地平组(12 mg/kg)及葛根素低、高剂量组(50 mg/kg,100 mg/kg),各15只。
除空白对照组外,其余各组均采用线栓法复制缺血性脑卒中大鼠模型。
建模成功后,各组大鼠灌胃相应药物或生理盐水,每日1次,连续14 d。
记录各组大鼠术后6 h及术后1,4,7,14 d的Longa评分;2,3,5-氯化三苯基四氮唑(TTC)染色,测定大鼠脑组织梗死体积;Nissl染色,观察大鼠海马组织神经细胞形态学;采用酶联免疫吸附(ELISA)法检测大鼠血清神经元特异性烯醇化酶(NSE)及星形胶质源性蛋白(S100β)水平,采用Western blot法检测大鼠海马组织匀浆中GSK-3β,p-GSK-3β,β-catenin蛋白表达水平。
结果与模型组比较,尼莫地平组及葛根素高剂量组大鼠术后4,7,14 d,葛根素低剂量组大鼠术后7,14 d的Longa评分均明显降低,且各用药组大鼠脑梗死体积明显缩小,海马区神经细胞损伤明显改善,血清中NSE和S100β蛋白表达水平均明显降低,脑组织匀浆中GSK-3β蛋白表达水平明显降低,p-GSK-3β和β-catenin蛋白表达水平均明显升高(P<0.05)。
结论葛根素能改善缺血性脑卒中模型大鼠的神经功能,减轻神经细胞损伤,其作用机制可能与调节Wnt/β-catenin信号通路相关。
【总页数】6页(P58-63)【关键词】葛根素;缺血性脑卒中;神经细胞;WNT/Β-CATENIN信号通路;作用机制;大鼠【作者】于心洋;张贺齐;刘云平;尚淑玲【作者单位】河北省唐山市协和医院神经内科【正文语种】中文【中图分类】R965;R971【相关文献】1.G蛋白偶联雌激素受体介导的雌激素减轻缺血性脑卒中大鼠神经损伤的作用及机制探讨2.丹参酮ⅡA磺酸钠对缺血性脑卒中大鼠受损组织的防护作用研究3.携载miRNA-124纳米粒子经鼻吸入对缺血性脑卒中模型大鼠的神经保护作用4.蛭龙活血通瘀胶囊对缺血性脑卒中模型大鼠的作用及对脑皮层神经元线粒体的影响5.头针联合中药对缺血性脑卒中失眠大鼠模型p38MAPK浓度的作用因版权原因,仅展示原文概要,查看原文内容请购买。
葛根异黄酮对衰老模型大鼠学习记忆及海马乙酰胆碱酯酶的影响
作用机制尚不十分清楚。本研究旨在观察植物雌激素——葛
根异黄酮对衰老模型大鼠学习记忆能力的改善作用,并检测大 鼠海马内乙酰胆碱酯酶(AChE)活性,进而探讨葛根异黄酮的 作用机制。
第一作者:王爱梅(1974-),女,硕士,讲师,主要从事神经生理及抗衰老 的研究。
动物分组及给药将大鼠随机分为生理盐水对照组(生
4参考文献
1
7 K,Mautes
Zhallg cardiac phys
M,)【uⅥ,Saini
dysfunction
HK,et to
a2.TNF—aIpha∞a pokntial mediator 0f
M/liel"B,Schwerdffeger
kines 26,8,and 10 in
A,et
a/.Differential release of intedeu—
(编辑张永贵/张慧)
葛根异黄酮对衰老模型大鼠学习记忆及海马乙酰胆碱酯酶的影响
王爱梅李
[摘要]
弋
(南阳医学高等专科学校,河南
南阳473061)
目的观察葛根异黄酮对衰老模型大鼠学习记忆及海马乙酰胆碱酯酶(ACHE)的影响。方法选用3—4月龄。雌性Wistar大鼠,体
重(180土20)g,随机分为生理盐水对照组(生理盐水组),衰老模型组(模型组)。用药(低、中、高剂量)组,每组11只。造模时,模型组、药物组每天 颈背部皮下注射l%D.半乳糖100 ms/kg,生理盐水组每天颈背部皮下注射等量生理盐水,连续6 W。造模成功后,第2天开始灌胃给药,药物(低、 中、高剂量)组每天分别给予(40、80、160 rag/ks)葛根异黄酮灌胃,模型组、生理盐水组每天给予80 Ing/kg生理盐水灌胃,连续8 W。用药结束后,用 跳台实验方法测试大鼠学习记忆功能。制备海马10%组织匀浆,检测其AChE活性。结果与模型组比较,低、中、高剂量用药组均能使衰老模型大 鼠的潜伏期延长,错误次数减少(P<O.05,P<O.01)。与模型组比较.中、高剂量组大鼠海马中AChE活性明显降低(P<o,05,P(O.01)。结论葛 根异黄酮对衰老模型大鼠学习记忆功能具有促进作用,其机制可能与AChE活性降低有关。 [关键词】葛根异黄酮;衰老;学习记忆;海马;AChE (中图分类号】R338.2 (文献标识码】A
葛根素对血管性痴呆大鼠脑组织NO、NOS含量及病理学的影响
葛根素对血管性痴呆大鼠脑组织NO、NOS含量及病理学的
影响
朱慧渊;胡晓蕾
【期刊名称】《中医药临床杂志》
【年(卷),期】2007(19)4
【摘要】目的:探讨葛根素对血管性痴呆(VD)大鼠脑组织一氧化氮(NO)、一氧化氮合酶(NOS)浓度及病理学改变的影响。
方法:中动脉阻塞法制备VD动物模型,检测脑组织NO、NOS含量,HE染色、尼氏染色检测病理变化。
结果:模型组较空白组海马及大脑皮质NO、NOS含量显著升高,神经元丢失及胶质细胞增生;葛根素组较模型组海马及大脑皮质NO、NOS含量显著降低,神经元损伤得以恢复。
结论:葛根素对血管性痴呆有一定防治作用。
【总页数】2页(P354-355)
【关键词】血管性痴呆;大鼠;葛根素;一氧化氮;一氧化氮合酶
【作者】朱慧渊;胡晓蕾
【作者单位】浙江中医药大学
【正文语种】中文
【中图分类】R749.1
【相关文献】
1.葛根素对血管性痴呆大鼠脑组织Na+-K+-ATP酶含量、梗死体积及病理学改变的影响 [J], 朱慧渊
2.脑通胶囊对血管性痴呆大鼠学习记忆能力及脑组织NO、SOD含量的影响 [J], 况时祥;刘继刚;肖燕;官志忠
3.智脑胶囊对血管性痴呆模型大鼠行为学及脑组织SOD活性、MDA含量的影响[J], 杨文明;王时光;洪亮;鲍远程;张波;汪美霞;董婷
4.智脑胶囊对血管性痴呆模型大鼠行为学及脑组织NO、ET、CGRP含量的影响[J], 杨文明;王时光;鲍远程;杨兴涛;张波;汪美霞;董婷
5.智脑胶囊对血管性痴呆模型大鼠学习记忆及脑组织Ca^(2+)含量的影响 [J], 杨文明;王时光;鲍远程;杨兴涛;张波;汪美霞;董婷
因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素对血管性痴呆大鼠海马中低氧诱导因子-1α和红细胞生成素表达的影响
葛根素对血管性痴呆大鼠海马中低氧诱导因子-1α和红细胞生成素表达的影响吴海琴;张蓓;张桂莲;展淑琴;张茹;赵英贤【期刊名称】《西安交通大学学报(医学版)》【年(卷),期】2006(027)002【摘要】目的观察葛根素对血管性痴呆(VD)大鼠海马中低氧诱导因子-1α和红细胞生成素表达的影响并探讨其可能机制.方法采用大鼠双侧颈总动脉永久性阻断法(2-VO)建立VD的动物模型,SD大鼠随机分为假手术组、2-VO组和葛根素组,每组又分为1周、3周、6周、2月(n=6)四个时间点;应用Y型水迷宫实验测定动物的学习记忆能力,采用免疫组化法检测大鼠海马CA1区HIF-1α和EPO蛋白的表达.结果①2-VO组和葛根素组大鼠游全程时间和误入盲端次数均显著增加,但各时间点葛根素组大鼠的学习记忆成绩均显著优于2-VO组.②各时间点,2-VO组和葛根素组大鼠海马CA1区HI-1α和EPO的表达均较假手术组明显增高,但葛根素组两种蛋白的表达均较2-VO组明显降低.结论葛根素可提高缺血脑组织细胞内氧浓度,改善学习记忆功能,对VD具有确切的保护作用.【总页数】5页(P132-136)【作者】吴海琴;张蓓;张桂莲;展淑琴;张茹;赵英贤【作者单位】西安交通大学医学院第二附属医院神经内科,陕西,西安,710004;西安交通大学医学院第二附属医院神经内科,陕西,西安,710004;西安交通大学医学院第二附属医院神经内科,陕西,西安,710004;西安交通大学医学院第二附属医院神经内科,陕西,西安,710004;西安交通大学医学院第二附属医院神经内科,陕西,西安,710004;西安交通大学医学院第二附属医院神经内科,陕西,西安,710004【正文语种】中文【中图分类】R743.3【相关文献】1.葛根素对血管性痴呆大鼠海马突触传递长时程增强的影响 [J], 张冰清;王玉良2.低氧对大鼠视网膜Müller细胞低氧诱导因子1α和促红细胞生成素蛋白表达的影响 [J], 曲虹;厉泉3.促红细胞生成素对血管性痴呆大鼠海马CA1区胆碱乙酰转移酶的影响 [J], 黄树其;牛富生;邵福源4.葛根素对血管性痴呆大鼠海马锥体细胞和BDNF表达的影响 [J], 张冰清;王玉良5.低氧诱导因子-1、促红细胞生成素在血管性痴呆大鼠海马的表达 [J], 张蓓;吴海琴;张海雄;张桂莲;展淑琴因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素对慢性低氧高二氧化碳大鼠海马超微结构的影响
葛根素对慢性低氧高二氧化碳大鼠海马超微结构的影响邵胜敏;王小同;黄汉津;陈松芳【期刊名称】《实用医学杂志》【年(卷),期】2006(22)13【摘要】目的:探讨慢性低氧(O2)高二氧化碳(CO2)大鼠海马超微结构、超氧化物歧化酶(SOD)、丙二醛(MDA)等改变及葛根素对其影响.方法:建立慢性低 O2高CO2肺动脉高压大鼠模型,给予葛根素干预,电镜观察海马超微结构并测定 SOD和MDA.结果:慢性低 O2高 CO2使海马神经元和神经胶质细胞水肿,并使 SOD活性降低、 MDA含量升高,葛根素干预可使海马神经细胞结构损害减轻.结论:葛根素对慢性低 O2高 CO2大鼠海马神经细胞结构损害有保护作用,其机制可能与抗氧化作用有关.【总页数】2页(P1496-1497)【作者】邵胜敏;王小同;黄汉津;陈松芳【作者单位】325027,温州医学院附属第二医院神经内科;325027,温州医学院附属第二医院神经内科;325027,温州医学院附属第二医院神经内科;325027,温州医学院附属第二医院神经内科【正文语种】中文【中图分类】R5【相关文献】1.葛根素对慢性低氧高二氧化碳大鼠肺动脉管壁胶原代谢的影响 [J], 龚永生;李继武;杨鹏麟;范小芳;胡良冈;郑绿珍;蒋仲荪2.慢性低氧高二氧化碳大鼠大脑超微结构改变和SOD、MDA、XOD变化及红花的保护作用 [J], 陈茜;陈少贤;王良兴;李勇3.慢性低氧高二氧化碳对大鼠海马NFкB的影响 [J], 李勇;柯将琼;邵胜敏;宫剑;陈松芳;王小同4.葛根素对慢性低氧高二氧化碳性肺动脉高压大鼠的保护作用 [J], 范小芳;李继武;胡良冈;龚永生;黄虹5.葛根素对慢性低氧高二氧化碳大鼠肺动脉高压形成的预防作用 [J], 李继武;季亢挺;范小芳;张素勤;龚永生;杨鹏麟因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素降低细胞内Ca^(2+)浓度并上调BDNF保护血管性痴呆大鼠海马神经细胞
葛根素降低细胞内Ca^(2+)浓度并上调BDNF保护血管性痴呆大鼠海马神经细胞任宏伟;陈明;王虎清;高震;吴海琴【期刊名称】《西安交通大学学报:医学版》【年(卷),期】2023(44)2【摘要】目的观察葛根素对血管性痴呆(VD)大鼠海马神经细胞内Ca^(2+)浓度及脑源性神经生长因子(BDNF)的影响,探讨葛根素保护神经细胞的可能机制。
方法雄性SD大鼠随机分为假手术组、模型组和葛根素干预组,采用间隔3 d结扎双侧颈总动脉法建立VD模型;术后2周用Morris水迷宫评估大鼠学习记忆能力,免疫组化和Western blotting检测大鼠海马组织BDNF的表达情况,流式细胞仪检测平均荧光强度来表示细胞内游离Ca^(2+)浓度。
结果葛根素干预组Morris水迷宫逃避潜伏期显著缩短,海马BDNF表达量显著增加,海马神经细胞内Ca^(2+)浓度减低;与模型组相比,差异有统计学意义(P<0.05)。
