切换至 "中华医学电子期刊资源库"

中华老年骨科与康复电子杂志 ›› 2018, Vol. 04 ›› Issue (06) : 341 -345. doi: 10.3877/cma.j.issn.2096-0263.2018.06.005

所属专题: 文献

基础研究

转染音猬因子对骨髓间充质干细胞成骨分化的影响
贾祎佳1, 孙吉平1, 刘强1, 郝海虎1, 朱剑1, 吴斗1,()   
  1. 1. 030032 太原,山西医学科学院 山西大医院骨科
  • 收稿日期:2018-06-19 出版日期:2018-12-05
  • 通信作者: 吴斗
  • 基金资助:
    山西省自然科学基金(2013011057-4)

The effects of bone marrow stromal cells transferred with lentivirat—mediated Sonic Hedgehog (Shh) gene in osteoporosis

Yijia Jia1, Jiping Sun1, Qiang Liu1, Haihu Hao1, Jian Zhu1, Dou Wu1,()   

  1. 1. Department of Orthopaedics, Shanxi Academy of Medical Sciences, Shanxi Dayi Hospital, Taiyuan 030032, China
  • Received:2018-06-19 Published:2018-12-05
  • Corresponding author: Dou Wu
  • About author:
    Corresponding author: Wu Dou, Email:
引用本文:

贾祎佳, 孙吉平, 刘强, 郝海虎, 朱剑, 吴斗. 转染音猬因子对骨髓间充质干细胞成骨分化的影响[J]. 中华老年骨科与康复电子杂志, 2018, 04(06): 341-345.

Yijia Jia, Jiping Sun, Qiang Liu, Haihu Hao, Jian Zhu, Dou Wu. The effects of bone marrow stromal cells transferred with lentivirat—mediated Sonic Hedgehog (Shh) gene in osteoporosis[J]. Chinese Journal of Geriatric Orthopaedics and Rehabilitation(Electronic Edition), 2018, 04(06): 341-345.

目的

建立能够持续稳定表达音猬因子(Shh)的骨髓基质干细胞(BMSCs),观察其成骨分化能力,为体内治疗骨质疏松提供可行性依据。

方法

使用Gateway Technology构建pDown-DsRed-Shh,贴壁培养法获取SD大鼠BMSCs,慢病毒转染法将pDown-DsRed-Shh、pDown-DsRed报告质粒转染进BMSCs,分为DsRed-BMSCs组(A组)和Shh-DsRed-BMSCs组(B组),荧光显微镜下观察DsRed表达,判断转染效率。48 h后使用RT-PCR法和Western印迹法检测Shh基因的表达情况,7 d后检测碱性磷酸酶(ALP)活性,28 d后茜素红染色检测骨髓间充质干细胞的成骨情况。

结果

慢病毒转染24 h后Shh-DsRed-BMSCs的转染率约为90%。同A组相比,B组能持续稳定高水平表达Shh mRNA和蛋白,同A组相比,B组的ALP活性更强,差异具有统计学意义(t=17.665,P<0.05),B组的茜素红染色表达情况明显高于A组。

结论

慢病毒转染音猬因子的骨髓间充值干细胞可以导致Shh的持续稳定高水平表达,并具有很高的成骨细胞分化能力,为骨质疏松的体内治疗提供了理论基础。

Objective

To observe osteogenous differentiation of modified bone marrow stromal cells (BMSCs) which can express Sonic Hedgehog (Shh) stably and provide the treatment of osteoporosis in vivo feasibility.

Methods

BMSCs were infected with lentiviruses using a lentiviral vector containing the DsRed or the Shh-DsRed gene and then divided into Shh-DsRed- BMSCs group (group A) and DsRed- BMSCs group (group B). The transfection efficiency were evaluated by fluorescence microscopy using DsRed expression. The expression of Shh was tested by RT-PCR and Western-blot analysis after 48 hours. The osteogenous differentiation of BMSCs were analysed by ALP testing after 7 days, and tested by alizarin red staining after 28 days.

