用户名: 密码: 验证码:
基于JNK、p38/Smads信号级联研究左归丸含药血清对MC3T3-E1细胞的调控机制
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:基于“肾主骨”理论,探讨补肾滋阴代表方剂左归丸促进骨形成作用,及JNK、p38/Smads信号级联对其影响,以探讨左归丸潜在促进骨形成的作用机制。
     材料与方法:采用血清药理学方法,将SPF级SD雌性大鼠60只(180~220g)按体质量随机分为空白对照组、左归丸组和倍美力组,每组各20只,依据人与大鼠等剂量换算公式折算大鼠左归丸等临床剂量为1.6g·Kg-1,大鼠倍美力结合雌激素片的等临床剂量为56.25μg·kg-1,使用前用蒸馏水将药物制成混悬液,空白对照组给予蒸馏水,灌服10ml·Kg-1体质量,2次·d-1,连续灌胃7d,末次给药2h后,麻醉大鼠,腹主动脉取血,室温静置2h,2500r·min-1,4℃离心20min,收集上清,同组血清混匀,56℃水浴灭活30min,制备大鼠含药血清。采用MC3T3-E1成骨细胞系体外研究模型,用含10%FBS的α-MEM培养液(100U·ml-1青霉素和100μg·ml-1链霉素)培养。细胞实验分为空白对照组(control)、SP600125组(SP)、SB203580组(SB)、左归丸组(ZG)、左归丸+SP600125组(ZG+SP)、左归丸+SB203580组(ZG+SB)、倍美力组(BML)、倍美力+SP600125组(BML+SP)、倍美力+SB203580组(BML+SB)。细胞接种24h后,换用含0.2%FBS的α-MEM培养液(100U·ml-1青霉素和100μg·ml-1链霉素),饥饿24h,吸弃旧培养液,加入含或不含JNK、p38MAPK特异抑制剂SP600125、SB203580(10μmol·L-1,溶于DMSO,体积分数<0.1%)的α-MEM培养液(100U·ml-1青霉素和100μg·ml-1链霉素),预处理60min后,各组加入相应血清(阴性对照孔、control组、SP组、SB组加入空白对照大鼠血清,ZG组、ZG+SP组、ZG+SB组加入左归丸大鼠含药血清,BML组、BML+SP组、BML+SB组加入倍美力大鼠含药血清),体积分数为15%,孵育48h。采用蛋白质印迹Western blot测定JNK和p38MAPK磷酸化蛋白表达水平,采用免疫荧光染色法进一步验证JNK和p38MAPK磷酸化蛋白表达水平。采用MTT法检测MC3T3-E1细胞增殖;采用流式细胞术分析细胞周期。采用对硝基苯磷酸盐(PNPP)法和改良钙钴染色法检测MC3T3-E1细胞碱性磷酸酶(ALP)活性;采用茜素红染色法检测MC3T3-E1细胞钙化结节形成。采用Western blot分析OB分泌蛋白情况,检测核结合因子(Runx2)、ALP、Ⅰ型胶原(ColI)、Smad2/3、 p-Smad2和p-Smad3蛋白表达水平。采用Q-PCR技术测定相关基因mRNA表达水平,检测MC3T3-E1细胞转化生长因子β1(TGF-β1)、Smad4、Runx2和ColI mRNA表达水平。
     结果:
     1.JNK信号通路参与左归丸含药血清诱导MC3T3-E1细胞分化的调控
     1.1左归丸含药血清能够诱导MC3T3-E1细胞JNK信号通路活化
     结果显示,与control组比较,ZG和BML均能显著诱导MC3T3-E1细胞JNK蛋白磷酸化;ZG显著优于BML;加入SP600125后,显著抑制ZG和BML对JNK蛋白磷酸化的诱导作用。
     1.2JNK信号通路主要在分化晚期参与左归丸含药血清对MC3T3-E1细胞ALP活性的调控
     结果显示,与control组比较,ZG和BML均能显著诱导MC3T3-E1细胞48h和7d的ALP活性(P<0.01);对于48h的ALP活性的影响,ZG组显著优于BML组(P<0.01);加入SP600125后,显著抑制BML对48h和7d的ALP活性的刺激作用(P<0.01),对ZG诱导48h的ALP活性的影响没有统计学意义(P>0.05),但显著抑制了ZG诱导7d的ALP活性。
     1.3JNK信号通路参与左归丸含药血清促进MC3T3-E1细胞矿化作用的调控
     结果显示,与control组比较,ZG和BML均能显著诱导MC3T3-E1细胞14d的矿化作用;加入SP600125后,显著抑制ZG和BML对矿化作用的诱导。
     1.4JNK信号通路对左归丸含药血清干预Runx2和ALP蛋白表达的影响
     结果显示,与control组比较,ZG和BML均能显著上调MC3T3-E1细胞48h的Runx2和ALP蛋白的表达;加入SP600125后,未能对抗ZG诱导48h的Runx2和ALP蛋白表达,也未能对抗BML诱导的Runx2蛋白的表达,但显著抑制了BML诱导的ALP蛋白表达。
     2.p38MAPK信号通路参与左归丸含药血清诱导MC3T3-E1细胞分化的调控
     2.1左归丸含药血清能够诱导MC3T3-E1细胞p38MAPK信号通路活化
     结果显示,与control组比较,ZG和BML均能显著上调MC3T3-E1细胞p38磷酸化蛋白的表达;加入SB203580后,显著抑制ZG和BML对p38蛋白磷酸化的诱导作用。
     2.2p38MAPK信号通路参与左归丸含药血清对MC3T3-E1细胞分化不同阶段ALP活性的调控
     结果显示,与control组比较,ZG和BML均能显著诱导MC3T3-E1细胞48h和7d的ALP活性(P<0.01);加入SB203580后,显著抑制ZG和BML对48h和7d的ALP活性的刺激作用(P<0.01)。
     2.3p38MAPK信号通路参与左归丸含药血清促进MC3T3-E1细胞14d矿化作用的调控
     结果显示,与control组比较,ZG和BML均能显著诱导MC3T3-E1细胞14d矿化作用;加入SB203580后,显著抑制ZG和BML对细胞矿化的诱导作用。
     2.4左归丸含药血清通过p38MAPK信号通路的活化上调Runx2蛋白的表达
     结果显示,与control组比较,ZG和BML均能显著上调MC3T3-E1细胞Runx2蛋白的表达;加入SB203580后,显著抑制ZG和BML对Runx2蛋白表达的诱导。
     3.JNK、p38MAPK/Smads信号级联参与左归丸含药血清对MC3T3-E1细胞功能的调控
     3.1左归丸含药血清上调MC3T3-E1细胞TGFβ1mRNA表达
     结果显示,与control组比较,ZG组和BML均组能显著上调MC3T3-E1细胞TGFβ1mRNA的表达(P<0.01)。
     3.2左归丸含药血清诱导MC3T3-E1细胞磷酸化JNK和磷酸化p38蛋白表达
     结果显示,与control组比较,ZG组和BML组均能显著诱导MC3T3-E1细胞磷酸化JNK和p38蛋白的表达;ZG组明显优于BML组;加入SP600125或SB203580后,显著抑制ZG和BML对磷酸化JNK或p38蛋白的诱导作用。
     3.3JNK和p38MAPK信号通路参与左归丸含药血清干预MC3T3-E1细胞增殖的调控
     结果显示,与control组比较,ZG和BML均能显著促进MC3T3-E1细胞增殖(P<0.01);ZG组显著优于BML组(P<0.01);加入SP600125或SB203580后,抑制ZG组和BML组对MC3T3-E1细胞增殖的诱导作用(P<0.05或P<0.01)。
     3.4JNK和p38MAPK信号通路参与左归丸含药血清干预MC3T3-E1细胞周期分布的调控
     细胞周期分析结果显示,与control组比较,ZG组G1期的细胞百分率显著下降(P<0.01),S期的细胞百分率变化不显著(P>0.05),G2/M期的细胞百分率显著增加(P<0.01);与ZG组比较,ZG+SP组和ZG+SB组显著抵抗了ZG对细胞周期的调控作用(P<0.01)。
     3.5左归丸含药血清活化JNK和p38MAPK信号通路有助于磷酸化Smad2/3和上调ColI蛋白表达
     结果显示,与control组比较,ZG和BML均能显著诱导MC3T3-E1细胞Smad2/3蛋白磷酸化和上调ColI蛋白表达水平(P<0.01);对p-Smad2的诱导作用,ZG组显著优于BML组(P<0.01),对ColI蛋白表达的上调作用,ZG组优于BML组(P<0.05),对p-Smad3的诱导作用,两组比较无统计学意义(P>0.05);加入SP600125或SB203580后,显著抑制ZG和BML对MC3T3-E1细胞Smad2/3蛋白磷酸化的诱导作用(P<0.01),显著抑制ZG对MC3T3-E1细胞ColI蛋白表达的上调作用(P<0.01),加入SP600125后显著抑制BML诱导的ColI蛋白表达(P<0.01),加入SB203580后抑制BML诱导的ColI蛋白的表达(P<0.05)。
     3.6左归丸含药血清干预Smad4、Runx2和ColI mRNA表达作用及JNK和p38MAPK通路对其影响
     结果显示,与control组比较,ZG和BML均能显著上调MC3T3-E1细胞Smad4、Runx2和ColI mRNA的表达(P<0.01);对Smad4和Runx2mRNA表达诱导作用ZG组与BML组比较无统计学意义(P>0.05),对ColI mRNA诱导作用ZG组显著优于BML组(P<0.01);加入SP600125后,对ZG诱导Smad4和Runx2mRNA表达的抑制作用没有统计学意义(P>0.05),可以显著抑制ZG诱导的ColI mRNA表达(P<0.01),对BML诱导的ColI mRNA表达的抑制作用没有统计学意义(P>0.05),可以抑制BML诱导的Smad4和Runx2mRNA的表达(P<0.05或P<0.01);加入SB203580后,对ZG诱导Smad4mRNA表达的抑制作用没有统计学意义(P>0.05),可以显著抑制ZG诱导的Runx2和ColI mRNA表达(P<0.01),对BML诱导的Smad4mRNA表达的抑制作用没有统计学意义(P>0.05),可以抑制BML诱导的Runx2和ColI mRNA的表达(P<0.05或P<0.01)。
     结论:
     1.左归丸含药血清具有诱导MC3T3-E1细胞增殖、分化和矿化作用。
     2.JNK、p38MAPK/Smads信号级联参与MC3T3-E1细胞的增殖与分化。
     3.左归丸含药血清可能部分通过活化JNK和p38MAPK通路调控MC3T3-E1细胞功能。
     4.左归丸含药血清可能部分通过对JNK、p38MAPK/Smads信号级联的调节而达到促骨形成作用。
Purpose:To explore the mechanisms of ZuoGui pill to promote bone formation and themediation of JNK/p38MAPK/Smads signaling cascade on it.
