用户名: 密码: 验证码:
手机频率电磁辐射对大鼠肝脏的影响及西洋参胶囊对其干预作用的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:以900MHz手机频率电磁辐射为干预因素,以大鼠肝脏组织为研究对象,以肝功能、组织形态学、抗氧化酶活性、Nrf2蛋白表达、DNA损伤及损伤修复蛋白ATM、γ-H2AX表达和大鼠肝细胞周期及凋亡的变化为主要观测指标,选择西洋参胶囊和水飞蓟宾胶囊作为实验用药,观察常见手机频率的电磁辐射对大鼠肝损伤的可能机制和西洋参胶囊对其作用。
     手机频率电磁辐射对大鼠肝脏的影响(论文第一、二、三部分)
     方法:
     1造模及分组清洁级SD雄性大鼠50只,体重200~220g。随机分正常组和辐射6d、12d、20d、30d组,共计5个实验组,每组10只,共50只。除正常组不辐射外,其余各组均置辐射笼内,打开自动开关每天4小时,分别连续辐射6d、12d、20d和30d,每天除辐射外正常饲养。
     2取材及检测指标各组实验结束后禁食12小时,10%水合氯醛腹腔麻醉后处死,股动脉取血离心取血清测ALT、AST含量;取肝脏称重后将相同部位肝组织(约0.3×0.5×0.5cm3)置于4%多聚甲醛溶液固定,用于HE染色;比色法检测肝组织MDA、SOD、GSH、GSH-PX含量;采取免疫组化及Western blot法检测Nrf2及ATM、γH2AX蛋白表达;流式细胞仪检测各组大鼠肝细胞周期和凋亡率。
     结果:
     1手机频率电磁辐射对大鼠肝脏功能的影响
     1各组大鼠一般情况观察
     各组大鼠的精神状态、进食水量、排泄、皮毛、活动等情况无显著差异。
     1.2各组大鼠血清ALT、AST的比较
     与正常组大鼠相比较,辐射30d大鼠血清AST水平显著性增高,(P<0.05),辐射6d、12d、20d组大鼠无显著变化(P>0.05);与正常组大鼠相比较,辐射6d、12d、20d、30d组大鼠血清ALT水平无显著变化(P>0.05)。
     1.3各组大鼠肝脏组织形态学变化的比较
     正常组肝组织肝小叶的结构清晰完整,肝细胞界限明显,大小基本相同,1~2个圆形或类圆形细胞核位于细胞中间。辐射6d组肝小叶结构紊乱,可见细胞界限不清,区域性肝细胞变性,细胞核消失,细胞浆出现空泡样变化,组织形态学异常。12d组肝小叶结构完整,肝细胞分界清,个别细胞核固缩或消失,组织形态学异常。20d组肝小叶结构完整,肝细胞分界清,个别细胞核固缩或消失。30d组肝小叶结构完整,肝细胞分界清,出现较明显的肝细胞核固缩现象,组织形态学异常。
     2手机频率电磁辐射对大鼠肝氧化损伤及抗氧化酶、Nrf2蛋白表达的影响
     2.1各组大鼠肝组织MDA、SOD、GSH、GSH-PX含量的比较
     与正常组大鼠相比,各辐射组大鼠肝组织MDA水平均显著增高(P<0.05)。辐射12d、30d组大鼠肝组织SOD含量均显著减少(P<0.05);辐射6d、20d组大鼠SOD含量无显著性变化(P>0.05)。各辐射组大鼠肝组织GSH水平均显著减少(P<0.05)。辐射20d、30d组大鼠肝组织GSH-PX水平均显著降低(P<0.05)。
     2.2免疫组化法检测大鼠肝细胞Nrf2蛋白表达
     正常组大鼠肝细胞浆中有Nrf2表达,肝细胞核内无明显表达,各辐射组均细胞核有Nrf2表达。与正常组相比,辐射6d、12d、20d、30d,Nrf2在肝细胞的表达出现先增强,后稍弱再增强的变化,Nrf2蛋白表达由强到弱顺序为:辐射6d>12d>30d>20d>正常组。辐射6d与12d之间差异不显著;20d与30d之间差异不显著。但辐射6d、12d与20d、30d之间差异显著(P<0.05)。
     2.3Western blot检测各组大鼠肝组织Nrf2蛋白表达
     与正常组相比,各辐射组大鼠肝组织Nrf2蛋白表达平均灰度比值明显增强(P<0.01);与免疫组化结果相符,辐射6d、12d、20d、30d,Nrf2在肝细胞的表达出现先增强,后稍弱再增强的变化,Nrf2蛋白表达由强到弱顺序为:辐射6d>12d>30d>20d>正常组。
     3手机频率电磁辐射对大鼠肝细胞DNA损伤修复蛋白ATM、H2AX表达及细胞周期和凋亡率的影响
     3.1免疫组织化学法检测ATM、γH2AX蛋白在各组大鼠肝组织的表达
     ATM蛋白在各组大鼠肝细胞核和细胞浆均有表达。与正常组大鼠相比,辐射12d、20d、30d组大鼠肝细胞核和细胞浆中表达均显著增强(P<0.01),随着辐射时间的延长,ATM蛋白表达平均光密度增加(P<0.05)。
     γH2AX在各组大鼠肝细胞核和细胞浆均有表达,与正常组大鼠相比,辐射6d、12d、20d、30d组大鼠肝细胞核和细胞浆中表达均显著增强(P<0.05;P<0.05;P<0.05;P<0.01);随着辐射时间的延长,γH2AX蛋白表达增加。
     3.2Western blot检测ATM、H2AX蛋白在大鼠肝组织的表达
     与正常组大鼠比较,辐射6d、12d、20d、30d组大鼠肝组织ATM蛋白的平均灰度比值逐渐增加(P<0.01)。
     与正常组大鼠比较,辐射6d、12d、20d、30d组大鼠肝组织H2AX蛋白的平均灰度比值显著增加,有统计学意义,(P<0.05,P<0.01,P<0.01,P<0.01)。
     3.3各组大鼠肝细胞周期的比较
     G0/G1期细胞:与正常组相比,辐射6d、12d、20d、30d组大鼠G0/G1期细胞比例均显著增加(P<0.01;P<0.05;P<0.01;P<0.01)。
     S期%细胞:与正常组相比,辐射6d、12d、20d组大鼠S期%细胞无显著性变化(P>0.05);辐射30d大鼠S期%细胞数量显著减少(P<0.05)。
     G2/M期%细胞:与正常组相比,辐射6d、12d、20d组大鼠G2/M期%细胞均显著性减少(P<0.01)。
     3.4各组大鼠肝细胞凋亡率的比较
     与正常组相比,辐射6d、12d、20d、30d组大鼠肝细胞凋亡率极显著增加(P<0.01);与辐射6d、12d和20d组相比,辐射30d组增加有显著性差异(P<0.05)。
     结论:
     1长时间手机频率的电磁辐射是一种致病因素,可导致肝组织形态学损伤,具有时间效应关系。
     2手机频率电磁辐射致大鼠肝损伤与抗氧化酶活性的变化及Nrf2表达变化具有相关性。
     3手机频率电磁辐射致大鼠肝组织形态学异常与肝细胞DNA损伤,激活修复相关的ATM和H2AX蛋白表达,影响细胞周期和细胞凋亡。西洋参胶囊对手机频率电磁辐射大鼠肝损伤的干预作用(论文第四部分)
     方法:
     1分组清洁级SD雄性大鼠40只,分为正常组、模型组(辐射12d组)、水飞蓟宾组(辐射12d加水飞蓟宾组)、西洋参胶囊组(辐射12d加西洋参胶囊组),各10只。
     2取材及检测指标各组实验结束后禁食12h,10%水合氯醛腹腔麻醉后处死,股动脉取血离心取血清测ALT、AST含量;取肝脏称重后将相同部位肝组织(约0.3×0.5×0.5cm3)置于4%多聚甲醛溶液固定,用于HE染色;比色法检测肝组织MDA、SOD、GSH、GSH-PX含量;采取免疫组化及/或Western blot法检测Nrf2及ATM、γH2AX蛋白表达;采用流式细胞法检测各组大鼠肝细胞周期和凋亡率。
     结果:
     1西洋参胶囊对辐射12d大鼠模型肝脏组织形态学的干预作用
     正常组大鼠肝组织细胞结构完整,分界清晰,细胞的大小比较一致,核位于细胞中间,胞质丰富。模型组大鼠肝小叶结构完整,肝细胞核缩小或部分消失,核固缩明显。与模型组相比,水飞蓟宾组和西洋参胶囊组大鼠肝细胞萎缩明显减轻,接近正常。
     2西洋参胶囊对辐射12d大鼠模型肝脏组织氧化损伤指标的干预作用
     与正常组比较,模型组MDA含量显著升高(P<0.05),SOD、GSH含量显著降低(P<0.05)。与模型组比较,水飞蓟宾组MDA含量显著降低(P<0.05),SOD、GSH、GSH-PX含量显著升高(P<0.05);西洋参胶囊组MDA含量显著降低(P<0.05),SOD、GSH含量显著升高(P<0.01,P<0.05)。
     3西洋参胶囊对辐射12d大鼠模型肝脏组织Nrf2蛋白表达的干预作用
     3.1免疫组化检测结果
     正常组仅在大鼠肝细胞浆中有Nrf2蛋白表达,肝细胞核内无明显表达,其余三组均主要在细胞核有Nrf2蛋白表达。与正常组比较,模型组大鼠肝脏组织Nrf2蛋白表达平均吸光度值显著升高(P<0.05),胞浆和胞核中均有表达。与模型组比较,水飞蓟宾组和西洋参胶囊组大鼠肝脏组织Nrf2蛋白表达平均吸光度值均显著降低(P<0.05)。
     3.2Western blot检测结果
     与正常组比较,模型组大鼠肝脏组织Nrf2蛋白表达平均灰度比值升高(P<0.01)。与模型组比较,水飞蓟宾组和西洋参胶囊组大鼠肝脏组织Nrf2蛋白表达平均灰度比值均降低(P<0.05,P<0.01)。
     4西洋参胶囊对辐射12d大鼠模型肝脏组织ATM、H2AX的干预作用
     与正常组比较,辐射后大鼠肝组织ATM平均灰度比值显著增加,表达增强(P<0.05)。与模型组比较,水飞蓟宾组大鼠肝组织ATM平均灰度比值显著降低,表达减弱(P<0.05);西洋参胶囊组大鼠组织ATM平均灰度比值显著降低,表达减弱(P<0.01)。
     与正常组比较,辐射后大鼠肝组织H2AX平均灰度比值显著增加,表达增强(P<0.01)。与模型组比较,水飞蓟宾组大鼠肝组织H2AX平均灰度比值显著降低,表达减弱(P<0.05);西洋参胶囊组大鼠组织H2AX平均灰度比值显著降低,表达减弱(P<0.01)。
     5西洋参胶囊对辐射12d大鼠模型肝细胞周期和细胞凋亡率的干预作用
     5.1西洋参胶囊对辐射12d大鼠模型肝细胞周期的干预作用
     与正常组大鼠相比较,辐射后大鼠肝细胞G0/G1期%显著增加(P<0.01)。与模型组比较,水飞蓟宾组大鼠G0/G1期肝细胞比例无显著变化(P>0.05);西洋参胶囊组大鼠G0/G1期%肝细胞数极显著降低(P<0.01),接近正常。
     与正常组大鼠相比较,辐射后大鼠肝细胞S期%无显著性变化(P>0.05)。与模型组比较,水飞蓟宾组大鼠肝细胞S期%明显降低(P<0.01);而西洋参胶囊组大鼠肝细胞S期%无显著变化(P>0.05)。
     与正常组大鼠相比较,辐射后大鼠肝细胞G2/M期%极显著减少(P<0.01)。与模型组比较,水飞蓟宾组大鼠肝细胞G2/M期%无显著性差异(P>0.05);西洋参胶囊组大鼠肝细胞G2/M期%极显著增高(P<0.01)。
     5.2西洋参胶囊对辐射12d大鼠模型肝细胞凋亡率的干预作用
     与正常组大鼠相比较,辐射后大鼠肝细胞凋亡率显著增加(P<0.01)。与辐射组比较,水飞蓟宾组大鼠无显著差异(P>0.05);西洋参胶囊组大鼠肝细胞凋亡率显著性降低(P<0.01)。
     结论:
     1长时间手机频率电磁辐射是一种致病因素,可导致肝组织损伤。其致病特点归属六淫之温热之邪,可使气阴耗伤。西洋参具有益气养阴作用,主治气阴不足之证。西洋参胶囊对辐射12d大鼠模型肝损伤具有干预作用。
     2西洋参胶囊可改善肝组织形态学异常变化,与降低氧化损伤、降低Nrf2在肝细胞的表达、降低ATM和H2AX蛋白表达,影响细胞周期和细胞凋亡有关。
     3西洋参胶囊对手机频率电磁辐射所致肝损伤模型具有保护作用。
Objective: This study investigated the possible mechanism of the injuryeffects of cellular phone electromagnetic radiation on mice liver tissues andthe intervention effects of American Ginseng and silymarin capsules on theinjury by observing the changes of liver function, histomorphology,antioxidase activity, Nrf2protein expression, DNA injury, expression of theinjury repair proteins ATM and γ-H2AX, and the mice liver proliferation cycleand apoptosis.
     Effects of cellular phone electromagnetic radiation on mice liver tissues
     Methods:
     1Animal models and groups: Fifty clean grade SD male mice with thebody weight of200-220g were randomized into one normal group and fourirradiation groups with10mice in each group. The four irradiation groupswere divided into four groups with each group receiving irradiation for6,12,20and30days in the irradiation cage. The normal group did not receive anyirradiation. All the mice received normal feeding.
