转染TIMP-1-shRNA基因的骨髓间充质干细胞治疗大鼠缺血性心肌病的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景:动物实验和初期的临床研究表明干细胞静脉途径移植后,干细胞归巢到缺血心肌组织,通过多种机制改善心功能,从而为心肌梗死的治疗开辟了一条崭新的途径。但干细胞在归巢过程中的大量缺失影响了干细胞归巢的治疗效果。一些研究显示,敲除MMP-2、MT1-MMP或TIMP-2基因使干细胞侵袭能力受损,而沉默的TIMP -1能够增强干细胞迁移能力。在体内外的实验中均证实TIMP-1基因在细胞的迁移过程中起阻碍作用,本试验利用慢病毒载体转染TIMP-1-shRNA基因的骨髓间充质干细胞,来研究沉默TIMP-1基因的骨髓干细胞能否在体内外提高干细胞的侵袭能力,增加干细胞归巢受损心肌的数量,并进一步探讨干细胞治疗缺血性心肌病的机理。
     第一部分携带TIMP-1-shRNA基因的慢病毒表达载体的构建
     目的:设计合成3对以骨髓间充质干细胞TIMP-1基因不同位点为靶点的短发夹状RNA(shRNA),构建携带沉默TIMP-1基因不同位点的3组慢病毒表达载体质粒,并对经鉴定纯化的载体质粒进行包装、滴度测定。
     方法:设计合成3对针对TIMP-1基因的特异性shRNA片段,退火后与经Hpa I和Xho 1双酶切的Pll3.7载体连接,扩增后提取质粒,经双酶切鉴定和测序后,将测序正确的载体质粒和慢病毒包装质粒共转染293T细胞,收集、纯化和浓缩富含慢病毒颗粒的细胞上清液。
     结果:双酶切鉴定和测序结果均表明3组重组的RNA干扰慢病毒载体均有一段与我们设计合成的靶向链完全一致。?
     结论:成功地合成了3组沉默大鼠TIMP-1基因的寡核甘酸序列,并构建了携带shRNA-TIMP-1基因的慢病毒表达载体质粒,经293T细胞包装生产出高滴度的重组慢病毒。
     第二部分使用慢病毒包装系统建立沉默TIMP-1基因的稳转骨髓间充质干细胞
     目的:使用慢病毒包装系统转染3组骨髓间充质干细胞株,利用RNA干扰技术沉默TIMP-1基因的不同靶点。在构建好的转染不同TIMP-1-shRNA的3组细胞株中筛选出沉默TIMP-1基因效率最高的细胞株。
     方法:使用慢病毒包装系统建立沉默TIMP-1基因不同位点的3组骨髓间充质干细胞株(SH1、SH2、SH3组),同时设立转染空载体组和阴性对照组,观察各组细胞转染前后形态和荧光的表达。再抽提蛋白,应用Western blot技术检测TIMP-1蛋白的表达。RT-PCR技术检测各组细胞的TIMP-1基因的mRNA水平。
     结果:慢病毒转染MSCs 5天后,各组均可检测到强荧光出现,转染前后细胞形态无改变。Western blot免疫印迹技术表明在5个实验组中,SH2组TIMP-1蛋白表达最低; RT-PCR技术检测结果表明转染1周后SH2组mRNA水平下降最大。
     结论:使用慢病毒包装系统成功建立沉默沉默TIMP-1基因的3组骨髓间充质干细胞株,并在构建好的沉默不同shRNA-TIMP-1基因位点的3组细胞株中成功筛选出沉默TIMP-1基因效率最高的细胞株。
     第三部分大鼠骨髓间充质干细胞(MSCs)的分离、培养和心梗模型的建立
     目的:从大鼠的骨髓中分离出具有多向分化潜能的骨髓间充质干细胞(Mesenchymal Stem Cells,MSCs)以及通过开胸结扎前降支建立大鼠缺血性心肌病的动物模型。
     目的:建立大鼠骨髓间充质干细胞体外培养方法,为干细胞归巢的实验研究提供细胞材料。
     方法:大鼠的骨髓MSCs分离纯化后,流式细胞仪检测细胞表面标记。
     结果:体外培养的原代MSCs 10~14d达到融合,细胞周期显示89.43%的细胞处于G0/G1期。超过90%大鼠骨髓间充质干细胞性表达CD29、CD90和CD44;但不表达造血干细胞表面标志CD34。
     结论:利用MSCs贴壁特性,在体外条件下成功分离、培养和扩增了大鼠骨髓MSCs,可用于干细胞的归巢研究。
     第二节大鼠心肌梗死模型的建立
     目的:经开胸结扎大鼠冠状动脉前降支(LAD),建立大鼠心肌梗死的模型。
     方法: 24只大鼠开胸结扎大鼠冠状动脉前降支,致前室壁梗死。结扎成功标志为左心室前壁及心尖周围心肌组织颜色晦暗或青紫和运动减弱。建模前后观察心电图变化,建模前和建模4周后行心脏超声、血液动力学检测,4周后行组织学检查。
     结果:与对照组相比,心梗(MI)组左室舒张未内径(LVEDd)增大(P<0.05),射血分数(EF)下降(P<0.05);左室收缩压(LVSP)、、左室内压最大上升和下降速率(±dp/dt )显著降低(P<0.01);而左室舒张末压(LVEDP)显著增加(P<0.01)。组织学检测证实MI组前室壁、室间隔形成了梗死灶。
     结论:这种模型接近临床病理生理过程,稳定可靠。
     第四部分转染TIMP-1-shRNA的MSCs和MSCs体外侵袭能力比较及其机制的研究
     目的:检测对比转染慢病毒空载体细胞、骨髓间充质干细胞和转染沉默TIMP-1基因细胞的体外趋化侵袭能力,同时检测细胞培养上清液的明胶酶活性以及TIMP-1和MMP-9的表达,研究影响细胞体外侵袭能力的因素并探讨其机制。方法:使用Costar穿膜小室系统研究转染慢病毒空载体细胞、P2代骨髓间充质
     干细胞和转染沉默TIMP-1基因细胞的体外趋化侵袭能力。检测不同细胞株培养上清液中明胶酶的活性及TIMP-1、MMP-9蛋白含量。
     第一节大鼠骨髓间充质干细胞(MSCs)的分离、培养及鉴定