     Material and method:Sixty special pathogen free female Sprague-Dawley rats (180~220g) were randomly distributed into three groups. Rats were orally administratedphysiological saline (negative group), ZGP (1.6g/kg) and Premarin suspension (56.25μg/kg,positive group) twice a day for7days. The serum was collected from femoral artery at thetime of2h after last orally administration, then the serum was inactivated at56°C for30min.Murine MC3T3-E1subclone14preosteoblast cell lines (MC3T3cells) were cultured inα-Minimum Essential Medium (α-MEM) supplemented with10%fetal bovine serum (FBS)containing100U/mL penicillin and100μg/mL streptomycin. Cells were assigned to ninegroups, including control, SP600125, SB203580, Zuogui Pills, Zuogui Pills plus SP600125,Zuogui Pills plus SB203580, premarin, premarin plus SP600125and premarin plus SB203580group. MC3T3cells growth arrested for24h in α-MEM containing0.2%FBS andpenicillin-streptomycin, then cells were preincubated in the absence or presence of a specificinhibitors of JNK or p38MAPK (SP600125or SB203580) for60min, then cultured inmedium containing15%control serum, ZGP and Premarin pharmacological serum in theabsence or presence of10μmol/L SP600125or SB203580for48h. Then the effects of ZGPpharmacological serum on cells were observed. phosphorylated level of JNK and p38wereassessed by western blot analysis and immunofluorescence. Cell viability was assayed byMTT method after the cell cultured with ZGP pharmacological serum for48h. Cell cycle wasanalyzed by flow cytometry after propidium iodide staining. The test for alkaline phosphatase(ALP) activity was carried out with PNPP for48h and ALP staining method on the seventhday. Alizarin red staining method was applied to the detection of the mineralizationdeposition. Runx2, ALP, type I collagen (ColI), Smad2/3, p-Smad2and p-Smad3proteinexpression were assayed by western blot analysis. TGFβ1, Smad4, Runx2and ColI mRNAexpression were evaluated by the method of real-time quantitative reverse transcriptase polymerase chain reaction (Q-PCR).
     Results:
     1.JNK signaling mediates ZGP pharmacological serum dependent increase of celldifferentiation
     1.1phosphorylated level of JNK could be increased by ZGP pharmacological serumsignificantly
     The results showed that phosphorylated level of JNK could be promoted by ZGP andpremarin pharmacological serum compared with controls significantly. The effect induced byZGP was better than premarin. The phosphorylated level of JNK induced by ZGPpharmacological serum and premarin was inhibited by SP600125.
     1.2JNK signaling mediates ZGP pharmacological serum dependent increase of ALP activityin telophase
     As compared with the control group, ZGP and premarin group markedly promoted ALPactivity in48h and7days incubation of MC3T3-E1cells (P<0.01). ALP activity inducedin48h incubation by ZGP pharmacological serum was better than premarin pharmacologicalserum (P<0.01). ALP activity induced by ZGP pharmacological serum in48h incubationwas unaffected by SP600125(P>0.05), but ALP activity induced by ZGP pharmacologicalserum in the seventh day incubation was opposed by SP600125.
     1.3JNK signaling mediates ZGP pharmacological serum dependent increase of mineralizeddeposition
     As compared with the control group, ZGP and premarin group significantly promotedmineralized deposition in14days incubation of MC3T3-E1cells. Mineralized depositioninduced by ZGP and premarin pharmacological serum in the fourteenth day incubation wasopposed by SP600125.
     1.4Effects of ZGP pharmacological serum on Runx2and ALP protein expression ofMC3T3-E1cells
     As compared with the control group, ZGP and premarin group markedly promoted Runx2andALP protein expression in48h incubation of MC3T3-E1cells. Runx2and ALP proteinexpression induced by ZGP pharmacological serum and Runx2protein expression induced by premarin pharmacological serum in48h incubation was unaffected by SP600125, but ALPprotein expression induced by premarin pharmacological serum in the seventh day incubationwas opposed by SP600125.
     2.p38MAPK signaling mediates ZGP pharmacological serum dependent increase of celldifferentiation
     2.1phosphorylated level of p38could be increased by ZGP pharmacological serumsignificantly
     The results showed that ZGP and premarin pharmacological serum significantly promotedphosphorylated level of p38compared with controls. SB203580inhibited the phosphorylatedlevel of p38induced by ZGP pharmacological serum and premarin via blocking the p38.
     2.2p38MAPK signaling mediates ZGP pharmacological serum dependent increase of ALPactivity
     As compared with the control group, ZGP and premarin group markedly promoted ALPactivity in48h and7days incubation of MC3T3-E1cells (P<0.01). ALP activity induced in48h incubation by ZGP pharmacological serum was better than premarin pharmacologicalserum (P<0.01). ALP activity induced by ZGP and premarin pharmacological serum in48hand7days incubation was opposed by SB203580(P<0.01).
     2.3p38MAPKsignaling mediates ZGP pharmacological serum dependent increase ofmineralized deposition
     As compared with the control group, ZGP and premarin group significantly promotedmineralized deposition in14days incubation of MC3T3-E1cells. Mineralized depositioninduced by ZGP and premarin pharmacological serum in the fourteenth day incubation wasopposed by SB203580.
     2.4p38MAPKsignaling mediates ZGP pharmacological serum dependent increase of Runx2protein expression
     As compared with the control group, ZGP and premarin group markedly promoted Runx2protein expression in48h incubation of MC3T3-E1cells. Runx2protein expression inducedby ZGP and premarin pharmacological serum in48h incubation was opposed by SB203580.3.JNK/p38MAPK/Smads signaling cascade mediates ZGP pharmacological serum dependentincrease of cell functions
     3.1TGFβ1mRNA expression was up-regulation induced by ZGP and premarinpharmacological serum in MC3T3-E1cells
     As compared with the control group, ZGP and premarin group markedly promoted TGFβ1mRNA expression in48h incubation of MC3T3-E1cells (P<0.01).
     3.2phosphorylated level of JNK and p38could be increased by ZGP pharmacological serumsignificantly
     The results showed that phosphorylated level of JNK and p38could be promoted by ZGP andpremarin pharmacological serum compared with controls significantly. The phosphorylatedlevel of JNK and p38induced by ZGP pharmacological serum and premarin were inhibited bySP600125or SB203580.