     2Sampling and testing parameters: After the experiment in each group,the mice were fasted for12hours, the mice were sacrificed by injecting10%chloralhydrate into the peritoneum of the mice. Blood sample was taken fromthe femoral artery and centrifuged. The blood serum was used to test the ALTand AST contents. The liver was taken and weighed, and some liver tissues inthe same location were taken (about0.3×0.5×0.5cm3) and fixed in4%paraformaldehyde solution for HE staining. The contents of MDA, SOD, GSHand GSH-PX in the hepatic tissues were tested by use of chromometry. Theprotein expression of Nrf2, ATM and γH2AX was tested by use ofimmunohistochemistry and Western blot. The cell cycle and apoptosis rate ofthe mice liver were tested by use of flow cytometry.
     Results:
     1Effects of cellular phone electromagnetic radiation on mice liver function
     1.1General observation on the mice in each group
     The mice spirit status, food and water intake, excretion, skin, hair,activities and so on were observed, with no significant differences.
     1.2Comparison of serum ALT and AST of the mice in every group
     Compared with the mice in the normal group, the mice serum AST wassignificantly increased (P<0.05) in the radiation group receiving radiation for30days but did not have significant differences (P>0.05) in the radiationgroups for6,12and20days. Compared with the mice in the normal group, nosignificant (P>0.05) differences existed in the serum ALT in any of theradiation groups for6,12,20or30days.
     1.3Comparison of the mice liver histomorphology
     In the normal group, the liver tissue had clear and complete structure ofthe liver lobules with clear liver cell boundaries, similar size and one to twocircular or oval cellular nucleus in the center of the cell. In the6-dayirradiation group, the hepatic lobule did not have a clear structure, and thecellular boundary was not clear. Local cellular degeneration was present in theliver, the cellular nucleus disappeared, and the cytoplasm had vacuolardegeneration, indicating abnormal histomorphology. In the12-day and20-dayirradiation groups, the hepatic lobules were complete, the hepatic cells hadclear boundary, and some individual nucleus had pyknosis or disappeared,indicating abnormal histomorphology. In the30-day irradiation group,apparent pyknosis was present in the liver. However, the hepatic lobules werecomplete, and the hepatic cells had clear boundary, indicating abnormalhistomorphology.
     2Effects of cell phone electromagnetic irradiation on the liver oxidativedamage and the expression of antioxidase and Nrf2protein
     2.1Comparison of the liver MDA, SOD, GSH and GSH-PX in every group
     Compared with the normal group, the MDA level increased significantly(P<0.05) in the mice in every irradiation group, the SOD content decreased significantly (P <0.05) in the12and30day irradiation groups but not in the6or20day irradiation group (P>0.05). The GSH level decreased significantly(P <0.05) in the mice in all the irradiation groups, and the GSH-PX level alsodecreased significantly (P<0.05) in the20and30day irradiation groups ascompared with the normal group.
     2.2Nrf2protein expression of the mice hepatic cells byimmunohistochemistry
     Nrf2was expressed only in the cytoplasm rather than the nucleus in themice hepatic cells in the normal group, whereas Nrf2was expressed in thenucleus in every irradiation group. Compared with the normal group, the Nrf2expression was first increased, a little decreased and enhanced once again afterirradiation for6,12,20and30days. The order of enhanced expression ofNrf2from strong to weak was6-d>12-d>30-d>20-d>the normal group. Nosignificant difference (P<0.05) existed between6-d and12d groups orbetween20-d and30-d groups. But a significant difference (P<0.05) existed ineither6or12day group as compared with either20or30day group.
     2.3Nrf2protein expression in the hepatic cells by Western blot
     Compared with the normal group, the grey level ratio of the Nrf2proteinexpression in the hepatic cells in every irradiation group was significantlyenhanced (P <0.01), consistent with the immunohistochemistry results. Afterirradiation for6,12,20and30days, the Nrf2expression in the hepatic cellswas firstly enhanced, then a little decreased and increased once more, and theorder of Nrf2expression was from strong to weak in the groups:6-d>12-d>30-d>20-d irradiation groups>the normal group.
     3Effects of cell phone electromagnetic irradiation on the mice hepatic injuryrepair protein ATM and H2AX expression and the cell cycle
     3.1Expression of ATM and γH2AX proteins in the hepatic tissues in everygroup by immunohistochemistry
     The ATM protein was expressed in both the nucleus and cytoplasm in thehepatic cells in every group of mice. Compared with the normal group, theexpression of ATM was enhanced significantly (P<0.01) in the12,20and30 day irradiation groups. With the elongation of the irradiation, the mean opticaldensity of the ATM protein expression increased (P<0.05).
     γH2AX was also expressed in both the nucleus and cytoplasm in the micehepatic cells. Compared with the normal group, the γH2AX expression wassignificantly enhanced in the6,12,20and30day irradiation groups (P<0.05,P<0.05, P<0.05, P<0.01, respectively);With the elongation of irradiation,the γH2AX protein expression increased significantly.
     3.2ATM and γH2AX protein expression in mice hepatic tissues by Westernblot
     Compared with the normal group, the mean grey level ratio of the ATMprotein in the mice hepatic tissues was gradually increased significantly (P<0.01) after irradiation for6,12,20and30days.
     Compared with the normal group, the mean grey level ratio of the H2AXprotein in the mice hepatic tissues was gradually increased significantly (P<0.05,P<0.01,P<0.01,P<0.01) after irradiation for6,12,20and30days,respectively.
     3.3Comparison of the cell cycle
     G0/G1stage cells (%): Compared with the normal group, the percentageof the G0/G1stage cells was highly significantly in the6,20and30daygroup(P<0.01;P<0.05; P<0.01; P<0.01).