     结果:与转染慢病毒空载体细胞、P2代骨髓间充质干细胞相比,敲除TIMP-1基因的MSCs株体外侵袭能力提高,细胞培养上清液中明胶酶活性增强,细胞培养上清液TIMP-1含量下降,而MMP-9含量升高(P<0.05)。
     结论:敲除MSCs的TIMP-1基因能够增加细胞培养上清液中MMP-9的活性和表达,提高了MSCs的体外侵袭能力。
     第五部分大鼠骨髓MSCs静脉输入治疗缺血性心肌病的实验研究
     目的:在大鼠的心梗模型平台上,研究输入转染TIMP-1-siRNA的MSCs能否在体内较MSCs、转染慢病毒空载体的MSCs更显著地改善缺血性心肌病的心脏功能,来探讨MSCs和基因联合应用治疗缺血性心肌病的可行性,以及研究干细胞归巢治疗缺血性心肌病的机制。
     方法:取1只SD大鼠骨髓,采用细胞差异贴壁筛选法分离纯化、体外扩增。利用慢病毒载体建立转染空载体的MSCs、正常传代的MSCs和转染shRNA-TIMP-1基因的细胞株。再选用SD大鼠(n=71),经开胸结扎大鼠前降支建立心梗模型,建模成功后存活56只大鼠随机分入4组:空白组(A组,n=14)、MSCs组(B组,n=14)、GFP-MSCs组(C组,n=14)和TIMP-1-KD-MSCs组(D组,n=14)。1周后经静脉途径植入细胞悬液。
     建模前后和细胞静脉途径移植4周后行心脏超声心动图检测,评估心功能。细胞静脉途径移植3天后,随机选取4只处死,TUNEL检测心肌细胞凋亡率。4周后,对比各组心肌梗死面积,评估梗死区和梗死周边区的有血流灌注的毛细血管的数量;另外,采用ELISA法检测血清TGF-β1在建模前和干细胞移植治疗心梗前后的变化。最后,流式细胞仪检测大鼠骨髓MSCs归巢到心肌组织的数量。
     结果:心超示:与建模前对比,梗死心脏的左室舒张末径(LVEDd)增加,收缩能力明显下降。干细胞移植后,B、C、D组的心功能较移植前有改善(P<0.05),D组的心功能改善优于其他3组(P<0.05)。移植的MSCs能防止梗死后的心室扩张,增加心肌收缩能力;TUNEL检测显示:MSCs组和GFP-MSCs组梗死区心肌细胞的凋亡率无显著差异(P>0.05),但低于空白组,高于TIMP-1-KD组(P<0.05)。TIMP-1-KD组、MSCs组和GFP-MSCs组心肌梗死面积与空白组对比明显缩小(P<0.05);TIMP-1-KD组心肌梗死面积又小于MSCs组和GFP-MSCs组。(P<0.05)。与空白组相比,B、C、D组梗死区的毛细血管密度明显增加(P<0.05)。D组的增加比B、C组显著(P<0.05)。血清TGF-β1的水平在梗死后有所增加,在细胞移植后逐渐下降,D组TGF-β1的水平下降最明显(P<0.05)。流式细胞仪检测证实D组MSCs归巢心肌组织的数量显著大于其他三组(P<0.05)。
     结论:MSCs能在宿主心肌组织中存活,通过减少梗死后心肌细胞凋亡数量、抑制收缩功能异常而改善左室功能;这种改善可能是MSCs通过心肌再生、血管再生和细胞外基质增生引起的。一些细胞因子在其中起非常重要的作用。敲除TIMP-1增加了MMP-9的活性,促进了MMCs侵袭能力,增加了MMCs归巢缺血心肌的数量,使参与心肌修复的干细胞的绝对数量增加,从而可更加显著改善心脏功能。
     沉默MSCs的TIMP-1基因,能够促进MSCs静脉移植后归巢损伤心肌的数量是治疗缺血性心肌病的一种有效的方法。
Background: Animal experiments and early clinical studies have shown that intravenous administration of adult bone marrow-derived Mesenchymal stem cells (MSCs) could home to the ischemic myocardial tissue, and improve cardiac function through a variety of mechanisms, thus intravenously administered MSCs may be a new therapeutic strategy for the treatment of myocardial infarction. However, a loss of a large number of stem cells in the process of homing affected stem cell homing therapeutic effect. The molecular mechanisms, however, that control MSC homing which require invasion through extracellular matrix (ECM) barriers are almost unknown. Matrix metalloproteinases (MMPs) were believed to play a pivotal role in invasion behavior of stem cells. Some studies by RNA interference revealed that gene knock-down of MMP-2, MT1-MMP, or TIMP-2 substantially impaired MSC invasion, whereas silencing of TIMP-1 enhanced cell migration. Lentiviral vectors provide an efficient means for TIMP-1-shRNA delivery into MSCs. Accordingly, we investigated whether intravenously transplanted TIMP-1-silencing MSCs can improve the invasion and homing capacity in vivo and in vitro, increase the number of homing MSCs, and further explore stem cell therapy for ischemic cardiomyopathy mechanism.