结论葛根素对VD大鼠具有神经保护作用,其作用机制可能与葛根素降低海马神经细胞内Ca^(2+)浓度及上调BDNF的表达有关。
【总页数】6页(P202-207)【作者】任宏伟;陈明;王虎清;高震;吴海琴【作者单位】西安交通大学第二附属医院神经内科【正文语种】中文【中图分类】R285.5【相关文献】1.前列腺素E1上调血管性痴呆大鼠海马组织VEGF、BDNF的表达2.纳洛酮对血管性痴呆大鼠学习记忆能力和海马神经细胞内钙离子浓度的影响3.慢性染铅对大鼠海马区神经细胞Ca^(2+)浓度及Ca^(2+)-ATP酶活性的影响4.针刺井穴对血管性痴呆大鼠脑神经细胞内Ca^(2+)浓度的影响5.铅对大鼠海马神经细胞Ca^(2+)浓度、Ca^(2+)-ATP酶、Na^+-ATP酶活性的影响因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素对缺氧缺血性脑病新生大鼠脑细胞凋亡及Bim蛋白表达的影响(1)
葛根素对缺氧缺血性脑病新生大鼠脑细胞凋亡及Bim 蛋白表达的影响陈 俊1,张丙宏2,陶旭炜2,赵日红2,张海霞2(1.广东省深圳市南山区妇幼保健院,广东深圳518052;2.武汉大学人民医院,湖北武汉430060) [摘要] 目的 观察葛根素对缺氧缺血性脑病(HIE )新生大鼠脑细胞凋亡及Bim 蛋白表达的影响。
方法 7日龄Wistar 大鼠随机分成假手术组、HIE 模型组、葛根素治疗组,每组22只,参照Rice 法制作HIE 模型,应用Western 2blot 和流式细胞术在不同时间点分别检测大脑皮质Bim 蛋白的表达和细胞凋亡情况。
结果 HIE 模型组和葛根素治疗组脑细胞Bim 蛋白表达灰度值在12h 分别为137.00±5.00和78.20±4.38,24h 分别为164.40±5.50和90.40±3.36,均显著高于假手术组(29.60±3.20和30.00±3.74),葛根素治疗组Bim 蛋白表达低于HIE 模型组。
HIE 模型组和葛根素治疗组脑皮质早期凋亡细胞比例在12h 分别为(16.2±1.86)%和(8.54±1.91)%,24h 分别为(20.03±2.02)%和(13.36±1164)%,均显著高于假手术组((0.76±0.09)%和(1.34±0.24)%),葛根素治疗组细胞凋亡低于HIE 模型组。
结论 葛根素能抑制HIE 新生大鼠脑细胞凋亡启动因子Bim 蛋白表达,具有抗脑细胞凋亡作用。
[关键词] 缺氧缺血性脑病;凋亡;Bim 蛋白;葛根素;大鼠[中图分类号] R285.5 [文献标识码] A [文章编号] 1008-8849(2009)35-4335-04I nfluence of puerarin on brain cell apoptosis and expression of Bim protein in hypoxicischemic encephalopathy neonatal R atsChen J un 1,Zhang Binghong 2,Tao Xuwei 2,Zhao Rihong 2,Zhang Haixia 2(1.The Maternal and Child Health Hospital of Nanshan District ,Shenzhen 518052,Guangdong ,China ;2.The People ’s Hospital of Wuhan University ,Wuhan 430060,Hubei ,China )Abstract :Objective It is to observe the influence of puerarin on brain cell apoptosis and the expression of Bim protein in hypoxic ischemic encephalopathy (HIE )neonatal Rats.Methods Seven 2day neonatal Wistar Rats were randomly divided into sham operated group ,HIE model group and puerarin treatment group in which there were 22Rats.HIE model was made in accordance with Rice method.The expression of Bim protein and apoptosis in cerebral cortex was respectively detected with western 2blot and flow cytometry in different time point.R esults The grayscale value of the expression of Bim protein in brain cell in HIE model group and puerarin treatment group at 12h was 137.00±5.00and 78.20±4.38res pectively and that at 24h was 164.40±5.50and 90.40±3.36res pectively ,which was both significant higher than that in sham operated group (29.60±3.20and 30.00±3.74).The expression of Bim protein in puerarin treatment group was lower than that in HIE model group.The ratio of viable apoptotic cell in cerebral cortex in HIE model group and puerarin treatment group at 12h was (16.2±1.86)%and (8.54±1.91)%respectively and that at 24h was (20.03±2.02)%and (13.36±1.64)%respectively ,which was both significant higher than that in sham operated group ((0.76±0.09)%and (1.34±0.24)%),respectively.The ratio of viable apoptotic cell in puerarin treatment group was lower than that in HIE model group.Conclusion Puerarin can inhibit the expression of brain cell apoptosis initiator Bim protein in HIE neonatal Rats and has the action of resisting brain cell apoptosis.K ey w ords :hypoxic ischemic encephalopathy ;apoptosis ;Bim protein ;puerarin ;Rats[作者简介] 陈俊(1975—),男,硕士研究生,主治医师,研究方向为新生儿缺氧缺血性脑病。
葛根素预处理对局灶性脑缺血再灌注大鼠海马 CA1区神经元损伤的影
关键词 葛根 素 ; 缺 血再 灌 注 损 伤 ; 神经保护; C a s p a s e - 3 ;
Ca s pa s e - 9
中图分类号
R 3 3 — 3 3 ; R 9 3 2
文献标志码 A 文章编 号 1 0 0 0—1 4 9 2 ( 2 0 1 5 ) 0 6— 0 7 2 3—0 4
响; 以期 能够 对葛 根 素 的 神 经保 护 作 用 进 行 初 步 探
讨, 从 而 为临 床上 缺 血 性 脑 损 伤方 面 疾 病 的治疗 提
供一 定 的理论 基础 和 科学依 据 。
1 材 料 与方 法
1 . 1 材料
0 . 0 5 ) 。结论
缺血前 1 h葛 根素预处理对局灶性 脑缺血 大
及对 促 凋 亡 基 因 C a s p a s e 一 3和 C a s p a s e 一 9表 达 的 影
鼠海马 C A 1区神 经元损伤的保护作 用及其 机制 。方法 雄 性s D大 鼠3 0只 , 随机均等分为假手术组 、 模 型组 、 葛根 素预
处理 组。采用线栓法 阻断 大鼠大脑 中动脉血供 , 6 0 m i n后拔 出栓 线造成局部脑缺 血再 灌注损 伤。葛根 素预 处理 组在 缺
2 0 1 5— 0 2—1 9接 收
o c c l u s i o n ,MC A O) 的脑 缺 血 动 物 模 型 , 6 0 m i n后 拔
分 为假 手术 组 、 模 型组 、 葛根 素预 处理组 。假手术 组 大 鼠只分离 血 管 , 不 插入 栓线 ; 采 用线 栓法 将模 型组
大 鼠制Байду номын сангаас 成 大 脑 中动 脉 阻塞 ( mi d d l e c e r e b r a l a r t e r y
葛根素对缺氧缺血性脑病新生大鼠脑细胞凋亡及Bim蛋白表达的影响
葛根素对缺氧缺血性脑病新生大鼠脑细胞凋亡及Bim蛋白表达的影响陈俊;张丙宏;陶旭炜;赵日红;张海霞【期刊名称】《现代中西医结合杂志》【年(卷),期】2009(018)035【摘要】目的观察葛根素对缺氧缺血性脑病(HIE)新生大鼠脑细胞凋亡及Bim蛋白表达的影响.方法 7日龄Wistar大鼠随机分成假手术组、HIE模型组、葛根素治疗组,每组22只,参照Rice法制作HIE模型,应用Western blot和流式细胞术在不同时间点分别检测大脑皮质Bim蛋白的表达和细胞凋亡情况.结果 HIE模型组和葛根素治疗组脑细胞Bim蛋白表达灰度值在12h分别为137.00±5.00和78.20±4.38,24h分别为164.40±5.50和90.40±3.36,均显著高于假手术组(29.60±3.20和30.00±3.74),葛根素治疗组Bim蛋白表达低于HIE模型组.HIE模型组和葛根素治疗组脑皮质早期凋亡细胞比例在12h分别为(16.2±1.86)%和(8.54±1.91)%,24h分别为(20.03±2.02)%和(13.36±1.64)%,均显著高于假手术组((0.76±0.09)%和(1.34±0.24)%),葛根素治疗组细胞凋亡低于HIE模型组.结论葛根素能抑制HIE新生大鼠脑细胞凋亡启动因子Bim蛋白表达,具有抗脑细胞凋亡作用.【总页数】4页(P4335-4338)【作者】陈俊;张丙宏;陶旭炜;赵日红;张海霞【作者单位】广东省深圳市南山区妇幼保健院,广东,深圳,518052;武汉大学人民医院,湖北,武汉,430060;武汉大学人民医院,湖北,武汉,430060;武汉大学人民医院,湖北,武汉,430060;武汉大学人民医院,湖北,武汉,430060【正文语种】中文【中图分类】R285.5【相关文献】1.针刺对局灶性脑缺血大鼠脑细胞凋亡及Bcl-2蛋白表达的影响 [J], 余晓慧;孙国杰2.血必净注射液对脑挫伤大鼠脑细胞凋亡及Bcl-2、Bax蛋白表达的影响 [J], 罗凤芹;张娟;叶玲3.缺氧缺血性脑病新生大鼠心肌Caspase-3、Cyt C蛋白表达与心肌细胞凋亡的关系 [J], 刘英;郭世杰;韩军;周文莉4.三七皂甙单体Rb1对大鼠脑缺血再灌注时脑细胞凋亡及cPLA2蛋白表达的影响[J], 王东吉;武凡5.葛根素对烟熏大鼠脑细胞凋亡及Fas蛋白表达的影响 [J], 李华;柳忠兰因版权原因,仅展示原文概要,查看原文内容请购买。