     S stage cells (%): Compared with the normal group, the percentage of theS stage cells had no significant (P>0.05) decrease in the6,12and20dayirradiation groups but significant decrease (P<0.01) in the30day irradiationgroup.
     G2/M stage cells (%): Compared with the normal group, the percentageof the G2/M stage cells had significant decrease (P<0.01) in the6,12and20day irradiation groups. No significant differences (P>0.05) existed among theirradiation groups.
     3.4Comparison of the Hepatic cell apoptosis rate
     Compared with the normal group, the hepatic apoptosis rate wassignificantly increased in the6,12,20and30day irradiation groups (P<0.01). Compared with the6,12,and20day irradiation groups,the30-d group hadsignificant differences(P<0.05).
     Conclusion:
     1Long-term cell phone electromagnetic irradiation is a cause of diseaseand may lead to the damage to the hepatic histomorphology in a time-efficientmanner.
     2The cell phone electromagnetic irradiation is closely related to thehepatic damage, the antioxidase activity change or the Nrf2expressionchange.
     3The abnormal histomorphology caused by cell phone electromagneticirradiation is closely related to the hepatic DNA damage, activation of therepair-related ATM and H2AX protein expression, and changes of the cellcycle and apoptosis.
     The intervention effects of American ginseng on the hepatic damagecaused by cell phone electromagnetic irradiation
     Methods:
     1Grouping: Forty clean grade SD male mice were randomized into thenormal,12-d irradiation group,12-d irradiation plus silymarin group, and12-dirradiation plus American ginseng group, with10mice in each group.
     2Sampling and testing parameters: After the experiment in each group,the mice were fasted for12hours, the mice were sacrificed by injecting10%chloralhydrate into the peritoneum of the mice. Blood sample was taken fromthe femoral artery and centrifuged. The blood serum was used to test the ALTand AST contents. The liver was taken and weighed, and some liver tissues inthe same location were taken (about0.3×0.5×0.5cm3) and fixed in4%paraformaldehyde solution for HE staining. The contents of MDA, SOD, GSHand GSH-PX in the hepatic tissues were tested by use of chromometry. Theprotein expression of Nrf2, ATM and γH2AX was tested by use ofimmunohistochemistry and Western blot. The cell cycle and apoptosis rate ofthe mice liver were tested by use of flow cytometry.
     Results:
     1The intervention effects of American ginseng capsules on the liver histologyof the mice model in the12-d irradiation group
     In the normal group, the cell structure was clear in the mice liver withdefinite demarcation. The cell had consistent size with the nucleus in the cellcenter and rich cytoplasm. In the12-d irradiation group, the hepatic lobuleshad complete structure, the nucleus of the hepatic cells shrank or partiallydisappeared with apparent karyopyknosis. Compared with the modelgroups,the hepatic atrophy was apparently less severe and was nearly normalin the silymarin or the American ginseng group.
     2The intervention effects of the American ginseng capsules on the oxidativedamage to the hepatic tissues in the12-d group
     Compared with the normal group, the hepatic MDA was significantlyincreased (P<0.05) but the hepatic SOD and GSH were significantly decreased(P<0.05) in the12-d group. Compared with the12-d group, the hepatic MDAwas significantly decreased (P<0.05) but the SOD, GSH and GSH-PXcontents were significantly increased (P <0.05) in the12-d irradiation plussilymarin group. Compared with the12-d group, the hepatic MDA wassignificantly decreased (P<0.05) but the SOD and GSH contents weresignificantly increased (P<0.01and P<0.05, respectively) in the12-dirradiation plus American ginseng group.
     3Intervention effects of American ginseng on the hepatic Nrf2proteinexpression of the12-d irradiation mice group
     3.1Immunohistochemistry
     In the normal group, the Nrf2protein expression was only in the hepaticcytoplasm of the mice with no apparent expression in the hepatic nucleus. TheNrf2protein expression was mainly in the nucleus in the other three groups.Compared with the normal group, the mean optical density absorbance of thehepatic Nrf2protein expression in the12-d irradiation mice model wassignificantly increased (P<0.01), with expression in both the cytoplasm andthe nucleus. Compared with the12-d irradiation group, the mean optical density absorbance of the hepatic Nrf2protein expression was significantly(P<0.005) decreased in the12-d irradiation plus silymarin or Americanginseng group.
     3.2Western blot results
     Compared with the normal group, the mean grey level ratio of the Nrf2protein expression in the12-d irradiation group was significantly (P<0.01)increased. Compared with the12-d irradiation group, the mean grey level ratioof the Nrf2protein expression in both the12-d irradiation plus silymarin andAmerican ginseng groups was significantly (P<0.05and P<0.01,respectively) decreased.
     4Intervention effects of American ginseng on the hepatic ATM and H2AX inthe12-d irradiation group
     Compared with the normal group, the mean grey level ratio of the hepatictissue ATM increased significantly (P<0.05) after irradiation with enhancedexpression. Compared with the12-d irradiation group, the mean grey levelratio of the hepatic tissue ATM in the12-d irradiation plus silymarin orAmerican ginseng group decreased significantly (P<0.05and P<0.01,respectively) with decreased expression.
     Compared with the normal group, the mean grey level ratio of the H2AXin the hepatic tissue in the12-d irradiation group increased significantly (P<0.05) with increased expression. Compared with the12-d irradiation group,the mean grey level ratio of the hepatic tissue H2AX in the12-d irradiationplus silymarin or American ginseng group decreased significantly (P<0.05andP <0.01, respectively) with decreased expression.
     5Intervention effects of American ginseng on the cell cycle and apoptosis rateof the12-d irradiation mice
     5.1Intervention effects of American ginseng on the cell cycle
     Compared with the normal group, the hepatic cells at the G0/G1stagesincreased significantly (P<0.05) after irradiation in the12-d irradiation group.Compared with the12-d irradiation group, the hepatic cells at the G0/G1stages had no significant change (P>0.05) in the12-d irradiation plus silymarin group. However, the hepatic cells at the G0/G1stages decreasedhighly significantly (P<0.01) near normal level in the12-d irradiation plusAmerican ginseng group.
     Compared with the normal group, the hepatic cells at S stage had nosignificant change (P>0.05) after irradiation in the12-d irradiation group.Compared with the12-d irradiation group, the hepatic cells at the S stage inthe12-d irradiation plus silymarin group decreased significantly (P<0.01)while those in the12-d irradiation plus American ginseng group had nosignificant change (P>0.05).
     Compared with the normal group, the hepatic cells at the G2/M stagesdecreased highly significantly (P<0.01) in the12-d irradiation group.Compared with the12-d irradiation group, the hepatic cells at the G2/M stagesin the12-d irradiation plus silymarin group had no significant (P>0.05)change while those in the12-d irradiation plus American ginseng groupincreased highly significantly (P<0.01).
     5.2Intervention effects of American ginseng on the hepatic apoptosis rate of12-d irradiation mice
     Compared with the normal group, the apoptosis rate of the hepatic cellsincreased significantly (P<0.01) after irradiation in the12-d irradiation group.Compared with the12-d irradiation group, no significant differences (P>0.05)existed in the hepatic apoptosis rate in the12-d irradiation plus silymaringroup whereas a significant decrease (P<0.01) existed in the hepatic apoptosisrate in the12-d irradiation plus American ginseng group.
     Conclusion:
     1Long-term cell phone electromagnetic irradiation is a cause of diseases,and this cause of disease is characterized as warm heat pathogen which is oneof the six external factors causing diseases. The warm heat pathogen can leadto damage to the QI YIN. The Chinese medicine, American ginseng, has thefunction of tonifying QI and YIN for the treatment of diseases caused by QIand YIN deficiency. The American ginseng capsules have an interventioneffect on the hepatic damage of the mice with12-d irradiation.
     2The American ginseng capsule can improve the abnormal changes ofthe hepatic histomorphology and is related to its function to decrease theoxidative damage, the Nrf2expression in the hepatic cells, and the proteinexpression of ATM and H2AX, and to affect the cell cycle and cell apoptosis.