     PartⅠConstruction of lentiviral expression vector of TIMP-1-shRNA gene
     Objective: Designed short hairpin RNA (shRNA), targeted to silence TIMP-1 gene of bone marrow-derived mesenchymal stem cells, constructed lentiviral expression vector carrying TIMP-1-shRNA gene, then dentified and purified packaged plasmid.
     Methods: Designed and synthesized three pairs for the TIMP-1 gene-specific oligonucleotide, annealed and connected with Pll3.7 vector that was digested by Hpa I and Xho1 enzyme. Plasmids were amplified, extracted, identified by double enzyme digestion and then sequenced. Vectors that have the correct sequence and the lentiviral packaging plasmid were co-transfected 293T cells. At last, collect, purify and concentrate the virus particles which are rich in cell supernatant.
     Results: Double enzyme digestion and sequencing results showed that three groups of recombinant lentiviral vector of shRNA exactly have a fragment and were consistent with the target chain that were designed and synthesized by our.
     Conclusion: Successfully synthesized oligonucleotide sequences to silence the rat TIMP-1 gene, and constructed carrying shRNA-TIMP-1 gene lentiviral expressing vector, then packaged by the 293T cells to produce high titer lentivirus particles.
     PartⅡUsing an HIV-based lentiviral vector to transfect Mesenchymal stem cells to knock-out TIMP-1 gene stably
     Objective: Using an HIV-based lentiviral vector to transfect Mesenchymal stem cells to knock-out TIMP-1 gene stably in vitro,then selecting out cell which have the highest efficiency of gene silencing in three cell lines
     Methods: Three lentiviral expression vector of carrying different shRNA-TIMP-1 gene was transfected into MSCs (SH1, SH2, SH3 group), and set up an empty vector transfection group and negative control group. The morphology of cells was observed in each group before and after cell transfection. After five day culturing, GFP (green) was detected in the fluorescence microscope. Protein was extracted, Western blot was carried out to detect TIMP-1 protein expression levels in each group cells and RT-PCR was carried out to detect TIMP-1 mRNA levels in each group cells,and GAPDH was used as a housekeeping gene.