浅析葛根素对大鼠局灶性缺血脑组织内海马神经元型一氧
化氮合酶的影响摘要本文研究了葛根素对大鼠局灶性缺血脑组织内海马神经元型一氧化氮合酶的影响。
通过实验发现,葛根素可以降低缺血大鼠海马神经元型一氧化氮合酶表达,从而起到一定的神经保护作用。
引言脑缺血是指由于脑血管受到阻塞、破裂或狭窄等因素导致脑部缺血缺氧的一种病理状态。
脑缺血可以导致大脑神经元坏死,此类神经细胞缺氧和3个生化过程即过氧化作用、胀性反应及酸性酶激活的协同作用是导致缺血后神经元损伤和死亡的关键因素,其中胀性反应和过氧化作用是重要的死亡通路,而一氧化氮在脑缺血后的神经元损伤中也有一定的作用。
而葛根素作为一种天然药物,因其具有一定的抗氧化、抗炎、抗缺血等作用,逐渐引起了人们的关注。
实验药品和试剂葛根素(puerarin,化学纯),一氧化氮合酶(nNOS)多克隆抗体(rabbit polyclonal,Santa Cruz)。
动物模型的建立雄性Wistar大鼠,体重250-280g,随机分为5组,每组10只,分别为正常组、假手术组、缺血组、葛根素治疗组(100mg/kg)和葛根素治疗组(200mg/kg)。
试验方法1.麻醉采血:给大鼠注射40mg/kg的氯丙嗪与10mg/kg的地西泮进行麻醉,取体重的10%进行静脉注射葛根素,25分钟后进行颈静脉采血。
2.物理交感神经动物模型的建立,4d后处死动物,进行HE染色观察和nNOS染色观察。
结果HE染色结果大鼠局灶性缺血脑组织内海马晕球细胞形态规则,胞体大小均一,胞浆充实,核染色质均匀染色,核质比值较高。
nNOS染色结果nNOS阴性细胞呈现出特定的淡蓝色色素沉积,而nNOS阳性细胞为深蓝色色素沉积。
如图所示,葛根素能显著抑制缺血后大鼠海马神经元型一氧化氮合酶表达量的上升,而且两种剂量的葛根素在抑制一氧化氮合酶表达方面并没有明显的差异。
讨论本实验结果表明葛根素能显著抑制缺血后大鼠海马神经元型一氧化氮合酶表达量的上升,从而在一定程度上发挥神经保护作用。
在缺血脑组织中一氧化氮水平增高,会导致ROS产生增加、氧化应激反应亢进、氧化性损伤加重及导致细胞死亡,而葛根素具有类似于一氧化氮的生理效应,有一定的抗氧化作用,能抑制ROS生成,增强一氧化氮的生物活性,减轻一氧化氮对损伤的影响。
血管性痴呆的中药实验研究进展
脂、心理障碍等高危因素,因此对VD模型的相关危险因素进行 研究有重要意义。通过制作相关的VD复合模型使其更接近临 床,以研究中药的防治机理。但受技术所限,此类的研究较少,且 多局限在模型制作方面。如莫飞智等Ⅲ1在复制肾性高血压大鼠 (RHR)的基础上反复阻断双侧颈总动脉及迷走神经,引起急性 不完全性脑缺血而造成RHR—VD的动物模型;宋莉莉等n5l在 链脲佐菌素诱导慢性实验性糖尿病(DM)大鼠模型的基础上永 久性结扎双侧颈总动脉建立DM—VD模型,结果显示DM合并 VD大鼠的空间学习和记忆能力显著低于VD大鼠,表明DM能 加重VD的认知障碍。雷燕等1261在食饵性高脂血症大鼠基础上 采用反复脑缺血再灌注造成大鼠脑损伤模型,发现复圣散能通 过调节脑内神经肽含量而减轻缺血性脑损伤;周海哲等心7I在造 成大鼠高脂血症的基础上结扎双侧CCA制作痰瘀交结型VD模 型,运用脑泰通颗粒治疗后,可提高VD模型大鼠学习能力及脑 组织中SS水平。 5分析与展望 从近年中药治疗VD的实验研究中,可见药物组成多以补 肾活血、解毒化痰、益气养血、开窍等方药为主,其作用机制可能 是通过作用于与记忆有关的神经结构、神经介质、脑组织形态、 神经细胞增殖、分化等方面,进而改变机体的功能状态,达到治 疗作用,这也体现了中药防治VD具有“多成分、多靶点、整合调 节作用”的特点。 中药对VD模型的实验研究虽然取得一定进展,但同时也 存在不少问题。如目前的VD模型只能复制人类VD发病的某一 个侧面,因此建立更加符合VD临床特征的动物模型是目前实 验研究中的首要问题。此外,当前研究中重复性较多,偏重于行 为学、神经生化指标,而对于复合模型及神经细胞的研究很少, 且中药药理和复方配伍机制、药物的相互作用及中药有效单体 的定位等还处于探索阶段。近年来使用先进手段进行中药治疗 VD的研究正成为热点。如通过中药激发内源性神经干细胞 (NSC)来替代受损神经细胞的功能,促进血管内皮生长因子 (VEGF)的表达,以保护大脑皮层免受缺血、缺氧的损伤。这为中 药治疗VD提供了新的研究思路和途径,将是今后研究的重 点。应充分借助现代分子生物学、药理学、病理学的理论与技术, 建立更接近人类VD的统一动物模型及相应的研究指标,使中
葛根素对D-半乳糖诱导糖基化大鼠脑损害的干预作用
葛根素对D-半乳糖诱导糖基化大鼠脑损害的干预作用吕俊华;张世平;沈飞海;潘竞锵;谭海荣【期刊名称】《中国中药杂志》【年(卷),期】2006(31)14【摘要】目的:研究葛根素(puerarin,Pue)对D-半乳糖诱导的糖基化模型大鼠并发脑损害的干预作用。
方法:D-半乳糖(150 mg.kg-1.d-1,ip)给药8周,诱导糖基化模型大鼠,并于第3周开始给予葛根素高、中、低(300,150,75mg.kg-1)剂量处理6周。
测定红细胞醛糖还原酶活性、糖化血红蛋白、血清果糖胺和晚期糖基化终末产物(AGEs)含量及脑组织中AGEs含量、脑神经细胞内钙离子水平,并以透射电镜观察脑内海马神经细胞线粒体的变化。
结果:葛根素高、中剂量明显降低模型组大鼠红细胞醛糖还细胞酶活性,抑制糖化产物的形成(P<0.01),降低脑组织中AGEs及脑细胞内钙的含量(P<0.05,P<0.01),保护海马神经细胞线粒体结构的完整性。
结论:葛根素具有抑制D-半乳糖诱导的蛋白糖基化反应,并对糖基化状态并发的脑神经细胞损害具有保护作用。
【总页数】4页(P1184-1187)【关键词】葛根素;D-半乳糖;糖基化;钙离子;线粒体【作者】吕俊华;张世平;沈飞海;潘竞锵;谭海荣【作者单位】暨南大学药学院;广州市中医中药研究所【正文语种】中文【中图分类】R285.5【相关文献】1.小檗碱对D-半乳糖诱导糖基化大鼠脑损害的干预作用 [J], 吕俊华;张世平;郑浩2.茶多酚对D-半乳糖诱导糖基化大鼠脑损害的干预作用 [J], 冯亮;徐辰;郑浩;沈文娟;吕俊华3.葛根素对D-半乳糖诱导大鼠抑制蛋白非酶糖基化及增强胰岛素敏感性的作用 [J], 钟艺;谭海荣;潘竞锵;肖柳英4.小檗碱对D-半乳糖诱导糖基化模型大鼠脑损害的干预作用 [J], 林媛;张世平;吕俊华5.D-半乳糖诱导大鼠肾脏损害的糖基化机制及药物干预作用 [J], 李冰;郑浩因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素对血管性痴呆大鼠脑组织Na+-K+-ATP酶含量、梗死体积及病理学改变的影响
葛根素对血管性痴呆大鼠脑组织Na+-K+-ATP酶含量、梗死体积及病理学改变的影响朱慧渊【摘要】目的观察葛根素对血管性痴呆(VaD)大鼠腩组织Na+-K+-A3TP酶的含量及病理学改变的影响.方法采用中动脉阻塞法制备VaD动物模型,检测脑组织含量,HE染色病理及梗死体积变化.结果模型组较空白组海马及大脑皮质Na+-K+-ATP酶含量减少,神经元丢失及胶质细胞增生,大脑梗死面积明显增加.葛根素组较模型组海马及大脑皮质Na+-K+-ATP酶含量、神经元损伤得以恢复,大脑梗死体积有所改善.结论葛根素对血管性痴呆有一定防治作用.【期刊名称】《中国中医急症》【年(卷),期】2010(019)003【总页数】2页(P483-484)【关键词】血管性痴呆;大鼠;葛根素;Na+-K+-ATP酶;梗死体积【作者】朱慧渊【作者单位】陕西中医学院,咸阳,712038【正文语种】中文【中图分类】R285.5血管性痴呆(VaD)是指缺血性、出血性脑血管疾病引起的脑功能损害所致的痴呆。
随着老龄化进程的加快和老龄化程度的加深,VaD患病率将逐渐上升,逐渐增多的痴呆患者已成为家庭和社会的沉重负担[1],因而探讨VaD的防治也已成为社会和医学界关注的重要课题。
研究发现VaD患者基底前脑、海马等广泛脑区存在大量神经元凋亡细胞。
本实验通过生化指标检测,以研究葛根素对VaD保护作用机制。
1 材料与方法1.1 材料(1)实验动物:雄性SD大鼠30只,购于浙江省中医药大学实验动物中心,8~12周龄,体质量280~300g。
(2)分组:动物随机分为空白组、模型组、葛根素组。
模型组、空白组不作药物处理,而采用蒸馏水灌胃进行模拟处理。
葛根素组采用葛根素100mg/kg灌胃,每日1次,连续7d。
(3)模型制备:第7次灌胃后1h对模型组和葛根素组进行VaD模型制备。
大鼠VaD模型根据MCAO法复制[1,2]。
大鼠使用10%水合氯醛腹腔注射(1g/kg)麻醉后,将其仰卧固定。
葛根素对血管性痴呆患者认知功能和事件相关电位P300的影响
葛根素对血管性痴呆患者认知功能和事件相关电位P300的影响林伟【期刊名称】《中国中医药咨讯》【年(卷),期】2009(001)002【摘要】目的观察葛根素对血管性痴呆(VaD)患者认知功能和听觉事件相关电位P300的影响。
方法将70例VaD患者随机分成两组各35例,葛根素治疗组和对照组。
应用简易精神状态检查量表(MMSE)评定两组患者治疗前后认知功能状况,并进行治疗前后P300检查。
同时记录药物不良反应。
结果两组各35例进入结果分析。
治疗前两组MMSE评分、P300的潜伏期及波幅差异无显著性(均P〉0.05)。
治疗14d、30d时,两组MMSE评分显著提高(均P〈0.01),P300潜伏期均有缩短,波幅均有提高(葛根素组P〈0.01,对照组P〈0.05);治疗后14d时认知功能改善葛根素组明显优于对照组(总有效率分别为91.4%,71.4%)(P〈0.05)。
两组治疗期间无严重不良反应。
结论葛根素能够改善VaD患者的认知功能,这可能与葛根素的扩血管、脑保护作用有关。
【总页数】2页(P232-233)【作者】林伟【作者单位】金坛市人民医院神经内科,213200【正文语种】中文【中图分类】R749.94【相关文献】1.早期干预治疗对血管性痴呆患者事件相关电位(P300)的影响 [J], 洪雁;梁智;方芳;苏宇2.通窍调神法对血管性痴呆患者认知功能及事件相关电位P300的影响 [J], 鹿俊磊;卢昌均;韦冰心;刘国成;周哲屹;王洁琼3.浅析对血管性痴呆患者进行团体心理治疗对其事件相关电位P300的影响 [J], 黄滢珊;罗庆禄;韩平4.天智颗粒对血管性痴呆认知功能及事件相关电位P300的影响 [J], 王少华5.血管性痴呆患者事件相关电位 P300改变与认知功能障碍的关系 [J], 郑健;赵莘瑜;李凤鹏因版权原因,仅展示原文概要,查看原文内容请购买。
葛根素对血管性痴呆大鼠海马锥体细胞和BDNF表达的影响
葛根素对血管性痴呆大鼠海马锥体细胞和BDNF表达的影响张冰清;王玉良【期刊名称】《神经解剖学杂志》【年(卷),期】2007(23)6【摘要】为了观察葛根素对血管性痴呆(VD)模型大鼠海马锥体细胞和BDNF表达的影响及其作用机制,本研究采用双侧颈总动脉缺血再灌注,同时腹腔注射硝普钠建立血管性痴呆大鼠模型,选出造模成功者随机分为模型组及葛根素干预组,各为24只,另以条件匹配的24只大鼠为假手术组。
分别在造模术后15d,1、2和4个月等时间点,采用水迷宫检测大鼠学习记忆能力的变化,HE染色和免疫组化染色观察大鼠海马神经元的形态学改变及BDNF表达的变化。