引文
1邱毅,张丽娥,陈小红,等.广西874名放射工作人员肝功能指标分析[J].环境与职业医学,2013,30(10):790-793
    2赵慧军,徐晓鸿.高压变电站电磁辐射对小鼠血液、肝功能的影响[J].湖北农业科学.2012,51(21):4827-4829
    3黄文才,沈钧康,陈胜平,等.早期肝脏放射性损伤病理组织学实验研究[J].苏州大学学报(医学版),2005,25(4):571-573
    4王放,左雅慧,王小莉,等.辐射致人正常肝细胞蛋白质组电泳图谱的改变[J].辐射防护,2009,29(6):385-340
    5田志杰,沈南,吕士杰,等.高强度微波辐射对Wistar大鼠血清SOD、HSP70和肝脏MDA、M it肿胀的影响[J].四川动物,2009,28(4):528-531
    6艾志伟,张四红,魏建华.等.手机电磁辐射对小鼠损伤实验的初步研究[J].北京生物医学工程.2005,24(3):211-213
    7曾群力.电磁辐射[J].中华预防医学杂志,2004,38(1):49
    8罗亚萍,李春香,马惠荣,等.不同时间900MHz手机频率辐射对SD雄鼠肝组织形态和功能的影响[J].河北医药,2013,35(20):3048-3049
    9马惠荣,李媛媛,栗晶晶,等.900MHz拟手机电磁辐射对大鼠肝功能和肝细胞Bax及bcl-2蛋白表达的影响[J].解放军医药杂志,2013,25(10):36-38
    10Hansson Mild K, Bach Andersen J, Fr lund Pedersen G. Is there anyexposure from a mobile phone in stand-by mode [J]. Electromagneticbiology and medicine,2012,31(1):52-56
    11Berg M B G. Are mobile phones harmful [J]. Acta Oncologica,2000,39(8):927-930
    12Wood A W. How dangerous are mobile phones, transmission masts, andelectricity pylons [J]. Archives of disease in childhood,2006,91(4):361-366
    13Esmekaya M A, Ozer C, Seyhan N.900MHz pulse-modulatedradiofrequency radiation induces oxidative stress on heart, lung, testis andliver tissues [J]. General physiology and biophysics,2011,30(1):84-89
    14Devrim E, Ergüder I B, Kili oglu B, et al. Effects of electromagneticradiation use on oxidant/antioxidant status and DNA turn-over enzymeactivities in erythrocytes and heart, kidney, liver, and ovary tissues fromrats: possible protective role of vitamin C [J]. Toxicology mechanisms andmethods,2008,18(9):679-683
    15Ozguner F, Aydin G, Mollaoglu H, et al. Prevention of mobile phoneinduced skin tissue changes by melatonin in rat: an experimental study [J].Toxicology and industrial health,2004,20(6-10):133-139
    16Weisbrot D, Lin H, Ye L, et al. Effects of mobile phone radiation onreproduction and development in Drosophila melanogaster [J]. Journal ofcellular biochemistry,2003,89(1):48-55
    17Aitken R J, Bennetts L E, Sawyer D, et al. Impact of radio frequencyelectromagnetic radiation on DNA integrity in the male germline [J].International journal of andrology,2005,28(3):171-179
    18Oral B, Guney M, Ozguner F, et al. Endometrial apoptosis induced by a900-MHz mobile phone: preventive effects of vitamins E and C [J].Advances in therapy,2006,23(6):957-973
    19Dasdag S, Akdag M Z, Ulukaya E, et al. Effect of mobile phone exposureon apoptotic glial cells and status of oxidative stress in rat brain [J].Electromagnetic biology and medicine,2009,28(4):342-354
    20Mailankot M, Kunnath A P, Jayalekshmi H, et al. Radio frequencyelectromagnetic radiation (RF-EMR) from GSM (0.9/1.8GHz) mobilephones induces oxidative stress and reduces sperm motility in rats [J].Clinics,2009,64(6):561-565
    21Atli Sekeroglu Z, Akar A, Sekeroglu V. Evaluation of the cytogenotoxicdamage in immature and mature rats exposed to900MHz radiofrequencyelectromagnetic fields [J]. International journal of radiation biology,2013,89(11):985-992
    22Jelodar G, Nazifi S, Akbari A. The prophylactic effect of vitamin C oninduced oxidative stress in rat testis following exposure to900MHz radiofrequency wave generated by a BTS antenna model [J]. Electromagneticbiology and medicine,2013,32(3):409-416
    23Ferreira A R, Knakievicz T, de Bittencourt Pasquali M A, et al. Ultra highfrequency-electromagnetic field irradiation during pregnancy leads to anincrease in erythrocytes micronuclei incidence in rat offspring[J]. Lifesciences,2006,80(1):43-50
    24Orlando V. Furtado-Filho, Juliana B. Borba, Alexsandro Dallegrave.et al.Effect of950MHz UHF electromagnetic radiation on biomarkers ofoxidative damage, metabolism of UFA and antioxidants in the livers ofyoung rats of different ages [J]. International journal of radiation biology,2014,90(2):159-168
    25高辉,裴银辉,李锋,等.胚胎期暴露手机辐射对雄性仔鼠生殖功能影响[J].中国妇幼保健.2012,27(31):4967-4968
    26高娴,罗芮,马斌,等.维生素E对手机辐射致孕鼠及胎鼠脑组织损伤的干预作用[J].卫生研究.2013,42(4):642-646
    27李乐,唐双阳,陈玉海,等.手机辐射对孕鼠及子代IFN-γ和IL-4的影响[J].微生物学免疫学进展.2011,39(3):33-35
    28孙华明,吴锡南,曹佳,等.低功率微波对大鼠精子畸形和骨髓嗜多染红细胞微核率的影响[J].第三军医大学学报,20013,25(19):1698-1770
    29马玲娟,赵晓梅,府伟灵.电磁辐射对雄性生殖系统损伤的研究进展[J].国际检验医学杂志,2012,33(8):943
    30唐国汉,黎世林.手持移动电话在信号不同强度环境下微波辐射的调查[J].中国公共卫生,2001,17(5):424-425
    31王亚民,张金明.手机使用状态的电磁辐射研究[J].环境保护,2002,(7):35-36
    32李媛媛.手机频率辐射损伤雄性大鼠“肾主生殖”功能的初步研究[D].河北医科大学硕士研究生论文.2012
    33Tsukimoto M, Tamaishi N, Homma T, et al. Low-dose gamma-rayirradiation induces translocation of Nrf2into nuclear in mousemacrophage RAW264.7cells [J]. J RadiatRes,2010,51(3):349-353
    34Mci ee DI, Soviet and Eastern European research on biological effects ofmicrowave radiation [J], Proc IEEE,1980;68(1):84
    35王小娟,陈景藻,李学荣,等.低功率毫米波辐射对小鼠肝脏作用的实验研究[J].第四军医大学学报,1990,11(2):92-95
    36Hardell L, Sage C. Biological Effects from Electromagnetic FieldExposure and Public Exposure Standards [J]. Biomed Pharmacother.2008,62(2):104-109
    37Santini M T, Rainaldi G, Indovina P L. Cellular Effects of Extremely LowFrequency (ELF) Electromagnetic Fields [J]. Int J Radiat Biol.2009,85(4):294-313
    38钟秀宏,徐俊杰,等.高功率微波辐射致大鼠心和肝损伤形态学观察[J].中国公共卫生.2010,26(12):1559-1560
    39李珍,黄晓峰,王春梅,等.高功率微波辐照后大鼠肝组织的形态学改变[J].中华放射医学与防护杂志,2005,25(2):180-182
    40安刚,周传艳,罗阳.低剂量电磁辐射对小鼠肝脏功能的影响研究[J].国际检验医学杂志.2013,34(14):1777-1778,1781
    41李妍,刘玉莲,纪朋艳,等.五味子水提物对微波辐射引起大鼠肝细胞氧化应激损伤的预防作用[J].吉林大学学报(医学版).2013,39(6):1173-1176
    42黄曾相,张惠权,潘钰卿.慢性肝炎血清酶值与肝病理变化的关系[J].浙江预防医学,2004,16(8):70
    1刘丹卓,赵新广.肝损伤病因病理机制及中医药治疗研究近况[J].国医论坛,2006,21(6):53-55
    2Poli G. Pathogenesis of liver fibrosis: role of oxidative stress [J].Molecular aspects of medicine,2000,21(3):49-98
    3Gebhardt R. Oxidative stress, plant-derived antioxidants and liver fibrosis[J]. Planta medica,2002,68(04):289-296
    4Dasdag S, Bilgin H M, Akdag M Z, et al. Effect of long term mobilephone exposure on oxidative-antioxidative processes and nitric oxide inrats [J]. Biotechnology and Biotechnological Equipment,2008,22(4):992-997