     3.3JNK and p38MAPK signaling mediates ZGP pharmacological serum dependent increaseof proliferation
     The results showed that cell proliferation could be stimulated by ZGP and premarinpharmacological serum compared with controls significantly (P<0.01). The effect induced byZGP was better than premarin (P<0.01). Cell proliferation induced by ZGP pharmacologicalserum and premarin was inhibited by SP600125or SB203580(P<0.05or P<0.01).
     3.4JNK and p38MAPK signaling mediates ZGP pharmacological serum dependentregulation of different cell cycle phases
     The cell cycle results were analyzed and showed that ZGP reduced the percentage of“G1-Period” Cells and dramatically increased the percentage of “G2/M-Period” Cells in CellCycle (P<0.01), while the percentage of “S-Period” was unaffected (P>0.05). The regulationof different cell cycle phases induced by ZGP pharmacological serum and premarin wasinhibited by SP600125or SB203580(P<0.01).
     3.5JNK and p38MAPK signaling mediates ZGP pharmacological serum dependent increaseof phosphorylated level of Smad2/3and ColI protein expressionAs compared with the control group, ZGP and premarin group markedly promotedphosphorylated level of Smad2/3and ColI protein expression in48h incubation ofMC3T3-E1cells (P<0.01). phosphorylated level of Smad2induced in48h incubation byZGP pharmacological serum was better than premarin pharmacological serum (P<0.01) andColI protein expression induced by ZGP pharmacological serum was better than premarin pharmacological serum (P<0.05). phosphorylated level of Smad3induced by ZGPpharmacological serum was not better than premarin pharmacological serum(P>0.05).phosphorylated level of Smad2and phosphorylated level of Smad3induced by ZGP andpremarin pharmacological serum were opposed by SP600125or SB203580(P<0.01). ColIprotein expression induced by ZGP pharmacological serum was opposed by SP600125orSB203580(P<0.01). ColI protein expression induced by premarin pharmacological serumwas opposed by SP600125(P<0.01), while ColI protein expression induced by premarinpharmacological serum was opposed by SB203580(P<0.05).
     3.6JNK and p38MAPK signaling mediates ZGP pharmacological serum dependent increaseof Smad4, Runx2and ColI mRNA expression
     As compared with the control group, ZGP and premarin group markedly promoted Smad4,Runx2and ColI mRNA expression in48h incubation of MC3T3-E1cells (P<0.01). Smad4and Runx2mRNA expression induced by ZGP was not better than premarin. ColI mRNAexpression induced by ZGP was better than premarin (P<0.01). Smad4and Runx2mRNAexpression induced by ZGP pharmacological serum were unaffected (P>0.05), while ColImRNA expression induced by ZGP pharmacological serum was opposed by SP600125(P<0.01). ColI mRNA expression induced by premarin pharmacological serum was unaffected(P>0.05), while Smad4and Runx2mRNA expression induced by premarin pharmacologicalserum were opposed by SP600125(P<0.05or P<0.01). Smad4mRNA expression inducedby ZGP pharmacological serum was unaffected (P>0.05), while Runx2and ColI mRNAexpression induced by ZGP pharmacological serum were opposed by SB203580(P<0.01).Smad4mRNA expression induced by premarin pharmacological serum was unaffected(P>0.05), while Runx2and ColI mRNA expression induced by premarin pharmacologicalserum were opposed by SB203580(P<0.05or P<0.01).
     Conclusion:
     1.Cell proliferation, differentiation and mineralization in MC3T3-E1cells could be inducedby ZGP pharmacological serum.
     2.JNK/p38MAPK/Smads signaling cascade were associated with cell proliferation anddifferentiation in MC3T3-E1cells.
     3.JNK/p38MAPK signaling mediates ZGP pharmacological serum dependent increase of cellfunctions.
     4.JNK/p38MAPK/Smads signaling cascade mediates ZGP pharmacological serum dependentpromotion of bone formation.
引文
[1]武密山,武博文,任立中,等.骨靶向亲和性引经药防治原发性骨质疏松症[J].中国组织工程研究,2012,16(30):5569-5576.
    [2]李恩,薛延,王洪复,等.骨质疏松鉴别诊断与治疗[M].北京:人民卫生出版社,2005.
    [3]Manolagas SC,Jilka RL.Emerging insights into the pathophysiology of osteoporosis[J].NEngl J Med,1995,332(5):305-311.
    [4]肖建德.实用骨质疏松学[M].北京:科学出版社.2004.
    [5]侯进,熊晓云,弥曼,等.xy9902对MC3T3-E1细胞的增殖和分化作用[J].中国药理学通报,2006,22(05):529-532.
    [6]郑丽,董进.IGF-1对MC3T3-E1细胞凋亡及凋亡调控蛋白Bax、Bcl-2表达的影响[J].中西医结合心脑血管病杂志,2008,6(06):688-689.
    [7]马攀,刘洪臣,马军利,等.格列美脲对大鼠下颌骨成骨细胞增殖分化及矿化的影响[J].中华老年口腔医学杂志,2012,10(01):1-4.
    [8]赵奇江,李芬芬,董新威,等.他汀类药物对TNF诱导成骨细胞生长抑制的保护作用[J].中国现代应用药学,2012,29(05):389-393.
    [9]刘铭,朱振安,汤亭亭.辛伐他汀对成骨样细胞MC3T3-E1增殖功能的影响[J].中国骨质疏松杂志,2008,14(02):101-105.
    [10]刘钰瑜,姚卫民,徐道华,等.卡托普利对MC3T3-E1细胞增殖、分化及Ⅰ型胶原mRNA表达的影响[J].中国药理学通报,2011,27(09):1259-1262.
    [11]张娟,董进.胰岛素样生长因子-1对MC3T3-E1细胞增殖及Ⅰ型胶原蛋白合成的影响[J].中西医结合心脑血管病杂志,2009,7(06):705-707.
    [12]张金超,刘丹丹,易长青,等.粒子对MC3T3-E1细胞增殖、分化和矿化功能的影响[J].科学通报,2010,(06):435-441.
    [13]王福山,牟慧琴.骨质疏松症的中医药治疗和研究进展[J].中国民族民间医药,2011,20(03):20-21.
    [14]徐祖健,汪付,尹思源,等.绝经后妇女原发性骨质疏松症辨证分型与症候特征的探讨[J].中医正骨,2008,20(11):1-2+85.
    [15]宋敏,柳申鹏.骨质疏松症的中医药研究述评[J].中国中医骨伤科杂志,2005,13(06):71-73.
    [16]刘维嘉,麦敏军,刘永坤.中医药治疗原发性骨质疏松症的研究近况[J].中国骨质疏松杂志,2009,15(05):374-376.
    [17]李中伟,许勇.中医药防治骨质疏松的理论基础与临床应用[J].中国中医骨伤科杂志,2010,18(12):71-73.
    [18]刘建宁,王晔.中医药治疗绝经后骨质疏松症概况[J].中国中医基础医学杂志,2011,17(04):465-466.
    [19]金华,章薇,袁静.单味中药防治骨质疏松症的研究进展[J].中国中医骨伤科杂志,2006,14(04):75-77.
    [20]董远芳.淫羊藿等中药治疗骨质疏松症的临床观察[J].中药材,2004,27(08):620-622.
    [21]马中书,王蕊,邱明才,等.四种补肾中药对去卵巢大鼠骨质疏松骨形态的作用[J].中华妇产科杂志,1999,34(02):17-20.
    [22]卞玉群,李超,谭峰,等.补肾方药治疗骨质疏松症的实验研究近况述评[J].山西中医学院学报,2012,13(03):141-143+162.
    [23]宋钦兰.骨碎补、续断、西洋参对MC3T3-E1细胞细胞增殖的影响[J].山东中医药大学学报,2007,31(04):332-333.
    [24]冯淑华,陈虹,刘薇.8味中药提取物对MC3T3-E1细胞增殖及碱性磷酸酶活性的影响[J].中成药,2010,32(04):661-663.
    [25]刘波,张睿,徐彭,等.淫羊藿对去卵巢大鼠骨质疏松的影响[J].中国实验方剂学杂志,2013,19(07):168-171.
    [26]宋渊,李盛华,何志军,等.淫羊藿苷对去势雌性大鼠骨质疏松的影响[J].军医进修学院学报,2012,33(04):400-403.
    [27]吴涛,徐俊昌,南开辉,等.淫羊藿苷促进羊骨髓间充质干细胞的增殖和成骨分化[J].中国组织工程研究与临床康复,2009,13(19):3725-3729.
    [28]吴昊,查振刚,姚平,等.淫羊藿苷对骨髓间充质干细胞骨向诱导的实验研究[J].中国中西医结合杂志,2010,30(04):410-415.