结果显示:(1)模型组大鼠的逃逸潜伏期(EL)均明显长于假手术组(P<0.01),葛根素干预组大鼠的EL较模型组明显缩短(P<0.05),但仍长于假手术组(P<0.05);(2)模型组大鼠海马CA1区锥体细胞数比假手术组明显减少(P<0.01),葛根素干预组2个月和4个月时点锥体细胞数较模型组明显增多(P<0.01),但仍少于假手术组(P<0.01);(3)模型组大鼠海马BDNF阳性细胞明显减少(P<0.01),除15d和1个月组DG区外,葛根素干预组大鼠海马BDNF 阳性细胞数较模型组明显增多(P<0.05),但仍低于假手术组(P<0.05);(4)模型组大鼠海马BDNF阳性细胞平均吸光度值较假手术组明显降低(P<0.01),而葛根素干预组比模型组和假手术组均明显降低(P<0.05)。
本研究结果提示,脑缺血再灌注后,海马BDNF阳性神经元和锥体细胞持续减少,在VD学习记忆障碍的发生和发展过程中起重要作用;葛根素对脑缺血再灌注损伤具有保护作用,其机制可能与葛根素上调BDNF的表达、减少锥体细胞的丢失有关。
【总页数】6页(P615-620)【关键词】血管性痴呆;脑源性神经营养因子;葛根素;学习记忆;大鼠【作者】张冰清;王玉良【作者单位】潍坊医学院生理学教研室【正文语种】中文【中图分类】Q426【相关文献】1.养血清脑颗粒对血管性痴呆大鼠海马CA1区BDNF和bFGF蛋白表达的影响[J], 张晋霞;伞云琨;李晶;马原源;刘斌2.丰富环境对血管性痴呆大鼠学习记忆和海马区BDNF、TrkB表达的影响 [J], 林俏明;范玉华;李妙嫦3.电针对血管性痴呆大鼠记忆力及海马BDNF、PSD-95蛋白表达的影响 [J], 梁慧英; 廖琳; 游国清; 林阳阳; 燕铁斌; 董军涛4.艾地苯醌对血管性痴呆大鼠海马BDNF mRNA及受体TrkB mRNA表达的影响[J], 钱旭东; 王东; 徐倩倩; 李国芸; 王红梅; 张晓璇; 马征5.山茱萸多糖对血管性痴呆大鼠海马BDNF和NGF表达的影响 [J], 李永格因版权原因,仅展示原文概要,查看原文内容请购买。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
NEURAL REGENERATION RESEARCH Volume 7, Issue 6, February 2012Cite this article as: Neural Regen Res. 2012;7(6):421-425.421Haiqin Wu ☆, M.D.,Professor, Department of Neurology, the Second Affiliated Hospital, Medical College of Xi’an Jiaotong University, Xi’an 710004, Shaanxi Province, ChinaCorresponding author: Haiqin Wu, Department of Neurology, the Second Affiliated Hospital, Medical College of Xi’an Jiaotong University, Xi’an 710004, Shaanxi Province, China whq60@Received: 2011-10-10 Accepted: 2012-01-16 (N20110804001/YJ)Wu HQ, Wang HQ, Zhang B, Zhang GL, Zhang R, Zhang LF. Puerarin decreases hypoxia inducible factor-1 alpha in the hippocampus of vascular dementia rats. Neural Regen Res. 2012;7(6):421-425.doi:10.3969/j.issn.1673-5374.2012.06.003Puerarin decreases hypoxia inducible factor-1 alpha in the hippocampus of vascular dementia rats ☆Haiqin Wu, Huqing Wang, Bei Zhang, Guilian Zhang, Ru Zhang, Lingfeng ZhangDepartment of Neurology, the Second Affiliated Hospital, Medical College of Xi’an Jiaotong University, Xi’an 710004, Shaanxi Province, ChinaAbstractIn this study, a rat vascular dementia model was established by permanent bilateral common carotid arterial occlusion. Rats were intraperitoneally injected with puerarin 3 days before modeling, for 45 successive days. Results demonstrated that in treated animals hippocampal structures were clear, nerve cells arranged neatly, and cytoplasm was rich in Nissl bodies. The number of cells positive for hypoxia inducible factor-1 alpha, erythropoietin and endothelial nitric oxide synthase was reduced; and the learning and memory abilities of rats were significantly improved. Our experimental findings indicate that puerarin can significantly improve learning and memory in a vascular dementia model, and that the underlying mechanism may be associated with the regulation of the expression of hypoxia inducible factor-1 alpha.Key Words: puerarin; vascular dementia; hypoxia-inducible factor-1 alpha; erythropoietin; endothelial nitric-oxide synthaseINTRODUCTIONDonepezil, Huperzine and othercholinesterase inhibitors are the drugs presently approved and widely used for treatment of vascular dementia. However, these drugs do not always producesatisfactory results and have side effects [1-2]. Puerarin, a major active ingredient of the traditional Chinese medicine pueraria radix, is classified as one of the isoflavones. It can dilate vessels and improve microcirculation [3]. It is widely prescribed for patients with cardiovascular and cerebrovasculardiseases [4-8]. Clinical application of puerarin for vascular dementia patients has obtained good curative effects [9], obviously improving the patient’s prognosis.Erythropoietin (EPO) and endothelial nitric oxide synthase (eNOS) are criticaldownstream factors of the hypoxia-inducible factor-1 alpha (HIF-1α) pathway [10-12] involved in the regulation of ischemictolerance. Brain-derived EPO antagonizes toxicity from excitatory amino acids and free radicals, has anti-inflammatory andanti-apoptotic effects, enhances synaptic transmission, and has neurotrophicfactor-like effects [13-15]. When brain tissue undergoes ischemic injury, eNOS in nerve cells and endothelial cells generates protective nitric oxide, which induces angiogenesis, enhances the function of ischemic tissue revascularization andpromotes recovery of brain function [16-17]. Through observing the effects of puerarin on HIF-1α and the related factors EPO and eNOS in the hippocampus of vasculardementia rats, this study sought to explore the mechanism underlying puerarin in the treatment of vascular dementia.RESULTSQuantitative analysis of experimental animalsThirty-five rats were used in the experiment. The rats were randomly divided into three groups: sham-operated (n = 10), dementia (n = 13) and puerarin (n = 12). Rats in the puerarin and dementia groups were treated with a permanent bilateral common carotid arterial occlusion. Rats in the puerarin group were injected intraperitoneally with puerarin for 45 days, beginning 3 days before surgery. In the dementia group, two rats