    5Esmekaya M A, Ozer C, Seyhan N.900MHz pulse-modulatedradiofrequency radiation induces oxidative stress on heart, lung, testis andliver tissues [J]. General physiology and biophysics,2011,30(1):84-89
    6李航,段惠军.Nrf2/ARE信号通路及其调控的抗氧化蛋白[J].中国药理学通报,2011,27(3):300-3
    7Rubiolo J A,Mithieux G,Vega F V. Resveratrol protects primary rathepatocytes against oxidative stress damage: Activation of the Nrf2transcription factor and augmented activities of antioxidant enzymes [J].Eur J Pharmacol,2008,591(1-3):66-72
    8邵帅,马增春,洪倩,等.基于Nrf2-ARE通路的抗辐射有效活性成分筛选研究[J].中国药理学通报,2012,28(1):29-33
    9关翠雯,金晶,李佳,等.丹参酮IIA激活Nrf2/ARE通路保护雷公藤甲素所致急性肝损伤[J].药学学报,2013,48(9):13971402
    10Cho H Y, Reddy S P, DebiaseA, et al. Gene expression profiling ofNRF2-mediated protection againstoxidative injury [J]. FreeRadic BiolMed,2005,38(3):325-343
    11戴志强,俞月桂,袁幸菊,等.氧代赖氨酸抗四氯化碳所致小鼠肝细胞损害的超微结构观察[J].中国药理学和毒理学杂志,1994.8(2):139
    12赵东红,秦德安,何学民.微波(2450Hz)对整体小白鼠红细胞膜的损伤效应及药物防护[J].华东师范大学(自然科学版),1995,(2):108-112
    13田志杰,沈南,吕士杰,等.高强度微波辐射对W istar大鼠血清SOD、HSP70和肝脏MDA、Mit肿胀的影响[J].四川动物,2009,28(7):528-530
    14李妍,刘玉莲,纪朋艳,等.五味子水提物对微波辐射引起大鼠肝细胞氧化应激损伤的预防作用[J].吉林大学学报(医学版).2013,39(6):1173-1176
    15车勇良.扇贝多肽(pcf)对H2O2损伤胸腺细胞的保护作用[J].西北农林科技人学学报:自然科学版.2005.33(4):13
    16袁平戈,张大志.还原型谷胱甘肽的作用机制及临床应用.药品评价[J],2006,3(5):385-390
    17石小枫,刘杞,郭树华.还原型谷胱甘肽对小鼠急性损伤的保护作用[J].现代医药卫生,2002.18(10):849-850
    18何梅,占美,柳汝明,等.还原型谷胱甘肽治疗药物性肝病的系统评价[J].中国药房.2010,21(32):3049-3052
    19Sies H. Oxidative stress: oxidants and antioxidants [J]. Experimentalphysiology,1997,82(2):291-295
    20Ozgur E, Güler G, Seyhan N. Mobile phone radiation-induced free radicaldamage in the liver is inhibited by the antioxidants N-acetyl cysteine andepigallocatechin-gallate [J]. International journal of radiation biology,2010,86(11):935-945
    21Dasdag S, Bilgin H M, Akdag M Z, et al. Effect of long term mobilephone exposure on oxidative-antioxidative processes and nitric oxide inrats [J]. Biotechnology and Biotechnological Equipment,2008,22(4):992-997
    22Niture S K, Khatri R, Jaiswal A K. Regulation of Nrf2—an update [J].Free Radical Biology and Medicine,2014,66:36-44
    23Moi P, Chan K, Asunis I, et al. Isolation of NF-E2-related factor2(Nrf2),aNF-E2-like basic leucine zipper transcriptional activator that binds to thetandem NF-E2/AP1repeat of the beta-globin locus control region [J].Proc Natl Acad Sci USA,1994,91(21):9926-9930
    24Zhu H, Itoh K, Yamamoto M, et al. Role of Nrf2signaling in regulationof antioxidants and phase2enzymes in cardiac fibroblasts: protectionagainst reactive oxygen and nitrogen species-induced cell injury [J].FEBS Leett,2005,579(14):3029-3036
    25Lee JM, Li J, Johnson DA, et al. Nrf2, a multi-organ protector[J]. FASEBJ,2005,19(9):1061-1066
    26Okada K,Warabi E,Sugimoto H,et al. Nrf2inhibits hepatic iron accumulation and counteracts oxidative stress-induced liver injury in nutritional steatohepatitis [J]. J Gastroenterol,2012,47(8):924-935
    27周艳利.Nrf2在溃疡性结肠炎患者中的表达及其与氧化应激的关系[D].石家庄:河北医科大学.2013
    28McDonald J T, Kim K, Norris A J, et al. Ionizing radiation activates theNrf2antioxidant response [J]. Cancer Res,2010,70(21):8886-8895
    29Nishimura J, Dewa Y, Okamura T, et al. Role of Nrf2andoxidative stresson fenofibrate-induced hepatocarcinogenesis in rats [J]. Toxicol Sci,2008,106(2):339-349
    30Ma Q. Role of Nrf2in oxidative stress and toxicity [J]. Annual RevPharmacol Toxicol,2013,53:401-426
    31Surh YJ, Kundu JK,Na HK.Nrf2as a master redox switch in turning onthe cellular signaling involved in the induction of cytoprotective genes bysame chemopreventive phytochemicals [J]. Planta Med,2008,74(13):1526-1539
    1俞英欣,万琪.ATM介导的蛋白磷酸化与DNA损伤的信号转导机制[J].国外医学分子生物学分册.2013,25(2):97-99
    2Paull TT, Rogakou EP,Yamazaki V,Kirchgessner CU,Gellert M,BonnerWM.A role for histone H2AX in recruitment of repair factors to nuclearfocicritical after DNA damage [J]. Curr Biol,2000,10(15):886-895
    3Mah L J,El-Osta A,Karagiannis T C.γH2AX:a sensitive molecular markerof DNA damage and repair [J]. Leukemia,2010,24(4):679-686
    4由莉,赵永成.γH2AX分析及其在监测电离辐射所致DNA双链断裂中的应用前景[J].国际放射医学核医学杂志.2006,30(1):50-52
    5赵晋,郭忠,马建秀.γH2AX和ATM与内源性氧化剂所致DNA损伤关系的研究进展.基础医学与临床.2010,30(4):437-441
    6余艳柯,陆源,余应年,γH2AX:DNA双链断裂的标志[J].中国药理学与毒理学杂志.2005,19(3):237-240
    7武慧欣,李宏玲,吴锡南.1800MHz电磁波辐射对大鼠外周血细胞DNA的损伤作用[J].昆明医学院学报,2009,20(8):47-49
    8司徒镇强,吴军正.细胞培养[M].北京:世界图书出版公司,2007:135-138
    9那广水,于洪儒,王洪新,等.一种小鼠原代肝细胞培养方法[J].锦州医学院学报,2003,24(3):22-23
    10张丽芳,王萍,胡晓.三种原代大鼠肝细胞分离方法的比较[J].江西医学院学报.2009,49(5):40-43
    11Zhao H, Traganos F, Albino A P, et al. Oxidative stress induces cellcycle-dependent Mre11recruitment,ATM and Chk2activation and histoneH2AX phosphorylation [J]. Cell Cycle,2008,7(10):1490-1495
    12Tanaka T, Halicka H D, Huang X,et al. Constitutive histone H2AXphosphorylation and ATM activation, the reporters of DNA damage byendogenous oxidants [J]. Cell cycle (Georgetown, Tex.),2006,5(17):1940
    13杨瑞华,陈景元,黄文华,等.高功率微波对视网膜神经节细胞脂质过氧化作用的实验研究[J].中华眼底病杂志,2000,16(1):32-34
    14龚茜芬,杨学森,李科,等.抗氧化营养素对电磁辐射致大鼠肝损伤的防护效果.第三军医大学学报.2010,32(2):119-122
    15谢健华,朱连标,陈锦生,等.铁路电力牵引工频电磁场对作业人员淋巴细胞DNA损伤的探讨[J].海峡预防医学杂志,2004,10(3):33-34
    16Adams K E, Medhurst A L, Dart D A, et al. Recruitment of ATR to sites ofionising radiation-induced DNA damage requires ATM and components ofthe MRN protein complex [J]. Oncogene,2006,25(28):3894-3904
    17Jeggo P, L brich M. Radiation-induced DNA damage responses [J].Radiation protection dosimetry,2006,122(1-4):124-127
    18Bao S, Wu Q, McLendon R E, et al. Glioma stem cells promoteradioresistance by preferential activation of the DNA damage response [J].Nature,2006,444(7120):756-760
    19姚莉.辐射损伤与细胞周期[J].国外医学·放射医学核医学分册.2004,28(1):33-36
    20Svitlana Malyarchuk,Reneau Castore, Rhua Shi. Artemis is required toimprove the accuracy of repair of double-strand breaks with5′-blockedtermini generated from non-DSB-clustered lesions [J]. Mutagenesis,2013,28(3):357-366