    [29]毛项颖,卞琴,沈自尹.淫羊藿苷介导MAPK信号通路促进间充质干细胞株C3H10T1/2成骨分化的体外研究[J].中西医结合学报,2012,10(11):1272-1278.
    [30]蔡玉霞,张剑宇.补骨脂水煎剂对去卵巢骨质疏松大鼠骨代谢的影响[J].中国组织工程研究与临床康复,2009,13(02):268-271.
    [31]翟远坤,武祥龙,潘亚磊,等.补骨脂抗骨质疏松研究概况[J].中医杂志,2012,53(14):1244-1248.
    [32]宋芹,董小萍,郁小兵.补骨脂提取物对体外培养小鼠成骨细胞株MC3T3-E1细胞分化的影响[J].中国中药杂志,2009,34(10):1264-1267.
    [33]宋芹,颜军,郭晓强,等.补骨脂与罗非鱼鳞胶原蛋白多肽对MC3T3-E1细胞ALP、Ⅰ型胶原蛋白mRNA表达的影响[J].中药药理与临床,2011,27(01):47-49.
    [34]贾红蔚,王宝利,邝晨钟,等.骨碎补与雌激素对去卵巢大鼠骨质疏松作用的对照研究[J].中国中西医结合杂志,2006(S1):116-119.
    [35]李展春,程光齐,臧危平,等.骨碎补治疗去卵巢大鼠骨质疏松的实验研究[J].中国中医骨伤科杂志,2011,19(04):9-11.
    [36]张上上,刘心昱,郑姝宁,等.骨碎补醇提物对骨质疏松预防作用的大鼠尿液UPLC-MS/MS代谢组学研究[J].中国中药杂志,2012,37(05):658-662.
    [37]唐琪,陈莉丽,严杰.骨碎补提取物促小鼠成骨细胞株MC3T3-E1细胞增殖、分化和钙化作用的研究[J].中国中药杂志,2004,29(02):73-77.
    [38]阳波,杨静.黄芪对绝经后骨质疏松症患者影响的临床研究[J].四川医学,2007,28(03):291-293.
    [39]欧莉,程虎印,曾小红.黄芪配伍熟地治疗绝经后骨质疏松29例[J].中国老年学杂志,2012,32(18):3990-3991.
    [40]张宏波,曹文波,崔文璟,等.黄芪多糖抗去势雌鼠骨质疏松的实验研究[J].玉林师范学院学报,2012,33(05):51-55.
    [41]刘心萍,陈方亮,程亚军.黄芪总黄酮防止大鼠骨质疏松的实验研究[J].浙江中西医结合杂志,2005,15(05):282-283.
    [42]孙玉明,王培民,查炜,等.葛根对原发性骨质疏松症患者临床症状影响的研究[J].中国中医骨伤科杂志,2008,16(04):14-16.
    [43]王新祥,张允岭,吴坚,等.葛根对睾丸切除骨质疏松模型小鼠骨密度和骨构造的作用[J].中国组织工程研究与临床康复,2010,14(07):1262-1266.
    [44]李俊华,潘子毅.葛根素对绝经后骨质疏松症患者血清IL-4、IL-6、IL-10和雌激素水平的影响[J].中国中医骨伤科杂志,2007,15(05):28-29.
    [45]李海,王金花,黄海玲,等.葛根素联合雌二醇对去卵巢大鼠骨质疏松的治疗作用[J].中国老年学杂志,2012,32(18):3950-3952.
    [46]李海,王金花,黄海玲,等.葛根素联合雌二醇对去卵巢大鼠骨质疏松症的治疗作用(英文)[J].中国组织工程研究与临床康复,2011,15(46):8698-8701.
    [47]扶晓明,周艳,李梓民,等.葛根异黄酮联合维生素D防治骨质疏松的协同效应(英文)[J].中国组织工程研究与临床康复,2011,15(11):2083-2086.
    [48]欧莉,曾小红,赵鹏.熟地、黄芪为主药治疗绝经后骨质疏松症的临床观察[J].北京中医药,2011,30(08):605-606.
    [49]陶怡,沈涛,马晖.熟地黄及其不同配伍药对治疗糖尿病大鼠骨质疏松的药效比较[J].中国实验方剂学杂志,2012,18(08):249-251.
    [50]武密山,赵素芝,李恩,等.地黄活性成分梓醇对小鼠MC3T3-E1细胞增殖、分化和矿化的影响[J].中国药理学通报,2010,26(04):509-513.
    [51]肖柳斌,刘国雄,邹丽宜.丹参防治小鼠肝纤维化性骨质疏松的作用研究[J].中国中西医结合外科杂志,2006,12(02):143-146.
    [52]崔燎,邹丽宜,刘钰瑜,等.丹参水提物和丹参素促进成骨细胞活性和防治泼尼松所致大鼠骨质疏松[J].中国药理学通报,2004,20(03):286-291.
    [53]肖柳斌,刘国雄,王晖.丹参水提液对抗大鼠泼尼松性骨质疏松的作用研究[J].中国临床药理学与治疗学,2008,13(01):94-98.
    [54]李艳,董佳梓,贾朝娟,等.五味子对去卵巢致骨质疏松大鼠胫骨护骨素、核因子-κB受体活化因子配体蛋白表达的影响[J].中医杂志,2010,51(11):1028-1030.
    [55]樊秦,赵文君,李应东.华中五味子含药血清对成骨细胞增殖分化的影响[J].中国实验方剂学杂志,2009,15(02):33-35.
    [56]蒙海燕,曲晓波,李娜,等.鹿茸及鹿角胶对去卵巢大鼠骨质疏松症的影响[J].中药材,2009,32(02):179-182.
    [57]刘盟,马玉,高晓黎,等.新疆马鹿角提取物对MC3T3-E1细胞增殖、分化和矿化的影响[J].中成药,2012,34(01):148-151.
    [58]程孟春,刘艳秋,王莉,等.何首乌和菟丝子对破骨细胞和成骨细胞增殖及分化的影响[J].中国中药杂志,2011,36(19):2737-2740.
    [59]蔡西国,赵素霞.菟丝子黄酮干预去卵巢大鼠骨代谢研究[J].中药药理与临床,2007,23(06):27-29.
    [60]贾朝娟,鞠大宏,刘梅洁,等.山药对卵巢切除大鼠骨质疏松症的治疗作用及其机理探讨[J].中国中医基础医学杂志,2009,15(04):268-271.
    [61]马锋,高俊,沈军,等.含枸杞多糖血清对MC3T3-E1成骨细胞增殖、分化和矿化的影响[J].宁夏医学杂志,2012,34(02):108-111.
    [62]冯伟,傅文彧,朱雅萍,等.接骨中药对培养成骨细胞增殖、分化和矿化功能的影响[J].浙江中医学院学报,2004,28(02):65-67.
    [63]常德有,杨靖,董福慧.阿胶对体外培养大鼠成骨细胞增殖、分化功能的影响[J].中国老年学杂志,2009,29(24):3230-3232.
    [64]杨玮,徐道华,周晨慧,等.小檗碱对骨髓间充质干细胞成骨分化的影响[J].中国药理学通报,2010,26(12):1560-1563.
    [65]卢建华,王维佳,何昱,等.红曲有效成分对体外培养成骨细胞增殖分化及矿化功能的影响[J].浙江中医药大学学报,2011,35(04):552-554+580.
    [66]卢建华,王维佳,陈冰.红曲对体外培养成骨细胞增殖、分化及矿化功能的影响[J].中国中医骨伤科杂志,2006,14(02):41-45.
    [67]丁佩惠,唐琪,陈莉丽.柚皮苷对小鼠MC3T3-E1细胞增殖、分化和矿化的影响[J].中国中药杂志,2009,34(13):1712-1716.
    [68]赵丹,潘智敏.原发性骨质疏松症的中医药治疗近展[J].江西中医药,2011,42(06):72-73.
    [69]邹军,吕爽,屠嘉衡,等.有氧运动配合左归丸对去卵巢所致大鼠骨质疏松的影响[J].中国中医基础医学杂志,2009,15(04):272-273+276.
    [70]刘梅洁,潘静华,李艳,等.左归丸对糖皮质激素所致骨质疏松大鼠血清中BGP,IGF-Ⅰ含量的影响[J].中国实验方剂学杂志,2011,17(16):133-136.
    [71]李鸿泓,鞠大宏,滕静如,等.左归丸对糖皮质激素所致骨质疏松大鼠血清中E_2、PTH含量的影响[J].中国中医基础医学杂志,2011,17(07):744-745+763.
    [72]吕海波,任艳玲,王莹,等.左归丸防治去卵巢大鼠骨质疏松症的实验研究[J].中国骨质疏松杂志,2010,16(11):847-850+823.