died during modeling and one died during the intervention. In the puerarin group, two rats died during the modeling. Thirty rats were involved in the final analysis of results, with 10 rats in each group.Effects of puerarin on behaviors of vascular dementia ratsAfter ligation, rats in the dementia group showed blepharoptosis, excitation, and ataxia. They quickly recovered withoutdyskinesia, but spontaneous movement was reduced and bradykinesia remained. The symptoms of rats in the puerarin group weremuch improved compared with those in the dementia group. There was no significant change in thesham-operated group before and after ligation. Puerarin enhanced the learning and memory abilities of vascular dementia ratsCompared with the sham-operated group, thelearning-memory scores of both the dementia and puerarin groups were significantly decreased 45 days after ligation of the common carotid arteries (P < 0.01). However, the puerarin group showed improvedlearning-memory scores compared with the dementia group (P < 0.05; Table 1).Puerarin ameliorated the hippocampal morphological changes in vascular dementia ratsIn the sham-operated group, light microscopy showed that hippocampal structures were clear, nerve cells were arranged regularly, and cytoplasm was rich in Nissl bodies. In contrast, in the dementia group, hippocampal structures were chaotic, nerve cells were enlarged and sparsely distributed, and cytoplasmic Nissl bodies were decreased or absent. In the puerarin group hippocampal structures were clearer than in the dementia group, nerve cells were regularly arranged and cytoplasm was rich in Nissl bodies (Figure 1). Puerarin inhibited the expression of HIF-1α, EPO and eNOS in the hippocampus of vascular dementia ratsImmunohistochemical staining showed that hippocampal tissues of all three groups expressed HIF-1α, EPO and eNOS. The immunoreactive products were stained brown. The positive cells were mainly distributed in the pyramidal cell layer in the CA1 region of the hippocampus, consistent with the characteristics of pyramidal cells (Figure 2). HIF-1α, EPO and eNOS positive cells in the CA1 region of the hippocampus were markedly increased in both the dementia and puerarin groups compared with thesham-operated group. Significant decreases in HIF-1α, EPO and eNOS positive cells were observed in the puerarin treated group compared with the control group (Table 2).Figure 1 Hippocampal morphology in rats from different groups (Nissl staining, × 400).(A) Sham-operated group: Hippocampal structures were clear, nerve cells arranged neatly, cytoplasm was rich in Nissl bodies.(B) Dementia group: Hippocampal structures were chaotic, nerve cells were enlarged and sparsely arranged, andNissl bodies decreased or disappeared.(C) Puerarin group: Hippocampal structures were clearer than in the dementia group. Neurons were neatly arranged and rich in cytoplasmic Nissl bodies.A B CFigure 2 Expression of hypoxia-inducible factor-1 alpha 1α), erythropoietin (EPO) and endothelial nitric-oxide422DISCUSSIONIn this study, a cerebral ischemia model was produced by occlusion of bilateral common carotid arteries. After ligation, rats in the dementia group showed blepharoptosis, excitation, and ataxia. They quickly recovered without dyskinesia, but spontaneous movement was reduced and bradykinesia remained. The learning-memory scores of the dementia group rats were markedly decreased in the Y-shaped water maze test. In the dementia group, hippocampal structures were chaotic; nerve cells were enlarged and arranged sparsely; and Nissl bodies in the cytoplasm were decreased or absent. Thus, this animal model, which is simple to prepare and shows features in line with clinical symptoms of vascular dementia, is an ideal model for studying vascular dementia.Puerarin is widely prescribed for patients with ischemic cardiovascular and cerebrovascular diseases. The therapeutic dose is 200-400 mg per day according to the description. There is no definite conclusion in the literature about the drug dose of puerarin for use in animal experiments. According to previously described findings[18-19] and our previous studies and preliminary experiments, we found that an intraperitoneal injection of 100 mg/kg was the optimal dose for treatment of vascular dementia rats. In the present study, the puerarin group remained healthy and demonstrated a better learning and memory performance than the dementia group. Histological examination showed a significant increase in the number of Nissl bodies, clear hippocampal structures, and significant changes in expression of HIF-1α, EPO and eNOS in the hippocampal CA1 region. Such structural and functional changes suggested that an intraperitoneal injection of 100 mg/kg puerarin is appropriate for vascular dementia in rats.Previous studies investigating the effects of puerarin on HIF-1α, EPO and eNOS of hypoxic-ischemic animals showed varied results. In the cerebralischemia-reperfusion