    21Dahm K. Functions and regulation of human artemis in double strandbreak repair [J]. J Cell Biochem.2007,100(6):1346-51
    22Guo Z, Kozlov S, Lavin M F, et al. ATM activation by oxidative stress [J].Science,2010,330(6003):517-521
    23Guo Z, Deshpande R, Paull T T. ATM activation in the presence ofoxidative stress [J]. Cell Cycle,2010,9(24):4805-4811
    24李梦侠.DNA损伤修复基因在氧化应激中的作用及其机制研究[D].第三军医大学博士学位论文.2011
    25Darroudi F, Wiegant W, Meijers M, et al. Role of Artemis in DSB repairand guarding chromosomal stability following exposure to ionizingradiation at different stages of cell cycle [J]. Mutat Res.2007,615(1-2):111-24
    26Zhang X, Succi J, Feng Z, et al. Artemis is a phosphorylation target ofATM and ATR and is involved in the G2/M DNA damage checkpointresponse [J]. Molecular and cellular biology,2004,24(20):9207-9220
    27Wang J, Pluth J M, Cooper P K, et al. Artemis deficiency confers a DNAdouble-strand break repair defect and Artemis phosphorylation status isaltered by DNA damage and cell cycle progression [J]. DNA repair,2005,4(5):556-570
    28Rogakou EP, Pilch DR, Orr AH, et al. DNA double-stranded breaks inducehistone H2AX phosphorylation on serine139[J].J Biol Chem,1998,273(10):5858-5868
    29Wang H, Wang M, Wang H, et al. Complex H2AX phosphorylationpatternsbymultiple kinases including ATM and DNA-PK in human cellsexposed to ionizing radiation and treated with kinase inhibitors [J]. J CellPhysiol,2005,202(2):492-502
    30Dickey J S, Redon C E, Nakamura A J, et al. H2AX: functional roles andpotential applications [J]. Chromosoma,2009,118(6):683-692
    31Redon C, Pilch D, Rogakou E, Sedelnikova O, Newrock K, BonnerW.Histone H2A variants H2AX and H2AZ [J]. Curr Opin Genet Dev,2002,12(2):162-169
    32由莉,赵永成.γH2AX分析及其在监测电离辐射所致DNA双链断裂中的应用前景[J].国际放射医学核医学杂志.2006,30(1):50-52
    33Mah L J, Orlowski C, Ververis K, et al. Evaluation of the efficacy ofradiation-modifying compounds using γH2AX as a molecular marker ofDNA double-strand breaks [J]. Genome Integr,2011,2(1):3
    34Paull TT, Rogakou EP, Yamazaki V, et al. A critical role for Histone H2AXin recruitment of repair factors to nuclear foci after DNA damage [J]. CurrBiol,2000,10(15):886-895
    35MacPhail SH, Banath JP, Yu TY, Chu EH, Lambur H, Olive PL.Expressionof phosphorylated histone H2AX in cultured cell lines following exposureto X-rays [J]. Int J Radiat Biol,2003,79(5):351-358
    36Bartova E, Krejci J, Harnicarova A, et al. Histone modifications and ucleararchitecture: a review [J]. J Histo chem Cytochem,2008,56(8):711-721
    37Ismail IH, Wadhra TI, Hammarsten O. An optimized method for detectingγ-H2AX in blood cells reveals a significant interindividual variation in theγ-H2AX response among humans [J]. NucleicAcids Res,2007,35(5): e36
    38Tanaka T, Kurose A, Huang X, et al. ATM activation and histone H2AXphosphorylation as indicators of DNA damage by DNA topoisomerase Iinhibitor topotecan and during apoptosis [J]. Cell proliferation,2006,39(1):49-60
    39宋建元,曹建平,朱巍,等.外源性毛细血管扩张性共济失调突变基因对辐射诱导的细胞周期的影响[J].苏州大学学报(医学版).2008,28(1):1-3
    40于廷曦,朱应葆,童坦君.DNA损伤与细胞周期调控[J].生物化学与生物物理进展,1999,26:350-354
    41鞠桂芝,苏旭,范冰,等.低剂量X射线照射诱导G1期阻滞的适应性反应[J].中华放射医学与防护杂志,1998,18(3):148-150
    42刘丽,金宏,王先远,等.大豆异黄酮对电离辐射小鼠脾脏细胞周期、凋亡与增殖的影响[J].中国应用生理学杂志.2006,22(4):497-500
    43Darroudi F, Wiegant W, Meijers M, et al. Role of Artemis in DSB repairand guarding chromosomal stability following exposure to ionizingradiation at different stages of cell cycle [J]. Mutat Res.2007,615(1-2):111-24
    44Mohapatra S, Kawahara M, Khan IS,et al. Restoration of G1chemo/radioresistance and double-strand-break repair proficiency bywild-type but not endonuclease-deficient Artemis [J]. Nucleic Acids Res.2011;39(15):6500-10
    45刘赟,洪蓉,喻云梅,等.极低频电磁场对小鼠脑和肝细胞凋亡及细胞周期的影响[J].中华劳动卫生职业病杂志,2003,21(5):339-341
    46姚克,王凯军,谭健,等.低强度2450MHz微波阻断兔晶状体上皮细胞周期及对相关基因表达的影响[J].中华眼科杂志,2004,40(12):836-840
    47张桂珍,高申,郑德明,等.过量氟对大鼠脑肝肾细胞周期与细胞凋亡的影响[J].中国地方病学杂志,2001,20(5):344-346
    48刘赟,洪蓉,喻云梅,等.极低频电磁场对小鼠脑和肝细胞凋亡及细胞周期的影响[J].中华劳动卫生职业病杂志,2003,21(5):339-341
    49胡恭华,庄志雄,夏芳莲,等.氢醌对L-02肝细胞DNA损伤及细胞周期的影响[J].环境与健康杂志,2007,24(4):201-203
    1罗琦.自拟养阴生津方对小鼠辐射损伤的防护作用研究[D].衡阳:南华大学,2011
    2沈世林,张汝学,贾正平具有抗辐射损伤作用的中药复方研究进展概述[J].中国实验方剂学杂志.2007.13(11):64-66
    3刘霞,杨景云,梁振明,等.八珍制剂对60Co辐射小鼠微生态失调的促恢复作用[J].中国微生态学杂志,2004,16(5):263-267
    4苏颖,郭军鹏,李欣.等.益气中药复方对电离辐射小鼠T淋巴细胞亚群的影响[J].时珍国医国药.2012.23(6):1430-1432
    5龚茜芬,杨学森,涂灵,等.复方中药制剂对电磁辐射致大鼠海马神经损伤的防治作用[J].中国应用生理学杂志.2013.29(4):346-350
    6刘哲峰,赵宏.生脉注射液提高机体抗辐射能力临床研究[J].科学技术与工程,2005,14(5):980
    7邓伟国,付艳,赵桂兰,等.人参皂苷对小鼠脂质及细胞膜辐射防护作用[J].白求恩医科大学学报,1998,24(1):29-30
    8汪德清,沈文梅,田亚平,等.黄芪总黄酮对犬模拟肺癌术中放疗抗辐射损伤作用的研究[J].中华放射医学与防护杂志,1996,16(6):339-401
    9杨映雪,陈建业,费中海,等.黄芪总黄酮对辐射损伤小鼠的防护作用研究[J].重庆医科大学学报,2010,35(4):504-507
    10郭毅,曹新山,李宗山,等.黄芪抗γ射线辐射对小鼠淋巴细胞增殖和IL-2的作用研究[J].时珍国医国药,2004,15(9):554-555
    11杨永建,梁莉,李梅,等.南沙参多糖对亚慢性辐射损伤的防护作用[J].中药药理与临床,2001,18(5):21-23
    12葛明珠,张勇,任少琳.南沙参多糖的辐射防护作用[J].中华放射医学与防护杂志,1996,16(2):123-125
    13于永超,张佳丽,林兵,等.西洋参多糖对60Co辐照小鼠免疫功能的调节作用[J].现代预防医学,2012,39(11):2685-2687
    14赵玉珍,刘蕾,陈立平,等.西洋参茎叶皂甙对大鼠实验性肝损伤的影响[J].中成药,2000,22(3):219-220
    15包海花,袁晓环,韩雪山,等.西洋参皂苷对H2O2致PC12细胞损伤的保护作用[J].中国生化药物杂志,2009,30(5):302-305
    16马骏,毛平,陈艳艳等.补气中药对正常及辐射小鼠脾脏巨噬细胞的影响[J].上海中医药大学学报,2006,20(4):76-78
    17邢艺凡.西洋参通过调节Nrf2抑制主动脉弓缩窄术诱导的小鼠心室重构的研究[D].济南:山东大学,2012
    18赵玉珍,栗坤,马春丽,等.国产西洋参茎叶皂苷减轻四氯化碳致大鼠肝损伤[J].佳木斯医学院学报,1997,20(2):11-12
    19侯新然,王晓波,李忠亮,等.耐力胶囊对60Coγ射线损伤小鼠的保护作用[J].2012,18(23):225-229
    20侯军峰,张盈华,史恒军,等养阴抗毒散对受照小鼠红细胞免疫粘附功能的影响4[J].第四军医大学学报,2000,21(4):496-498
    21侯军峰,焦凯,张盈华.中药复方对X线照射小鼠T淋巴细胞亚群和抗氧化能力的作用[J].中国免疫学杂志,2003,19(1):54-56
    22张盈华,殷缨,侯军峰,等.养阴抗毒散对X射线辐射损伤小鼠巨噬细胞免疫活性的防护作用[J].中华放射医学与防护杂志,2003,23(1):41-42
    23刘宗林.西洋参茎叶水溶性多糖结构分析及其生理活性的研究[J].食品工业科技,2001,22(2):20-21
    24吴东方,彭仁秀,叶丽萍.水飞蓟素对小鼠肝细胞微粒体及线粒体膜流动性的影响[J].中国中药杂志,2003,9:870-872
    25吴波明,应茵.水飞蓟宾胶囊对非酒精性脂肪性肝病患者肝功能及血脂的影响[J].中国药业,2013,22(7):26-27
    26甄月映.水飞蓟宾胶囊治疗抗艾滋病毒药物致肝损害的效果观察[J].中国当代医药,2013,20(15):63-66