    [73]赵文武.六味地黄丸合二至丸治疗更年期综合征30例[J].职业与健康,2002,18(02):133-134.
    [74]邹崇祺.六味地黄汤加味治疗绝经后骨质疏松症的临床观察[J].广州中医药大学学报,2005,22(04):267-268+272.
    [75]林雄浩,吴锦忠.中药复方二至丸的化学成分及抗骨质疏松研究进展[J].解放军药学学报,2009,25(05):421-424.
    [76]虞巧英.二至丸治疗更年期骨质疏松症临床疗效观察[J].海峡药学,2009,21(11):169-170.
    [77]邢薇薇,张宏,吴锦忠,等.二至丸对肾阴虚骨质疏松大鼠的影响[J].福建中医药,2008,39(06):45-47.
    [78]华刚,管爱芬,张敏.右归丸加减治疗骨质疏松症82例[J].四川中医,2008,26(04):105.
    [79]梁启明,许小志,潘国铨,等.右归丸加减治疗肾虚型骨质疏松症的临床研究[J].中国医药指南,2011,9(33):5-7.
    [80]罗汉文,关宏刚.右归丸对骨质疏松模型大鼠垂体-肾上腺轴影响的实验研究[J].贵阳中医学院学报,2006,28(02):60-62.
    [81]杨铸,卞琴,赵永见,等.左归丸、右归丸加减方对去卵巢骨质疏松小鼠的疗效比较和基因芯片分析[J].中国河南洛阳.2012,4.
    [82.王建伟,马勇,尹恒.金匮肾气丸联用葡萄糖酸钙对原发性骨质疏松症疼痛临床疗效观察[J].辽宁中医药大学学报,2012,14(02):5-6.
    [83]王建伟,马勇,张亚峰,等.金匮肾气丸联用葡萄糖酸钙治疗原发性骨质疏松症的临床研究[J].中国骨质疏松杂志,2011,17(10):912-914+911.
    [84]王建伟,马勇,周玲玲,等.金匮肾气丸联合葡萄糖酸钙对去势大鼠骨质疏松的影响[J].中国骨质疏松杂志,2011,11(01):60-63.
    [85]孔令青,李鸣镝.中医方剂“五子衍宗丸”组方的历史源流[J].中国中医基础医学杂志,2009,15(01):67-68.
    [86]牛丽颖,刘娇,李清,等.五子衍宗丸对去势致骨质疏松大鼠骨形态计量学参数的影响[J].中药药理与临床,2008,24(04):5-7.
    [87]郑雨,陈东风,周健洪,等.加味左归饮对去卵巢骨质疏松大鼠下丘脑雌激素α受体的影响[J].中药新药与临床药理,2004,15(04):229-232.
    [88]童培建,胡柏松,金红婷,等.右归饮对激素性股骨头坏死大鼠体外诱导培养成骨细胞作用的研究[J].中国骨伤,2010,23(01):23-27.
    [89]孙峰,王文君,李大金,等.“更年春”方对大鼠成骨细胞增殖与分化及矿化的影响[J].上海中医药杂志,2006,40(07):60-62.
    [90]张红.从补肾健脾法谈中医药对骨质疏松症的防治[J].时珍国医国药,2007,18(09):2250.
    [91]林越,张宁,刘世巍,等.补肾活血方对hPTH(1-34)干预下小鼠MC3T3-E1细胞增殖及分化的影响[J].中华临床医师杂志(电子版),2012,6(11):3065-3068.
    [92]臧洪敏,陈君长,刘亦恒,等.补肾活血方对大鼠成骨细胞增殖、分化及矿化的影响[J].中国中西医结合杂志,2005,25(12):1112-1114.
    [93]许旺.中医药治疗骨质疏松症的研究进展[J].中草药,2005,36(02):314-315.
    [94]郭俊,李磊,高波,等.中医药治疗骨质疏松症研究进展[J].广西中医学院学报,2009,12(01):70-72.
    [95]姜向坤,刘敏如.补肾复方胶囊含药血清对成骨细胞增殖、分化和矿化功能的影响[J].成都中医药大学学报,2009,32(01):46-50.
    [96]万蕾,卢建华.骨质疏松症中医药治疗研究进展[J].中医正骨,2008,20(11):75-76.
    [97]陈述祥,康乐.中药促成骨细胞增殖和分化的机制与作用[J].中国组织工程研究,2012,16(07):1299-1302.
    [98]李春年,李淑娟,杨冬茹,等.左归丸和戊酸雌二醇对实验性骨质疏松牙周炎大鼠牙周组织及血清中碱性磷酸酶、钙、磷影响的研究[J].中国实用口腔科杂志,2010,3(08):474-476.
    [99]蒋天骄.中药左归丸联合维D磷酸氢钙片治疗骨质疏松症疗效观察[J].临床合理用药杂志,2011,4(07):54.
    [100]赵尧春,刘立云,王璘琳.加味左归丸治疗原发性骨质疏松症(肾阴虚型)的临床疗效观察[J].中国医药指南,2010,8(28):60-61.
    [101]俞庆三,王青.左归丸联合复方骨肽注射液治疗绝经后骨质疏松症[J].中国实验方剂学杂志,2012,18(10):302-303.
    [102]严炜,吕荣,李道鸿,等.左归丸治疗肝肾亏虚型老年性骨质疏松症临床观察[J].吉林医学,2012,33(35):7670-7671.
    [103]张秋万.左归丸加减治疗绝经后骨质疏松症20例[J].中国民间疗法,2007,15(12):29-30.
    [104]黄波.左归丸合二至丸治疗肾阴虚型围绝经期诸症64例[J].现代医药卫生,2008,24(03):404-405.
    [105]曹静.益心舒联合左归丸治疗绝经期综合征的临床观察[J].中西医结合心脑血管病杂志,2012,10(08):1010-1011.
    [106]肖文,李仓霞,薛海龙,等.左归丸合解郁丸治疗围绝经期妇女抑郁症45例[J].中国实验方剂学杂志,2012,18(19):278-280.
    [107]赵旭,代平,金钊,等.经方左归丸抗糖皮质激素性骨质疏松症的效应及机制研究[J].时珍国医国药,2009,20(09):2243-2244.
    [108]夏花英,张江虹,谢月霞,等.左归丸对D-半乳糖致衰老大鼠血液抗氧化能力和淋巴细胞DNA损伤的影响[J].中华中医药杂志,2012,27(11):2937-2939.
    [109]孙琳林,卢林,王书惠,等.左归丸与六味地黄丸对衰老大鼠抗自由基能力及P_(16)蛋白表达影响的比较研究[J].中国临床保健杂志,2012,15(03):274-276.
    [110]姚建平,金国琴,戴薇薇,等.左归丸和右归丸对老年大鼠海马GRmRNA表达与HPA轴活性的影响[J].中国实验方剂学杂志,2009,15(10):67-69.
    [111]李红梅,苗竹林,王奇玲,等.左归丸对免疫性卵巢早衰小鼠性激素的影响[J].中国现代应用药学,2010,27(13):1161-1164.
    [112]李红梅,朱玲,钟志勇,等.左归丸对免疫性卵巢早衰小鼠卵巢GDF-9、BMP-15表达的影响[J].临床和实验医学杂志,2012,11(19):1517-1518.
    [113]朱玲,罗颂平,许丽绵,等.左归丸对免疫性卵巢早衰小鼠卵巢Bcl-2、Bax蛋白表达的影响[J].中药新药与临床药理,2012,23(04):381-386.
    [114]李春年,李淑娟,杨冬茹,等.左归丸对实验性骨质疏松牙周炎大鼠牙周组织OPG、RANKL表达的影响[J].实用口腔医学杂志,2011,27(02):270-273.
    [115]姚建平,金国琴,戴薇薇,等.左归丸和右归丸对老年大鼠杏仁核促肾上腺皮质激素释放激素表达与HPA轴活性作用的实验研究[J].时珍国医国药,2010,21(09):2170-2171.
    [116]刘梅洁,鞠大宏,赵宏艳,等.“肾主骨”的机理研究—左归丸含药血清对破骨细胞分化调控因子OPG、 RANKL蛋白表达的影响[J].中国中医基础医学杂志2009,15(03):184-187+196.
    [117]鞠大宏,刘梅洁,赵宏艳,等.左归丸含药血清对成骨细胞OPG、RANKL mRNA表达的影响[J].北京中医药大学学报,2008,31(05):312-315.
    [118]王小琴,袁军,马晓红,等.左归丸对5/6肾大部切除模型并肾性骨病大鼠骨代谢指标的影响[J].中国中西医结合肾病杂志,2012,13(07):584-586.