model, Luo et al [18] found that the expression of EPO in cortex and basal ganglia in the cerebral ischemic-reperfusion rats was significantly increased after puerarin treatment. In contrast, results from Gu et al [19] showed that expression of eNOS in the brain tissue of rats with cerebral ischemia was decreased after puerarin treatment, but was still higher compared with the sham-operated group. In the rat model of myocardial infarction, Zhang et al[20] discovered that the expression of HIF-1 mRNA and eNOS mRNA in myocardial tissue in the puerarin group was significantly decreased. As reported by Wen et al [21], puerarin can elevate the level of eNOS protein and mRNA in myocardial cells. In this study, puerarin improved learning and memory, improved the hippocampal formation, and increased the intraneuronal Nissl bodies in vascular dementia rats, showing that it has protective effects against vascular dementia. Furthermore, in the hippocampal CA1 region, we observed the expression of HIF-1α, EPO and eNOS. Significantly more positive cells were present in the dementia group compared with the sham-operated group. The results indicated that HIF-1α, EPO and eNOS were closely related to vascular dementia. Following induction of vascular dementia, hippocampal neurons showed hypoxic-ischemic injury. HIF-1α was increased and played a role in self-protection by up-regulating the expression of EPO and eNOS which have neuroprotective effects. In this study, there were fewer positive cells in the puerarin group than in the dementia group, suggesting that puerarindown-regulated the expression of HIF-1α, EPO and eNOS in ischemic hippocampal CA1. This result is different from previous studies. We speculate that the possible causes are as follows: (1) Through increasing blood flow and improving cerebral microcirculation, puerarin improves hippocampal perfusion and increases oxygen concentration in nerve cells[7], thus negatively regulating the expression of HIF-1α and downstream protective factors EPO and eNOS. (2) Puerarin possesses an estrogen substitution effect[3, 22]. It is likely to increase tolerance of cells for hypoxia by acting at estrogen receptors, therefore down-regulating the expression of HIF-1α and t he downstream protective factors EPO and eNOS. Indeed, in this study puerarin down-regulated the expression of the protective HIF-1α pathway and downstream factors EPO and eNOS. Additionally, the learning and memory abilities of puerarin treated rats were significantly improved compared with the dementia group, but were still poorer than the sham-operated group. At the same time, there were fewer positive cells in the puerarin group than in the dementia group, but more than in the sham-operated group. This shows that puerarin can improve clinical symptoms of vascular dementia but cannot completely reverse the process of vascular dementia. Therefore, the most important thing is to prevent the occurrence of vascular dementia in clinic.Our study confirms that puerarin can improve clinical symptoms of vascular dementia and demonstrates that this action is associated with down-regulation of HIF-1α and its downstream factors, EPO and eNOS.423MATERIALS AND METHODSDesignA randomized, controlled animal experiment.Time and settingThe study was performed at the Medical College of Xi’an Jiaotong University, China from December 2009 to September 2010.MaterialsAnimalsA total of 35 male, specific pathogen freeSprague-Dawley rats, weighing 280 ± 20 g, aged 2- 3 months, were provided by the Experimental Animal Center of Medical School of Xi’an Jiaotong University, China (certificate No. SCXK (Shaan) 2007-001). Rats were housed on a 12-hour light/dark cycle, at 23.0 ±0.5°C and a relative humidity of 50.0 ± 0.5%, with free access to food and water. Animal procedures were in strict accordance with the Guidance Suggestions for the Care and Use of Laboratory Animals, issued by the Ministry of Science and Technology of China[23].DrugsPuerarin injection (4, 7-dihydroxy-8-β-D-glucosyl- isoflavone; Ankang Pharmaceutical Factory, Shaanxi, China; lot No. H20053144, 100 mg: 2 mL/bottle). The structure of puerarin is shown in Figure 3.MethodsEstablishment of vascular dementia rat models According to the methods described by de la Torre et al [24], rats in the puerarin group and dementia group were treated with a permanent bilateral common carotid arterial occlusion under anesthesia with peritoneal injection of 10% hydral chlorate (300 mg/kg). A longitudinal incision was made in the ventral neck to expose the common carotid arteries which were carefully dissected free from the vagus nerve and adjacent tissues. Then the common carotid artery was ligated with a silk suture. Animals assigned to the sham-operated group underwent the same surgical procedure without vessel occlusion. Puerarin interventionFrom 3 days before surgery, animals in the puerarin group were injected intraperitoneally with puerarin at a dose of 100 