    27张国富,王娜,陈建生,等.水飞蓟宾胶囊治疗抗精神病药物所致肝损伤多中心随机对照研究[J].中国肝脏病杂志(电子版),2013,5(1):31-36
    2828宋维芳,许瑞龄,王登妮,等.水飞蓟宾对大鼠实验性肝纤维化发生发展作用机制的研究[J].山西医科大学学报,2009,40(5):423-426.480
    29薛洪源,侯艳宁,刘会臣,等.水飞蓟宾胶囊对异烟肼和利福平肝损害小鼠的保护作用[J].中成药,2003,25(4):317-310
    30肖时湘,韩涛,李岩.水飞蓟宾胶囊治疗慢性乙型病毒性肝炎合并脂肪肝的疗效[J].中国新药杂志,2009,18(16):1530-1532
    31李荣萍,任成山,赵晓宴.水飞蓟宾对急性肝损伤中肝细胞胀亡的影响及其机制[J].中国中西医结合急救杂志,2006,13(4):201-205
    32陈正跃,吴子钊,许建文,等.水飞蓟宾-磷脂酰胆碱复合物与水飞蓟宾对四氯化碳所致小鼠急性肝损伤保护作用的对比[J].中国生化药物杂志,2004,25(4):221-223
    33Rajnarayanak, Reddyms, Vidyasagar J, etal.Studyon the influence ofsilymarin pretreatment on metabolism and dis-position of metronidazole[J]. Arznelmittelforschung,2004,54(2):109-113
    34Sailer R, Meier R, Brignolir. The use ofsilymarin in thetreatment of liverdiseases [J]. Drugs,2001,61(14):2035-2063.-4
    35吴华彰,赵云利,费鸿君等.西洋参皂甙的抗氧化功能及其对小鼠遗传损伤的保护作用[J].中国生物制品学杂志,2012,25(1):61-64
    36刘敏,肖颖,左爱仁,等.槲皮素、根皮素、水飞蓟宾清除自由基和抑制脂质过氧化活性研究[J].中成药,2012,34(4):753-756
    37汪艳丽,余燕影,田洁等.水飞蓟宾乳糖修饰对急性酒精性肝损伤保护作用的影响[J],时珍国医国药,2013,24(10):2360-2363
    38黄治官,马仁强.中药方剂1号对小鼠免疫功能和抗氧化能力的影响[J].中药材,2013,36(8):1304-1308
    39Shelton L M, Park B K, Copple I M.Role of Nrf2in protection against acute kidney injury [J]. Kidney international,2013,84(6):1090-109
    40Muthumani M, Prabu S M. Silibinin potentially attenuates arsenic-induced oxidative stress mediated cardiotoxicity and dyslipidemia in rats [J].Cardiovascular toxicology,2013:1-15
    41任连生,张蕻,汤莹山.全甲洋参胶囊对环磷酰胺致畸和抑瘤作用的影响[J].中国肿瘤临床与康复,2003,7(4):4
    42陈勤,张道宏,史竹兵.丝裂霉素致小鼠DNA损伤及西洋参的干预作用研究.中医药理论与应用研究-安徽中医药继承与创新博士科技论坛论文集.中国安徽合肥,2008,195-197
    43朴云峰,明月,李靖涛.西洋参多糖Ⅰ、Ⅱ、Ⅲ对肝癌细胞DNA合成抑制作用的研究[J].临床肝胆病杂志,1999,15(4):213-214
    44Sonnenbichler J, Goldbero M, Hane L, et al. Stimulatory effect ofSilibinin on the DNA synthesis in partially hepatectomized rat livers:non-response in hepatoma and other malign cell lines [J]. Biochemicalpharmacology,1986,35(3):538-541
    45Hoh C, Boocock D, Marczylo T, et al. Pilot study of oral silibinin, aputative chemopreventive agent, in colorectal cancer patients: silibininlevels in plasma,colorectum,and liver and their pharmacodynamicconsequences[J]. Clinical cancer research,2006,12(9):2944-2950
    46史艳宇,西洋参有效部位诱导肿瘤细胞凋亡及其分子机制研究[D].吉林:吉林大学,2006
    47史艳宇,李红,杨世杰。西洋参有效部位对K562细胞凋亡诱导的实验研究[J].中国药理学通报.2005,21(12):1494~1497
    48王智昊,赵学忠.西洋参叶二醇组皂苷抑制血管平滑肌细胞增殖及影响原癌基因表达的研究[J].长春大学学报,2008,18(4):75-79
    49任孟军,左国庆.水飞蓟宾对人肝癌细胞SMMC-7721体外增殖的影响,现代肿瘤医学现代肿瘤医学[J].2007,25(9):1229-1231
    1韩硬海,李树桐.临床肝脏病学[M].济南:山东科学技术出版社,2004:1-10,211-371
    2Cohen JI, N agy LE. Pathogenesis of alcoholic liverdisease:in-teractions between parenchymal and non-parenchymal cells[J].J Dig Dis,2011,12(1):3-9
    3Khan AQ, Nafees SSultana S. Perillyl alcohol protects against ethanolinduced acute liver injury in Wistar rats by inhibiting oxidative stress,NFK-B activation and proinflammatory cyto-kineroduction[J]. Toxicology,2011,279(1-3):108-114
    4于晨辉,杜仲燕,高佳,等.4-HNE通过抑制TNF-α介导的NF-κB活化诱导酒精性肝损伤[J].中国病理生理杂志,2013,29(6):1046-1052
    5吴晓宁,尤红,贾继东.2003-2007年国内药物性肝损伤临床特点文献综合分析[J].肝脏,2008,13(6):463-466
    6黄道林,向娟,刘晓东,等.药源性肝损伤中药的研究进展[J].2012,24(10):13-15
    7徐鑫.药物性肝损伤机制[J].医学综述,2008,14(5):747-749
    8Date M, Matsuzaki K, MatsushitaM, et al. Differential regulation ofac-tivin Afor hepatocyte growth and fibronectin synthesis in rat liverinjury [J]. J Hepatol,2000,32(2):251-260
    9马小超,屠曾宏.细胞色素P450在药源性肝损伤中的作用[J].世界华人消化杂志,2003,11(3):338-341
    10张石革.药物性肝损伤的机制和药物选择[J].中国全科医学,2009,18(9):1659-1661
    11Lei X, Chen Y, Du G,et al.Gossypol induces Bax/Bak-independentactivation of apoptosis and cytochrome c release via a conformationalchange in Bcl-2[J]. FASEB J,2006,20(12):2147-2149
    12韩瑞刚,彭瑞云.电离辐射对骨髓损伤的研究进展[J].中华放射医学与防护杂志,2005,25(6):586-588
    13Nakazawa Y, Chisuwa H, Mita A, et al. Life-threatening veno-occlusivedisease after living-related liver transplantation [J]. Transplantation,2003,75:727-730
    14王舟,曾令福,肖元梅,等,绿茶抗辐射损伤作用研究[J].四川大学学报(医学版),2003,34(2):303-305
    15贾海泉,金宏,许志勤,等.大豆异黄酮对辐射小鼠造血系统损伤防护作用的实验研究[J].氨基酸和生物资源,2005,27(2):23-25
    16姚莉,梁晓俐,王德文,等.高场强电磁脉冲辐射对小鼠肝细胞DNA含量和倍体影响的定量学研究[J].2004,24,(4):386-388
    17潘晓燕,何继亮,张美辨,等.低强度微波与4-硝基喹啉氧化物对人淋巴细胞DNA损伤的研究[J].中华放射医学与防护杂,2003,23(4):301-303
    18杨瑞福.DMN致小鼠肝损伤模型的建立及用于中药护肝效果评价[D].广州中医药大学,2008
    19张琪,陈辉,彭顺利,等.急性肝损伤动物模型制备研究进展[J],2011,32(4):216-220
    20Masuda Y. Learning toxicology from carbon tetrachloride-in-ducedhepatotoxicity[J]. Yakugaku Zasshi,2006,126(10):885-899
    21刘莲,孙晖,王喜军,等.茵陈蒿汤对乙醇诱导的大鼠急性肝损伤的保护作用研究[J].世界科学技术-中医药现代化基础研究,2007,9(4):35-40
    22于强,于洋.肝损伤动物模型及中药治疗研究概况[J].中国现代中药,2006,8(11):25-28
    23邱英锋,缪晓辉,蔡雄,等.卡介苗加脂多糖建立的大鼠急性免疫性肝损伤模型的研究[J].西北国防医学杂志,2004,25(5):345-347
    2424黄正明,杨新波,等.化学性及免疫性肝损伤模型的方法研究[J].解放军药学学报,2005,25(1):42-46
    25马玲娟,50Hz磁场暴露后大鼠生物学变化的临床经验分析[D],第三军医大学,2012
    26马惠荣,李媛媛,等.观察900MHz拟手机电磁辐射对大鼠肝功能和肝细胞Bax及Bcl-2表达的影响[J].解放军医药杂志,2013,25(10):36-44
    27艾志伟,张四红,魏建华.手机电磁辐射对小鼠损伤实验的初步研究[J].北京生物医学工程,2005,24(3):211-213
    28陈驰.[18F]-FDG-PET/CT可对骨髓、肝脏、小肠的放射后损伤进行快速分层诊断并评估预后[D].南方医科大学,2012
    29李艳辉,肖恩华.实验性肝损伤模型的建立和评价[J].放射学实践,2006,21(10):1075-1077
    30冀润利,赵青娥.水飞蓟素对四氯化碳致小鼠肝损伤的保护作用[J].中国应用生理学杂志,2012,28(3):279-281
    31姚会枝,李吉学,郑海娜.冬凌草提取物对四氯化碳诱导的小鼠慢性肝损伤的保护作用[J].时珍国医国药,2010,21(3):575-577
    32俞亚静,方晶.车前草水提取物对肝脏保护作用的实验研究[J].中国实用医药,2008,3(16):71-72
    33田红剑,陈斌.当归对大鼠免疫性肝损伤的保护作用[J].中南药学,2003,1(5):271
    34孙长海,王瑜等.中药莪术对四氯化碳所致急性肝损伤的保护作用[J].时珍国医国药,2010,21(10):2460-2461
    35刘刚,雷立,等.赤灵芝及其孢子粉提取物缓解小鼠肝脏CCl4损伤的研究[J].西南师范大学学报(自然科学版),2009,34(5):117
    36赵红宇,李文斌.灵芝抱子粉抗肝纤维化作用的实验研究[J].中国实用医药,2007,2(5):12
    37王明宇,刘强,车庆明,等.灵芝三萜类化合物对3种小鼠肝损伤模型的影响[J].药学学报,2000,35(5):326
    38杨丽丽,邓志钦,等.灵芝孢子粉对抗亚急性酒精性肝损伤的实验研究[J].时珍国医国药,2013,24(3):513-514
    39李天一,汪丽佩,吴国琳.黄芪多糖对免疫性肝损伤小鼠免疫调节的影响[J].中国中医急症,2014,23(1):25-27
    40赵军,刘涛,马龙,苏德奇.类叶升麻苷对大鼠原代肝细胞免疫性肝损伤的保护作用[J].西北药学杂志,2013,28(3):269-271
    41陈奕冰.葛花多糖对辐射损伤小鼠保护作用的研究[D].吉林大学,2013
    42赵杰,孙设宗,等.半枝莲多糖对四氯化碳致小鼠肝损伤保护作用的研究[J].中国中医药科技,2012,19(1):39-40
    43陈宝芝,等.北五味子粗多糖对四氯化碳致小鼠急性肝损伤的保护作用及其机制[J].吉林大学学报(医学版),2014,40(1):92-94.