    [119]丁富平,黄进,张进,等.左归丸对肾虚大鼠MSCs增殖的影响[J].时珍国医国药,2011,22(05):1062-1064.
    [120]谭峰,樊巧玲,王明艳,李沐涵,尹鑫.左归丸对SD大鼠骨髓间充质干细胞增殖的影响[J].中国实验方剂学杂志,2011,17(13):145-148.
    [121]徐凌霄,王芳,郭敦明,等.左归丸对间充质干细胞向软骨细胞分化过程中Ⅱ型胶原及蛋白多糖基因表达的影响[J].中国中西医结合杂志,2011,31(12):1662-1668.
    [122]]刘梅洁,李艳,潘静华,等.Effects of Zuogui Pill (左归丸) on Wnt Singal Transduction inRats with Glucocorticoid-induced Osteoporosis[J].Journal of Traditional ChineseMedicine,2011,32(02):98-102.
    [123]白雪东,胡蕴玉,李航,等.小鼠p38MAPK重组腺病毒载体的构建及其在成骨细胞系中的表达[J].细胞与分子免疫学杂志,2011,27(04):367-369+373.
    [124]Brennan TC,Rybchyn MS,Green W,et al.Osteoblasts play key roles in the mechanismsof action of strontium ranelate[J].Br J Pharmacol,2009,157(7):1291-1300.
    [125]Leong WF, Zhou T, Lim GL,et al.Protein palmitoylation regulates osteoblastdifferentiation through BMP-induced osterix expression[J].PLoS One,2009,4(1):4135.
    [126]Rodan GA, Martin TJ.Therapeutic approaches to bone diseases[J]. Science,2000,289(5484):1508-1514.
    [127]Hsieh TP, Sheu SY, Sun JS,et al.Icariin isolated from Epimedium pubescens regulatesosteoblasts anabolism through BMP-2, SMAD4, and Cbfa1expression[J]. Phytomedicine2010;17(6):414-423.
    [128]朱晓峰,张荣华,孙升云,等.淫羊藿素通过雌激素受体和骨形态发生蛋白信号诱导MC3T3-E1subclone14细胞分化[J].中国病理生理杂志,2011,27(12):2351-2356.
    [129]Don MJ,Lin LC,Chiou WF.Neobavaisoflavone stimulates osteogenesis viap38-mediated up-regulation of transcription factors and osteoid genes expression inMC3T3-E1cells[J].Phytomedicine,2012,19(6):551-561.
    [130]姚新苗,冷涛,张云鹏.益骨口服液含药血清对成骨细胞增殖、分化及矿化功能的影响[J].中国中医骨伤科杂志,2010(01):6-8.
    [131]马锋,高俊,王一农,沈军,马敬祖.枸杞多糖含药血清对MC3T3-E1细胞内Ca~(2+)及Ⅰ型胶原蛋白合成的影响[J].中国组织工程研究,2012,16(20):3743-3746.
    [132]Raman M,Chen W,Cobb MH.Differential regulation and properties ofMAPKs[J].Oncogene,2007,26(22):3100-3112.
    [133]Molina JR,Adjei AA.The Ras/Raf/MAPK pathway[J].J Thorac Oncol,2006,1(1):7-9.
    [134]Johnson GL,Lapadat R.Mitogen-activated protein kinase pathways mediated byERK,JNK,and p38protein kinases[J].Science,2002,298(5600):1911-1912.
    [135]袁凌青,罗湘杭,郭丽娟,等.组织金属蛋白酶抑制因子3重组蛋白诱导小鼠MC3T3-E1细胞凋亡[J].中华内分泌代谢杂志,2006,22(03):229-233.
    [136]周筠,张秀珍,宋利格.MAPK信号通路参与17β-雌二醇对维生素D受体表达的调控[J].第二军医大学学报,2011,32(12):1291-1295.
    [137]彭文芳,张秀珍.丝裂原活化蛋白激酶信号转导通路与骨质疏松[J].国外医学内分泌学分册,2004,24(04):233-236.
    [138]李烨,刘文锋,刘如石,等.丙二醛通过激活p38和JNK通路抑制间充质干细胞成骨分化[J].中国生物化学与分子生物学报,2012,28(09):804-810.
    [139]Kim do Y,Jung MS,Park YG,et al.Ginsenoside Rh2(S) induces the differentiation andmineralization of osteoblastic MC3T3-E1cells through activation of PKD and p38MAPKpathways[J].BMB Rep,2011,44(10):659-664.
    [140]Choi EM.Protective effect of quercitrin against hydrogen peroxide-induced dysfunctionin osteoblastic MC3T3-E1cells[J].Exp Toxicol Pathol2010,64(3):211-216.
    [141]Tang CH,Yang RS,Chien MY,et al.Enhancement of bone morphogenetic protein-2expression and bone formation by coumarin derivatives via p38and ERK-dependent pathwayin osteoblasts[J].Eur J Pharmacol,2008,579(1-3):40-49.
    [142]高怀林,王玲玲,贾振华,等.P38MAPK通路在内皮细胞F-actin蛋白表达中的作用及通络药物的影响[J].中国中医基础医学杂志,2010,16(09):769-771.
    [143]徐道晶,王锦,何娟文,等.p38蛋白激酶参与BMP9诱导的C3H10T1/2细胞成骨分化[J].中国生物工程杂志,2011,31(05):15-21.
    [144]Tang SY,Xie H,Yuan LQ,et al.Apelin stimulates proliferation and suppresses apoptosisof mouse osteoblastic cell line MC3T3-E1via JNK and PI3-K/Akt signalingpathways[J].Peptides2007,28(3):708-718.
    [145]Liang D,Yang M,Guo B,et al.Zinc inhibits H(2)O(2)-induced MC3T3-E1cells apoptosisvia MAPK and PI3K/AKT pathways[J].Biol Trace Elem Res,2012,148(3):420-429.
    [146]王卫华.JNK抑制剂对鼠实验性牙周病后基质金属蛋白酶-2mRNA影响的研究[J].口腔医学,2012,32(04):232-234.
    [147]兰爱平,莫利求,郑东诞,等.JNK信号通路介导化学性缺氧对PC12细胞的损伤作用[J].中国药理学通报,2012,28(01):62-66.
    [148]刘立萍,任艳玲,李然,等.左归丸含药血清通过JNK信号通路诱导MC3T3成骨细胞分化的研究[J].中成药,2012,34(08):1433-1437.
    [149]刘立萍,任艳玲,李然,等.JNK通路对左归丸含药血清调控MC3T3成骨细胞Runx2mRNA表达的影响[J].中国实验方剂学杂志,2012,18(14):143-146.
    [150]Ge C,Xiao G,Jiang D,et al.Critical role of the extracellular signal-regulatedkinase-MAPK pathway in osteoblast differentiation and skeletal development[J].J CellBiol,2007,176(5):709-718.
    [151]蒿长英,任艳玲,赵金茹.左归丸含药血清通过ERK/Smads信号通路干预MC3T3-E1细胞的功能基因表达[J].中国药理学通报,2012,28(06):872-876.
    [152]Yano M,Inoue Y,Tobimatsu T,Hendy G,et al.Smad7inhibits differentiation andmineralization of mouse osteoblastic cells[J].Endocr J,2012,59(8):653-662.
    [153.]吴昆鹏,杨巧珠,马凤燕,等.小分子肽对成骨前体细胞MC3T3-E1骨保护素和核转录因子κ B受体活化因子配体表达的影响[J].中国组织工程研究与临床康复,2011,15(24):4375-4378.
    [154]刘义,吴昆鹏,杨巧珠,等.小分子肽(KP)对小鼠成骨前体细胞MC3T3-E1增殖分化及NF-κB表达的影响[J].中国药理学通报,2011,27(03):423-427.
    [155]张志梅,李玉坤.他汀类药物对骨代谢影响的研究进展[J].国际药学研究杂志,2009,36(02):100-104.
    [156]刘盟,马玉,冯崴,等.新疆马鹿角提取物对MC3T3-E1成骨细胞RANKL/OPG mRNA表达的影响[J].中药新药与临床药理,2011,22(06):605-609.
    [157]牛银波,李宇华,黄海涛,等.染料木素促成骨样细胞增殖作用及其机制研究[J].中国药理学通报,2010,26(10):1309-1312.
    [158]Somjen D,Katzburg S,Sharon O,et al.The effects of estrogen receptors alpha-andbeta-specific agonists and antagonists on cell proliferation and energy metabolism in humanbone cell line[J].J Cell Biochem,2011,112(2):625-632.
    [159]Jin M,Jin F,Zhang L,et al.Two estrogen replacement therapies differentially regulateexpression of estrogen receptors alpha and beta in the hippocampus and cortex ofovariectomized rat[J].Brain Res Mol Brain Res,2005,142(2):107-114.