mg/kg per day for 45 days. Animals were injected 1 hour before ligation in the day of surgery. At the same time, animals in the sham-operated group and dementia group were injected intraperitoneally with 0.9% sodium chloride solution at a dose of 100 mg/kg.Y-type water maze test for detection of rat behavior The learning-memory abilities of rats were assessed in the Y-type water maze test[25]. The water maze consisted of a long arm (50 cm × 30 cm × 45 cm) and two short arms (40 cm × 30 cm × 45 cm). The long arm was the initiation region. One short arm was the blind zone (on the top of which there was an opaque plastic board, thereby decreasing the visibility of the canal); the other short arm was the safety zone with a platform of 30 cm ×10 cm and lamplight. Rats were trained over ten-trials. Their latency to arrive at safety zone from initiation region and the numbers of errors displayed by entering the blind zone were recorded as learning performances. After a 24-hour interval, the rats were tested again to obtain their memory performances. Before the experiment, the rats whose latency to arrive at safety zone were longer than 25 seconds were eliminated. After 45 days of intervention, the learning and memory abilities of the rats were once again evaluated with the Y-type water maze test.Collection of hippocampal samplesAfter 45 days of intervention, the rats were anesthetized, the chest was opened, the animals were perfused through the heart and the whole brain was removed. Brain tissues including hippocampus were then taken from coronal sections located from 3 mm after the bregma to the superior colliculus[26], and were paraformaldehyde fixed and paraffin-embedded.Nissl staining for cell morphology in the rat hippocampusThe hippocampal tissues were cut into 3-μm thick paraffin sections. They were deparaffinized with two changes of xylene for 15 minutes each, followed by 95% alcohol and 80% alcohol for 5 minutes each, then washed for 5 minutes with running water. The sections were next incubated in 1% toluidin blue (Tianyuan Biotechnology Co., Ltd., Shanghai, China) at 55°C for 30 minutes, and rinsed for 5 minutes. The sections were then rinsed with 80% alcohol, 90% alcohol, and 95% alcohol for 5 minutes each, then with three changes of xylene for 15 minutes each[27]. Last, the sections were mounted with neutral gum. The sections were observed under an optical microscope (B-type biomicroscope; Global Motic Group, Xiamen, China). Immunohistochemical staining for HIF-1α, EPO and eNOs expressionThe streptavidin-peroxidase immunohistochemical staining was executed in hippocampal tissues sections according to the kit instructions (Boster Biological Engineering Company, Wuhan, China). In brief, hippocampal tissue sections underwent dehydration through the ethanol gradient. The sections were treated with 3% H2O2 for 40 minutes at room temperature, microwave treated for 10 minutes for antigen retrieval, and cooled for 2 hours at room temperature. After incubation for 40 minutes in normal rabbit serum, they were incubated with primary antibodies, including rabbit anti-HIF-1α poly clonal antibody (1: 200; Boster Biological Engineering Company, Wuhan, China)[28], sheepanti-EPO multiclonal antibody (1: 500; Santa CruzFigure 3 Structure of puerarin.424Biotechnology, Delaware Santa Cruz, CA, USA)[29] and rabbit anti-eNOS polyclonal antibody (1: 100; Boster Biological Engineering Company)[19]. Subsequently, they were kept at 4°C overnight, treated with the biotinylated secondary antibodies, sheep anti rabbit IgG (HIF-1α, eNOS, 1: 200) or horse anti sheep IgG (EPO, 1: 500) for 40 minutes at room temperature, and incubated for30 minutes in streptavidin with horseradish peroxidase (1: 200) at room temperature. This was followed by DAB for 5 minutes, counterstaining with hematoxylin, dehydration, and coverslipping. Negative control sections were treated with TBS instead of primary antibody.Six sections from each group were observed under an optical microscope. The positive cells were nerve cells in hippocampus showing buffy granules. The data were expressed as the mean of number of positive cells/mm2 in five random high-power fields (400 × magnification) from the CA1 region of the hippocampus.Statistical analysisData were analyzed using SPSS 12.0 software (SPSS, Chicago, IL, USA) and a level of P < 0.05 was assessed as a statistically significant difference. All values in the figures of present study indicate mean ± SD. Group comparisons were analyzed with analysis of two-sample t-test.Author contributions: Haiqin Wu designed this study and revised the manuscript. Huqing Wang and Bei Zhang provided experimental data, participated in statistical analysis and wrote the manuscript. Guilian Zhang and Ru Zhang provided technical support and statistical calculations. Lingfeng Zhang provided information support.Conflicts of interest:None declared.Ethical approval:This study had the approval of the Animals Ethics Committee of Medical College of Xi’an Jiaotong University in China.Acknowledgments:We would like to thank Ming Li, from Department of Anatomy, Medical College of Xi’an Jiaotong University, China for technical support.REFERENCES[1] Román GC, Salloway S, Black SE, et al. Randomized,placebo-controlled, clinical trial of donepezil in vascular dementia: differential effects by hippocampal size. Stroke. 