Results

The transfeetion efficiency was about 90% after 24 hours in Shh-DsRed-BMSCs. Compared with the group B, the Shh-DsRed-BMSCs can stably secrete Shh mRNA and protein. The ALP activity in Shh-DsRed-BMSCs group was significantly greater than those in DsRed-BMSCs. The alizarin red staining expression in group A was markedly higher than those in group B.

Conclusion

Shh-DsRed-BMSCs can long-term and stably secrete high levels of Shh, has a high ability of osteoblast differentiation and was then the basis for the subsequent treatment for osteoporosis in vivo.

图4~5 慢病毒载体转染BMSCs 5 d后。图4 DsRed-BMSCs(×100);图5 Shh-DsRed-BMSCs(×100)
图6 Shh基因的mRNA的表达
图7 Shh-BMSCs中Shh蛋白的表达情况
图8~9 两组成骨诱导分化28 d后茜素红染色情况。图8 A组茜素红染色情况(×100);图9 B组素红染色情况(×100)
图10 两组成骨诱导分化28 d后茜素红染色吸光度值的比较
图11 两组ALP活性定量检测比较
1
Migliaccio S, Brama M, Malavolta N. Management of glucocorticoids-inducedosteoporosis: role ofteriparatide[J]. Ther Clin Risk Manag, 2009, 5(2):305-310.
2
Adler RA. Glucocorticoid-induced osteoporosis:management update[J]. Curr Osteoporos Rep, 2010, 8(1):10-14.
3
Ma XL, Zhang XE, Jia YF, et al. Dexamethasone induces osteogenesis via regulation of hedgehog signalling molecules in rat mesenchymal stem cells[J]. Int Orthop, 2013, 37(7):1399-1404.
4
Das S, Crockett JC. Osteoporosis - a current view of pharmacological prevention and treatment[J]. Drug Des Devel Ther, 2013, 7:435-447.
5
原林,王军,王春雷,等.人体内新的功能系统--支持储备及自体监控系统新学说[J].科技导报, 2006, 24(6):85-89.
6
Bai Y, Yuan L, Soh KS, et al. Possible applications for fascial anatomy and fasciaology in traditional Chinese medicine[J]. J Acupunct Meridian Stud, 2010, 3(2):125-132.
7
张晓玲,戴尅戎,汤亭亭,等.未分化骨髓间充质干细胞的免疫学特性研究[J].中华实验外科杂志, 2006, 23(2):135-137.
8
Lien CY, Ho KC, Lee OK, et al. Restoration of bone mass and strength in Glucocorticoid-Treated mice by systemic transplantation of CXCR4 and cbfa-1 Co-Expressing mesenchymal stem cells[J]. J Bone Miner Res, 2009, 24(5):837-848.
9
Soltanoff CS, Yang S, Chen W, et al. Transient inhibitiion of the hedgehog pathway in young mice causes permanent defects in bone structure[J]. Crit Rev Eukaryot Gene Expr, 2009, 19(1):1-46.
10
Blank U, Karlsson G, Karlsson S. Signaling pathways governing stem-cell fate[J]. Blood, 2008, 111(2):492-503.
11
Foppiano S, Hu D, Marcucio RS, et al. Signaling by bone morphogenetic proteins directs formation of an ectodermal signaling center that regulates craniofacial development[J]. Dev Biol, 2007, 312(1):103-114.
12