    [160]何陨,唐婉容,吴恙,等.选择性雌激素受体调节剂对MC3T3-E1细胞生长分化的调节[J].第三军医大学学报,2012,34(16):1617-1620.
    [161]鲍捷,王国祥.PI3K/Akt信号通路应力与应激对β-连环蛋白及骨代谢的共刺激[J].中国组织工程研究,2012,16(37):7003-7008.
    [162]高建林,李青南.第二信使cAMP、cGMP信号通路调节骨形成的研究进展[J].中国药理学通报,2010,26(12):1545-1549.
    [163]黄莺飞.中医药防治绝经后骨质疏松症的实验研究进展[J].内科,2008,3(01):127-129.
    [164]刘立萍,李然,任艳玲.基于肾藏精理论解析《神农本草经》之益精药[J].中华中医药杂志,2012,27(03):528-530.
    [165]赵治友,邬亚军.骨质疏松症的中医辨证思路与治法研究[J].浙江中医药大学学报,2007,31(03):275-276.
    [166]梁茂新,范颖.论西医疾病辨证规范所属症状虚拟性及其成因[J].中华中医药杂志,2012,27(03):544-547.
    [167]孙益,童培建,肖鲁伟.绝经后骨质疏松症与中医体质的相关性[J].中医杂志,2009,50(08):696-698.
    [168]马坤.六味地黄丸与左归丸在补阴方面的临床应用[J].吉林中医药,2011,31(03):260-261+276.
    [169]陈晓飞,陈素红,吕圭源,等.补肾活血方治疗骨质疏松症机制研究及探讨[J].中成药,2011,33(07):1130-1134.
    [170]俞益火,陈久毅,王建.二至丸治疗绝经后骨质疏松症的疗效观察[J].中国中医骨伤科杂志,2011,19(04):42-43.
    [171]黄勇,黄秀深,胡一梅,等.“左归丸”配伍规律对骨髓源成体干细胞定向分化的调控作用[J].成都中医药大学学报,2010,33(01):48-52.
    [172]陈敏,李政木,高洁,等.“阴中求阳”理论及其主方肾气丸的研究探讨[J].河南中医学院学报,2007,22(03):12-14.
    [173]李春年,李淑娟,杨冬茹,等.骨质疏松性牙周炎大鼠左归丸与雌激素治疗后颌骨结构和骨量的变化[J].中国组织工程研究与临床康复,2011,15(33):6211-6215.
    [174]蒿长英,任艳玲,刘立萍,等.左归丸含药血清通过ERK/TGF-β/Smads信号级联调控MC3T3-E1细胞增殖与分化[J].中国病理生理杂志,2012,28(09):1670-1675.
    [175]原野,张明伟,于晓妉,等.FGF2对BMP-Smads信号转导通路的负调节机制初步研究[J].临床口腔医学杂志,2005,21(03):134-137.
    [176]关键,程宗生,王健平,等.成骨细胞中Runx2对机械离心力刺激的响应[J].华西口腔医学杂志,2010(01):38-40+44.
    [177]Lee HS,Jung EY,Bae SH,et al.Stimulation of osteoblastic differentiation andmineralization in MC3T3-E1cells by yeast hydrolysate[J]. Phytother Res,2011,25(5):716-723.
    [178]徐凌霄,高俊,张前德.左归丸含药血清对大鼠骨髓间充质干细胞骨向分化中碱性磷酸酶含量的影响[J].中国实验方剂学杂志,2011,17(07):149-152.
    [179]李茂强,朱振安,刘凤翔,等.钛颗粒对大鼠成骨细胞增殖、分化及矿化功能的影响[J].中国组织工程研究与临床康复,2009,13(26):5047-5051.
    [180]Kim HK,Kim JH,Abbas AA,et al.Alendronate enhances osteogenic differentiation ofbone marrow stromal cells: a preliminary study[J].Clin Orthop Relat Res,2009,467(12):3121-3128.
    [181]Itoh D,Yoneda S,Kuroda S,et al.Enhancement of osteogenesis on hydroxyapatite surfacecoated with synthetic peptide in vitro[J].J Biomed Mater Res,2002,62(2):292-298.
    [182]Davis RJ.Signal transduction by the JNK group of MAP kinases[J]. Cell,2000,103(2):239-252.
    [183]邱晓晓,戴雍月,宋张娟,等.SP600125对大鼠肺缺血/再灌注损伤的保护作用及机制[J].中国应用生理学杂志,2012,28(03):255-258+292.
    [184]Suzuki A,Ghayor C,Guicheux J,et al.Enhanced expression of the inorganic phosphatetransporter Pit-1is involved in BMP-2-induced matrix mineralization in osteoblast-likecells[J].J Bone Miner Res,2006,21(5):674-683.
    [185]Bennett BL,Sasaki DT,Murray BW,et al.SP600125,an anthrapyrazolone inhibitor of JunN-terminal kinase[J].Proc Natl Acad Sci U S A,2001,98(24):13681-13686.
    [186]Tokuda H, Takai S, Hanai Y, et al.Potentiation by platelet-derived growth factor-BB ofFGF-2-stimulated VEGF release in osteoblasts[J]. J Bone Miner Metab,2008,26(4):335-341.
    [187]Matsuguchi T,Chiba N,Bandow K,et al.JNK activity is essential for Atf4expression andlate-stage osteoblast differentiation[J].J Bone Miner,Res2009,24(3):398-410.
    [188]Rey A,Manen D,Rizzoli R,et al.Evidences for a role of p38MAP kinase in thestimulation of alkaline phosphatase and matrix mineralization induced by parathyroidhormone in osteoblastic cells[J].Bone,2007,41(1):59-67.
    [189]Hsu YL,Kuo PL.Diosmetin induces human osteoblastic differentiation through theprotein kinase C/p38and extracellular signal-regulated kinase1/2pathway[J].J Bone MinerRes,2008,23(6):949-960.
    [190]Yamaguchi T,Chattopadhyay N,Kifor O,et al.Activation of p42/44and p38mitogen-activated protein kinases by extracellular calcium-sensing receptor agonists inducesmitogenic responses in the mouse osteoblastic MC3T3-E1cell line[J].Biochem Biophys ResCommun,2000,279(2):363-368.
    [191]Higuchi C,Myoui A,Hashimoto N,et al.Continuous inhibition of MAPK signalingpromotes the early osteoblastic differentiation and mineralization of the extracellularmatrix[J].J Bone Miner Res2002,17(10):1785-1794.
    [192]Huang MS,Morony S,Lu J,et al.Atherogenic phospholipids attenuate osteogenicsignaling by BMP-2and parathyroid hormone in osteoblasts[J].J Biol Chem,2007,282(29):21237-21243.
    [193]Hu Y,Chan E,Wang SX,et al.Activation of p38mitogen-activated protein kinase isrequired for osteoblast differentiation[J].Endocrinology,2003,144(5):2068-2074.
    [194]Greenblatt MB,Shim JH,Zou W,et al.The p38MAPK pathway is essential forskeletogenesis and bone homeostasis in mice[J].J Clin Invest,2010,120(7):2457-2473.
    [195]Ge C,Yang Q,Zhao G,et al.Interactions between extracellular signal-regulated kinase1/2and p38MAP kinase pathways in the control of RUNX2phosphorylation and transcriptionalactivity[J].J Bone Miner Res,2011,27(3):538-551.
    [196]Liao QC,Xiao ZS,Qin YF, et al.Genistein stimulates osteoblastic differentiation via p38MAPK-Cbfa1pathway in bone marrow culture[J]. Acta Pharmacol Sin,2007,28(10):1597-1602.
    [197]Lee CH,Huang YL,Liao JF,Chiou WF.Ugonin K promotes osteoblastic differentiationand mineralization by activation of p38MAPK-and ERK-mediated expression of Runx2andosterix[J].Eur J Pharmacol,2011,668(3):383-389.
    [198]Chiou WF,Lee CH,Liao JF,et al.8-Prenylkaempferol accelerates osteoblast maturationthrough bone morphogenetic protein-2/p38pathway to activate Runx2transcription[J]. LifeSci,2011,88(7-8):335-342.
    [199]Lane NE,Kelman A.A review of anabolic therapies for osteoporosis[J].Arthritis ResTher,2003,5(5):214-222.
    [200]刘敏,周后德,何玉玲,等.MC3T3E1细胞分化过程中Cbfα1及相关基因的表达[J].中国骨质疏松杂志,2006,12(02):118-123.
    [201]刘婷,曹春雨,郝然,等.黔岭淫羊藿总黄酮类成分对hFOB1.19人SV40转染成骨细胞活性的影响[J].中国实验方剂学杂志,2013,19(02):267-270.