2010;41(6):1213-1221.[2] Wilkinson D, Róman G, Salloway S, et al. The long-term efficacyand tolerability of donepezil in patients with vascular dementia. Int J Geriatr Psychiatry. 2010;25(3):305-313.[3] Yin LH, Li YF, Meng FL. Puerarin's chemical compositionpharmacological effects and clinical application. HeilongjiangYiyao. 2010;23(3):371-373.[4] Sun GH. A review on the clinical application of Puerarin. ZhongyiLinchuang Yanjiu. 2010;2(6):100-101.[5] Ji AL, Zhang XJ. Effect of puerarin injection on inflammatoryfactors in patients with acute cerebral infarction. NanjingZhongyiyao Daxue Xuebao: Ziran Kexue Ban. 2009;25(2):145-147.[6] Yao PF, Fan HX, Qian YL. Study of puerarin injection on acuteischemic brain infarction in the aged. Xiandai Zhongxiyi JieheZazhi. 2009;18(14):1587-1590. [7] Yu WH, Chu GL. Researching development of the protectiveeffects of puerarin in hypoxic ischemic brain damage. YixueZongshu. 2009;15(20):3163-3165.[8] Zhang R, Cheng HX, Wu HQ, et al. Effect of Puerarin on nNOSexpression in hippocampal neurons after focal cerebral ischemiain rats. Xi'an Jiaotong Daxue Xuebao: Yixue Ban. 2010;31(6):669-672.[9] Zhang T, Zhang XQ, Zhu RH. The report of puerarin and citicolinein 36 cases with vascular dementia. Shandong Zhongyiyao Daxue Xuebao. 2000;24(1):37-38.[10] Semenza GL. HIF-1: upstream and downstream of cancermetabolism. Curr Opin Genet Dev. 2010;20(1):51-56.[11] Corzo CA, Condamine T, Lu L, et al. HIF-1α regulates functionand differentiation of myeloid-derived suppressor cells in thetumor microenvironment. J Exp Med. 2010;207(11):2439-2453. [12] Takubo K, Goda N, Yamada W, et al. Regulation of the HIF-1alphalevel is essential for hematopoietic stem cells. Cell Stem Cell.2010;7(3):391-402.[13] Noguchi CT, Asavaritikrai P, Teng R, et al. Role of erythropoietin inthe brain. Crit Rev Oncol Hematol. 2007;64(2):159-171.[14] Zhang F, Xing J, Liou AK, et al. Enhanced delivery oferythropoietin across the blood-brain barrier for neuroprotectionagainst ischemic neuronal Injury. Transl Stroke Res. 2010;1(2):113-121.[15] Genc K, Egrilmez MY, Genc S. Erythropoietin induces nucleartranslocation of Nrf2 and heme oxygenase-1 expression inSH-SY5Y cells. Cell Biochem Funct. 2010;28(3):197-201.[16] Ito Y, Ohkubo T, Asano Y, et al. Nitric oxide production duringcerebral ischemia and reperfusion in eNOS- and nNOS-knockoutmice. Curr Neurovasc Res. 2010;7(1):23-31.[17] Aoki T, Nishimura M, Kataoka H, et al. Complementary inhibitionof cerebral aneurysm formation by eNOS and nNOS. Lab Invest.2011;91(4):619-626.[18] Luo YM, Gao L, Ji XM, et al. Effects of puerarin on EPOexpressions in ischemic brain tissues of rats following MCAO.Zhongguo Laonian Xue Zazhi. 2008;28(12):1057-1059.[19] Gu CZ, Lu HL, Wu QH, et al. The effects of puerarin on theepression of eNOS in brain tissues after cerebral ischemiareperfusion in rats. Zuzhong yu Shenjing Jibing. 2010;17(6):336-338.[20] Zhang SY, Shen YJ, Chen SL, et al. The effects of puerarin onHIF-1, eNOS, VEGF genes expressions in myocardium and NO in serum of rats with myocardial infarction. Zhongyi Yaoli yuLinchuang. 2007;23(5):54-57.[21] Wen J, Chen SL, Filly C, et al. Effect of puerarin on nitric oxideproduction in H9C2 cells. Zhongguo Xinxueguan Bing Zazhi.2006;4(10):777-779.[22] Huang T, Jin BQ, Sun GJ, et al. Effects of puerarin on the bonemetabolism in ovariectomized rats. Zhongguo Laonian Xue Zazhi.2009;29(19):2482-2484.[23] The Ministry of Science and Technology of the People’s Republicof China. Guidance Suggestions for the Care and Use ofLaboratory Animals. 2006-09-30.[24] de la Torre JC, Fortin T, Park GA, et al. Chronic cerebrovascularinsufficiency induces dementia-like deficits in aged rats. Brain Res.1992;582(2):186-195.[25] Wang CF, Li DQ, Xue HY, et al. Oral supplementation of catalpolameliorates diabetic encephalopathy in rats. Brain Res. 2010;1307:158-165.[26] Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates.5th ed. London: Academic Press. 2005.[27] Yu SS, Zhao J, Zheng WP, et al. Neuroprotective effect of4-hydroxybenzyl alcohol against transient focal cerebral ischemiavia anti-apoptosis in rats. Brain Res. 2010;1308:167-175.[28] Peng Z, Ren P, Kang Z, et al. Up-regulated HIF-1alpha is involvedin the hypoxic tolerance induced by hyperbaric oxygenpreconditioning. Brain Res. 2008;1212:71-78.[29] Wu H, Wang H, Sha J, et al. Expression of hypoxia induciblefactor-1alpha and erythropoietin in the hippocampus of aging rats.Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2009;34(9):856-860.(Edited by Wang XD, Shi ZH/Yang Y/Wang L)425。