Spinella-Jaegle S, Rawadi G, Kawai S, et al. Sonic hedgehog increases the commitment of pluripotent mesenchymal cells into the osteoblastic lineage and abolishes adipocytic differentiation[J]. J Cell Sci, 2001, 114(11):2085-2094.
13
Ingram WJ, Wicking CA, Grimmond SM, et al. Novel genes regulated by Sonic Hedgehog in pluripotent mesenchymal cells[J]. Oncogene, 2002, 21(53):8196-8205.
14
Shefer S, Salen G, Batta AK, et al. Markedly inhibited 7-dehydrocholesterol delta 7-reductase activity in liver microsomes from Smith-Lemli-Opitz homozygotes[J]. J Clin Invest, 1995, 96(4):1779-1785.
15
Beloti MM, Bellesini LS, Rosa AL. Purmorphamine enhances osteogenic activity of human osteoblasts derived from bone marrow mesenchymal cells[J]. Cell Biol Int, 2005, 29(7):537-541.
16
Cai J, Huang Y, Chen X, et al. [Regulation of sonic hedgehog on vascularendothelial growth factor,basic fibroblast growth factor expression and secretion in bone marrow mesenchymal stem cells][J]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi, 2012, 26(1):112-116.
[1] 杨霁, 黄顺梅, 王安鸽, 吴月, 杨云梅. 杭州地区老年人群中肌少症患病情况及其与骨质疏松症的相关性分析[J]. 中华危重症医学杂志(电子版), 2023, 16(03): 207-210.
[2] 陆宜仙, 张震涛, 夏德萌, 王家林. 巨噬细胞极化在骨质疏松中调控作用及机制的研究进展[J]. 中华损伤与修复杂志(电子版), 2023, 18(06): 538-541.
[3] 陈跃圻, 罗睿, 向涵, 余泳妍, 余挺. 骨质疏松症与牙周炎的因果关系:一项两样本孟德尔随机化研究[J]. 中华口腔医学研究杂志(电子版), 2023, 17(04): 292-298.
[4] 唐英俊, 李华娟, 王赛妮, 徐旺, 刘峰, 李羲, 郝新宝, 黄华萍. 人脐带间充质干细胞治疗COPD小鼠及机制分析[J]. 中华肺部疾病杂志(电子版), 2023, 16(04): 476-480.
[5] 李晔, 何洁, 胡锦秀, 王金祥, 田川, 潘杭, 陈梦蝶, 赵晓娟, 叶丽, 张敏, 潘兴华. 高活性间充质干细胞干预猕猴卵巢衰老的研究[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 210-219.
[6] 龙慧玲, 林蜜, 邵婷. 三维球体间充质干细胞培养技术的研究进展及其应用[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 229-234.
[7] 刘文慧, 吴涛, 张曦. 间充质干细胞联合血小板生成素受体激动剂在异基因造血干细胞移植后血小板恢复中的研究进展[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 242-246.
[8] 王红敏, 谢云波, 王彦虎, 王福生. 间充质干细胞治疗新冠病毒感染的临床研究进展[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 247-256.
[9] 秦富豪, 郑正, 江滨. 间充质干细胞在克罗恩病肛瘘治疗中的研究进展[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(03): 172-177.
[10] 袁久莉, 刘丹, 李林藜, 刘晋宇. 毛囊间充质干细胞的基础研究及临床应用[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(03): 189-192.
[11] 杨蕴钊, 周诚, 石美涵, 赵静, 白雪源. 人羊水间充质干细胞对膜性肾病大鼠的治疗作用[J]. 中华肾病研究电子杂志, 2023, 12(04): 181-186.
[12] 宋艳琪, 任雪景, 王文娟, 韩秋霞, 续玥, 庄凯婷, 肖拓, 蔡广研. 间充质干细胞对顺铂诱导的小鼠急性肾损伤中细胞铁死亡的作用[J]. 中华肾病研究电子杂志, 2023, 12(04): 187-193.
[13] 陈客宏. 干细胞外泌体防治腹膜透析腹膜纤维化新技术研究[J]. 中华肾病研究电子杂志, 2023, 12(03): 180-180.
[14] 张茜, 刘叶青, 康雪莹, 孙兵兵, 刘岩, 胡丽叶, 周亚茹. 血清铁蛋白与绝经后骨质疏松症的相关性分析[J]. 中华老年骨科与康复电子杂志, 2023, 09(03): 166-171.
[15] 梁宇同, 丁旭, 马国慧, 黄艳红. 间充质干细胞在宫腔粘连治疗中的研究进展[J]. 中华临床医师杂志(电子版), 2023, 17(05): 596-599.
阅读次数
全文


摘要