    44廖磊,孙丽萍,刘魁英,等,花粉提取物对D-半乳糖胺盐酸盐致大鼠肝损伤的保护作用[J].解放军药学学报,2013,29(6):500-502
    45宋昊,赵文玺,王玉娇,等.祁州漏芦对四氯化碳所致肝脏氧化应激和DNA损伤的影响[J].中国药学杂志,2013,48(22):1915-1918
    4646李国栋,陈园园,王盼,等.野菊花中萜类和黄酮类化合物保肝作用研究[J].中草药,2013,44(24):3510-3514
    47齐艳萍.鹿茸对小鼠急性肝损伤的修复作用及相关机理研究[D].东北林业大学,2008
    48李妍,刘玉莲,纪朋艳,等.五味子水提物对微波辐射引起大鼠肝细胞氧化应激损伤的预防作用[J].吉林大学学报(医学版),2013,39(6):1173-1176
    49夏珂,张宁,王业秋,等.昼夜给予逍遥散对肝损伤大鼠的保护作用[J].中国实验方剂学杂志,2010,16(7):168-170
    50柴智,樊慧杰,王永辉,等.逍遥散对致大鼠急性肝损伤的防治作用研究[J].世界中西医结合杂志,2014,9(1):27-29
    51谢斌,俞大军,程绍民.加味小柴胡汤对醋氨酚所致肝损伤的保护作用[J].时珍国医国药,2008,19(l):130-131
    52陈粱.柴胡疏肝散对四氯化碳所致大鼠急性肝损伤的防治作用[J].中西医结合肝病杂志,2004,14(1):42
    53闫冰,蔡秀江,姚卫峰,等.二至丸保肝活性成分群对CCl4致小鼠急性肝损伤保护作用研究[J].中国中药杂志,2012,37(9):1303-1306
    54张莉,蔚红华.扶正化瘀中药治疗肝纤维化大鼠的实验研究[J].现代中西医合杂志,2011,20(1):24-25
    55刘成海,宜红萍,陶艳艳.基于FN-整合素信号途径探讨扶正化淤方抑制肝状细胞活化的作用机制[J].上海中医药杂志,2008,42(1):3-7
    56Jain AK, Jaiswal AK. Phosphorylation of tyrosine568controls nuclearexport of Nf2[J], J Bio Che,2006,281(17):12132-12142
    57吴硕.扶正化瘀方对肝纤维化小鼠肝细胞中Nrf2表达的影响[D].山东大学,2013
    58陈敏,严璐佳,等.六味地黄丸对非酒精性脂肪肝大鼠肝脏的保护作用[J].福建中医药大学学报,2013,23(1):21-22
    59陈敏.六味地黄丸对非酒精性脂肪肝大鼠肝脏的保护作用及其机制研究[D].福建中医药大学,2013
    60黄剑涛,唐琪,谢涛,等.六味地黄丸联合山楂防治大鼠非酒精性脂肪肝的实验研究[J].湖南中医杂志,2013,29(6):108-110
    61龚继勇,齐宝凤,刘利胜,等.加味当归补血汤对荷瘤小鼠X线辐射急性肝损伤血清转氨酶活性的影响[J].中国医药导报,2008,5(26):151-152
    1Hardell L. and C. Sage. Biological effects form eletromagnetic field andpublic expoure standards [J]. Biomed Pharmacother,2008,62(2):104-109
    2Sandstrom M.Electromanetic fields in offices [J]. Int J Occup Saf Ergon,2006.12(2):137-147
    3Kruglik, O.V. Morgulis, II, and R.G. Khlebopros.Effoct of microwaveelectromagnetic radiation on tumer cell viability in experiment [J]. DoklBiochem Biophys,2013,44(9):66-68
    4Khurana,V.G. et al. Cell phones and brain tumers:a review including thelong-term epidemiologic date [J]. Surg Neurol,2009,72(3):205-214
    5Behari, J. Biological responses of mobile phone frenquency exposure [J].Indian J Exp Biol,2010,48(10):959-981
    6Kundi M,Mild K. Mobile telephones and cancer-a review ofepidemiological evidence [J]. Toxicol Environ Health B Crit Rev,2004,7(5):351-384
    7Kundi M, Mild K, Hardell L. Mobile telephones and cancer-a review ofepidemiological evidence [J]. Toxicol Environ Health B Crit Rev,2004,7(5):351-384
    8Franke H, Ringelstein EB.Electromagnetic fields(GSM1800)do not alterblood-brain barrier permeability to sucrose in models in vitro with highbarrier tightness [J]. Bioelectromagnetics,2005,26(7):529-535
    9路国宾,李玉红,史长华等.2000μW/cm2电磁辐射对大鼠学习记忆功能的影响[J].现代预防医学,2013,40(10):1906-1909
    10刘勇,黄蔚初,程绪浩等.电磁辐射对家兔谷氨酸受体2蛋白质表达和磷酸化的影响[J].中国预防医学杂志,2008,10(9):879-882
    11Hutter HP, Moshammer H, Wallner P, et al.Subjective Symptoms,SleepingProblems,and Cognitive Performance in Subjects Living near MobilePhone Base Stations Occup [J]. Environ. Med,2006,63(5):307-313
    12Moshammer H, Hutter HP, Wallner P, et al. Subjective Symptoms,SleepingProblems,and Cognitive Performance in Subjects Living near MobilePhone Base Stations Occup [J]. Environ.Med,2006,63(5):307-313
    13De Pomerai D, Daniells C, David H,et al. Non-thermal heat-shockresponse to microwaves [J]. Nature,2000,405(6785):417-418
    14Franke H,Ringelstein EB,St gbauer F. Electromagnetic fields (GSM1800) do not alter blood-brain barrier permeability to sucrose in models invitro with high barrier tightness [J]. Bioelectromagnetics,2005,26(7):529-535
    15Hardell L, Hallqu ist A, Mild KH, et al. Cellular and cordless telephonesand the risk for brain tumours [J]. Eur J Can cer Prev,2002,11(4):377-386
    16Lonn S, Ah lbom A. Mobile phone use and the risk of acoust ic neu rom a[J]. Ep idem iology,2004,15:653-659
    17潘敏鸿,彭瑞云,高亚兵.高功率微波辐射后大鼠心脏β1肾上腺素能受体和M2胆碱能受体表达的变化[J].中华劳动卫生职业病杂志,2005,23(3):172-174
    18李振杰,乐秀鸿,杨军,等.长波脉冲电磁场对职业人群健康影响的三年动态观察[J].第六届全国生物电磁和第二届微波生物医学学术会议论文摘要集.1997,7(7):233-240
    19裴兆辉,丁红新,陈景藻.次声对大鼠心率与血压影响的实验研究[J].军事医学科学院院刊.2005,3(4):32-34
    20丁朝阳,吕志忠,马强等.雷达作业人员健康状况的调查和评价.铁道劳动安全与环保.1996,23(4):309
    21Braune S, Wrocklage C, Raczek J,et al.Resting blood pressure increaseduring exposure to a radio-frequency electromagnetic field [J].Lancet,1998,351:1857-1858
    22邓桦,王德文,彭瑞云.高功率脉冲微波和电磁脉冲辐照对心肌细胞膜电穿孔效应及机理的研究.生物医学工程学杂志,2005,22(4):672-676
    23姚莉,梁晓俐,王德文.高场强电磁脉冲辐射对小鼠肝损伤效应的初步研究[J].中华理医学与康复杂志,2004,26(2):69-72
    24赵慧军,徐晓鸿.高压变电站电磁辐射对小鼠血液、肝功能的影响[J].湖北农业科学2012,51(21):4827-4829
    25安钢,周传艳,罗阳.低剂量电磁辐射对小鼠辐射肝脏功能的影响研究[J].国际检验医学杂志,2013:1673-4230
    26Meral I, Mert H, Mert N, et al. Effects of900-MHz electromagnetic fieldemitted from cellular phone on brain oxidative stress and some vitaminlevels of guinea pigs[J]. Brain research,2007,1169:120-124
    27Koyu A, Yilmaz H, Uz E, et al. The protective effect0f caffeic acidphenethyl ester (CAPE) on oxidative stress in rat liver exposed to the900MHz electromagnetic field [J]. Toxicology and industrial health,2009,25(6):429-34
    28田志杰,沈南.高强微波辐射对Wistear大鼠血清SOD、HSP和肝脏MDA、Mit肿胀的影响[J].四川动物.2009.28(4):528-531
    29Ivan ica Tro i, Ivan Pavi i. Impact of Radiofrequency Radiation onRat’s DNA, Coll. Antropol.2011(4):1259–1264
    30Robison JG, Pendleton AR, Monson KO. Decreased DNA repair rates andprotection from heat induced apoptosis mediat ed by electromagnetic fieldex posure. Bio electronics,2002.23(2):106-112
    31陈烈平,黄渊清,陈起燕,等.低出生体重儿影响因素病例对照研究.中国公共卫生,2008,24(10):1258-1259
    32曹亚男.极低频电磁场对小鼠雌性生殖和子代生长发育的影响.中华劳动卫生职业病杂志,2006,24(8):468-470
    33杨娟,张元珍,刘文惠.935MHz微波电磁辐射对卵母细胞成熟的影响.武汉大学学报,医学版,2008,29(4):519-523
    34任东青,杨文清,曾桂英.微波辐照对雄性小鼠生殖系统的影响[J].中国临床康复,2002,6(6):822-823
    35OzgunerM, Koyu A,CesurG.Biological and morphological effects on thereproductive organ of rats after exposure to elec-tromagnetic field [J].SaudiMed J,2005,26(3):405-410
    36Djeridane Y, Touitou Y, de Seze R.Influence of electromagnetic fieldsemitted by GSM-900cellular telephones on the circadiam patterns ofgonadal,anrenal and pituitary hormones in men [J]. Radiat Res,2008,169(3):337-343
    37Ribeiro EP,Rhoden EL,Horn MM,et al.Effects of subchronic exposureto radio frequency from a conventional cellular telephone on testicularfunction in adultrats [J]. JUrol,2007,177(1):395-399
    38张巍,康锶鹏,陈清瑞.巴戟天对微波损伤的雄鼠睾丸生精功能的影响[J].解剖学研究,2010,32(5):338-340
    39Erogul O, Oztas E,Yildrim I. Effects of eletromagneticradiation form acellular phone on human sperm motility:an in vitro study [J]. Arch MedRes,2006,37(7):840-843
    40胡海翔,方红,罗少波.微波辐射对雷达操作人员精子头部及尾部超微结构的影响[J].中国男科学杂志,2010,24(9):33-35
    41韦春玲,胡竹林.低强度微波对晶状体的非热效应损伤的研究进展[J].医学综述,2009,15(18):2769-2770
    42陈鹏,梁洁,肖瑞.微波长时间辐照对视网膜结构和功能的影响[J].中国职业医学,2011,38(1):13-16

© 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