    [202]Takeuchi Y,Suzawa M,et al.Differentiation and transforming growth factor-beta receptordown-regulation by collagen-alpha2beta1integrin interaction is mediated by focal adhesionkinase and its downstream signals in murine osteoblastic cells[J].J BiolChem,1997,272(46):29309-29316.
    [203]王艳杰,任艳玲,宋囡,等.左归丸对去卵巢大鼠股骨中Ⅰ型胶原mRNA表达的影响[J].中国老年学杂志,2012,32(23):5170-5172.
    [204]Jadlowiec J,Koch H,Zhang X,et al.Phosphophoryn regulates the gene expression anddifferentiation of NIH3T3,MC3T3-E1,and human mesenchymal stem cells via theintegrin/MAPK signaling pathway[J].J Biol Chem,2004,279(51):53323-53330.
    [205]Mehrotra M,Krane SM,Walters K,et al.Differential regulation of platelet-derived growthfactor stimulated migration and proliferation in osteoblastic cells[J]. J CellBiochem,2004,93(4):741-752.
    [206]Liu H,Liu Y,Viggeswarapu M,et al.Activation of c-Jun NH(2)-terminal kinase1increases cellular responsiveness to BMP-2and decreases binding of inhibitory Smad6to thetype1BMP receptor[J].J Bone Miner Res,2011,26(5):1122-1132.
    [207]Kanno T,Takahashi T,Tsujisawa T,et al.Mechanical stress-mediated Runx2activation isdependent on Ras/ERK1/2MAPK signaling in osteoblasts[J]. J CellBiochem,2007,101(5):1266-1277.
    [208]Guicheux J,Lemonnier J,Ghayor C,et al.Activation of p38mitogen-activated proteinkinase and c-Jun-NH2-terminal kinase by BMP-2and their implication in the stimulation ofosteoblastic cell differentiation[J]. J Bone Miner Res,2003,18(11):2060-2068.
    [209]Costessi A,Pines A,D'Andrea P,et al.Extracellular nucleotides activate Runx2in theosteoblast-like HOBIT cell line: a possible molecular link between mechanical stress andosteoblasts' response[J].Bone,2005,36(3):418-432.
    [210]Fan X,Rahnert JA,Murphy TC,et al.Response to mechanical strain in an immortalizedpre-osteoblast cell is dependent on ERK1/2[J].J Cell Physiol,2006,207(2):454-460.
    [211]Xiao G,Jiang D,Thomas P,et al.MAPK pathways activate and phosphorylate theosteoblast-specific transcription factor,Cbfa1[J].J Biol Chem,2000,275(6):4453-4459.
    [212]Wang L,Li JY,Zhang XZ,et al.Involvement of p38MAPK/NF-kappaB signalingpathways in osteoblasts differentiation in response to mechanical stretch[J].Ann BiomedEng,2012,40(9):1884-1894.
    [213]Zayzafoon M,Botolin S,McCabe LR.P38and activating transcription factor-2involvement in osteoblast osmotic response to elevated extracellular glucose.J BiolChem,2002,277(40):37212-37218.
    [214]Jennings JC,Mohan S.Heterogeneity of latent transforming growth factor-beta isolatedfrom bone matrix proteins[J].Endocrinology,1990,126(2):1014-1021.
    [215]Centrella M,Horowitz MC,Wozney JM,et al.Transforming growth factor-beta genefamily members and bone[J].Endocr Rev,1994,15(1):27-39.
    [216]Rodan GA.Bone homeostasis[J].Proc Natl Acad Sci U S A,1998,95(23):13361-13362.
    [217]Beck LS,Amento EP,Xu Y,et al.TGF-beta1induces bone closure of skull defects:temporal dynamics of bone formation in defects exposed to rhTGF-beta1[J].J Bone MinerRes,1993,8(6):753-761.
    [218]雪原,王沛,齐清会,等.淫羊藿甙对成骨细胞Smad4mRNA作用的实验研究[J].中华骨科杂志,2005,25(02):58-62.
    [219]任艳玲,李娅玲,吕海波,等.左归丸对去卵巢骨质疏松大鼠肾脏TGF-β_1/Smad4mRNA表达的影响[J].中国实验方剂学杂志,2012,18(10):190-194.
    [220]Massague J,Chen YG.Controlling TGF-beta signaling[J]. Genes Dev,2000,14(6):627-644.
    [221]Drevelle O,Daviau A,Lauzon MA,et al.Effect of BMP-2and/or BMP-9onpreosteoblasts attached to polycaprolactone functionalized by adhesive peptides derived frombone sialoprotein[J].Biomaterials,2013,34(4):1051-1062.
    [222]Yano H,Hamanaka R,Nakamura M,et al.Smad,but not MAPK,pathway mediates theexpression of type I collagen in radiation induced fibrosis[J].Biochem Biophys Res,Commun2012,418(3):457-463.
    [223]胡振宇.TGF-β1促进rhBMP-2对成骨前体细胞增殖分化的观察[J].口腔颌面外科杂志,2012,22(01):24-26.
    [224]Ghayor C,Rey A,Caverzasio J.Prostaglandin-dependent activation of ERK mediates cellproliferation induced by transforming growth factor beta in mouse osteoblasticcells[J].Bone,2005,36(1):93-100.
    [225]潘秋辉,李益广,杨松海,等.骨形态发生蛋白2通过Smad途径上调Osterix的表达[J].中国生物化学与分子生物学报,2008,24(01):40-45.
    [226]Iu MF,Kaji H,Sowa H,et al.Dexamethasone suppresses Smad3pathway in osteoblasticcells[J].J Endocrinol,2005,185(1):131-138.
    [227]赵丹,王箭,罗进勇,等.骨形态发生蛋白9通过p38激酶途径调控间充质干细胞C3H10T1/2成骨分化[J].生物化学与生物物理进展,2011,38(11):1001-1010.
    [228]Wang Y,Bernard DJ.Activin A induction of murine and ovine follicle-stimulatinghormone beta transcription is SMAD-dependent and TAK1(MAP3K7)/p38MAPK-independent in gonadotrope-like cells[J].Cell Signal,2012,24(8):1632-1640.
    [229]Yuan Y,Wu ZJ,Yao HY,et al.Activation of p38mitogen-activated protein kinasecontribute to BMP4-induced alkaline phosphatase expression in MC3T3-E1preosteoblast[J].Chin Med J (Engl),2006,119(4):324-327.
    [230]Byers BA,Pavlath GK,Murphy TJ,et al.Cell-type-dependent up-regulation of in vitromineralization after overexpression of the osteoblast-specific transcription factorRunx2/Cbfal[J].J Bone Miner Res,2002,17(11):1931-1944.
    [231]Fan D,Chen Z,Wang D,et al.Osterix is a key target for mechanical signals in humanthoracic ligament flavum cells[J].J Cell Physiol,2007,211(3):577-584.
    [232]Komori T,Yagi H,Nomura S,et al.Targeted disruption of Cbfa1results in a completelack of bone formation owing to maturational arrest of osteoblasts[J]. Cell,1997,89(5):755-764.
    [233]Ma GK,Chiu R,Huang Z,et al.Polymethylmethacrylate particle exposure causes changesin p38MAPK and TGF-beta signaling in differentiating MC3T3-E1cells[J].J Biomed MaterRes A,2010,94(1):234-240.
    [234]Lee KS,Hong SH,Bae SC.Both the Smad and p38MAPK pathways play a crucial role inRunx2expression following induction by transforming growth factor-beta and bonemorphogenetic protein[J].Oncogene,2002,21(47):7156-7163.
    [235]Li J,Tsuji K,Komori T,et al.Smad2overexpression enhances Smad4gene expression andsuppresses CBFA1gene expression in osteoblastic osteosarcoma ROS17/2.8cells andprimary rat calvaria cells[J].J Biol Chem,1998,273(47):31009-31015.
    [236]Alliston T, Choy L, Ducy P,et al.TGF-beta-induced repression of CBFA1by Smad3decreases cbfa1and osteocalcin expression and inhibits osteoblast differentiation[J]. EmboJ,2001,20(9):2254-2272.
    [237]Lee YS,Choi EM.Costunolide stimulates the function of osteoblastic MC3T3-E1cells.Int Immunopharmacol,2011,11(6):712-718.
    [238]Sowa H,Kaji H,Yamaguchi T,et al.Activations of ERK1/2and JNK by transforminggrowth factor beta negatively regulate Smad3-induced alkaline phosphatase activity andmineralization in mouse osteoblastic cells[J].J Biol Chem,2002,277(39):36024-36031.

© 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