     Results: On the lentivirus-transfected MSCs 5th days, strong fluorescence can be detected in each groups, cell morphology did not change before and after transfection. Western blot analysis showed that SH2 group expressed the lowest TIMP-1 Protein levels; RT-PCR results showed that the largest decline in the levels of TIMP-1 mRNA expression could be detected distinctly in SH2 group.
     Conclusion: Lentiviral vector carrying shRNA-TIMP-1 gene could make MSCs stably to knockout TIMP-1 gene.Cell lines of SH2 group has the highest efficiency of gene silencing in three cell lines
     .PartⅢ
     Section A Culture of Mesenchymal Stem Cells from Rat Bone Marrow
     Objective: To establish a method for isolation and cultivation of mesenchymal stem cells (MSCs) from rat bone marrow and to study their phenotypical properties to offerg an experimental foundation for their further differentiation and actual application.
     Methods: Marrow was isolated from the long bonesof rat by centrifugation and plated in tissue-culture dishes. The proliferation and growth characteristics were observed in primary and passage culture. Cell cycle was analyzed by measuring DNA content with flowcytometer. Epitope analyses were detected by flow cytometry.
     Results: The adherent, fibroblast-like cells were confluent in single layer after plating for 10~14 days. The cell cycle analysis showed that 89.43% of MSCs was in G0/G1 phase. More than 90% of rat bone marrow-derived mesenchymal stem cells expressed CD29, CD90 and CD44, but not CD34 which was hematopoietic stem cell surface marker. Conclusion: Application of MSCs adherent properties in vitro conditions, rat bone marrow MSCs which were successful isolated, cultured and amplificated can be used to study stem cell homing. It is suggesting that the method can provide cell material for cell homing experiment.
     Section B A Model of Myocardial Infarction in Rat
     Objective: Myocardial infarction (MI) was induced by left anterior descending artery (LAD) ligation.
     Methods: MI was induced by ligation of the LAD in 24 rats. Doppler echocardiography was performed to compare myocardial function in two groups of rats. Electrocardiogram (ECG) changes were observed in the operation and postoperative. Pathology was also performed 4 weeks later.
     Results: Coronary artery ligation resulted in anterior and lateral infarction of the left ventricular wall. The immediate changes visible post ligation were immediately pallor of the myocardium. LVEDd (the LV end-diastolic dimension) in MI increased highly compare to control group (control versus MI: 5.68±0.41 mm vs 7.35±0.67 mm, p<0.01), and the ejection fraction (EF) decreased (control versus MI: 88.36±3.83% vs 49.36±6.25%, p<0.001) in the MI group. Histological tests confirmed that coronary artery ligation resulted in anterior and lateral infarction of the left ventricular wall.
     Conclusion: This rat model of MI is reliable, reproducible, and similar to the human condition.
     PartⅣContrast MSCs transfected TIMP-1-shRNA and MSCs in vitro invasive ability and investigate its mechanism in vitro.
     Objective: Contrast MSCs transfected lentiviral empty vector, MSCs and MSCs transfected shRNA-TIMP-1 gene chemotactic invasive ability in vitro, while detected the gelatin activity in cell culture supernatant, as well as TIMP-1 and MMP-9 expression, and investigated its mechanism affecting the cells invasive ability in vitro.
     Methods: Using Costar trans-membrane chamber system to study MSCs transfected lentiviral empty vector, P2 generation MSCs and MSCs transfected shRNA-TIMP-1 gene the chemotactic invasion ability in vitro.After culturing in serum-free medium in 5% CO2 at 37℃for 24 h,harvested culture supernatants, detected gelatinase activity and TIMP-1, MMP-9 levels in culture supernatants of different cell lines.
     Results: Compared with three groups, the invasive ability of MSCs line transfected shRNA-TIMP-1 gene was the highest (P<0.05), gelatin activity of cell culture supernatant also increased, TIMP-1 content of cell culture supernatant decreased, while MMP-9 expression levels increased(P<0.05).
     Conclusion: MSCs line transfected shRNA-TIMP-1 gene can increase the MMP-9 activity and expression in cell culture supernatant, and increase invasion ability of MSCs. The improvement of MSCs invasion ability in vitro was associated with knockout TIMP-1 gene.
     PartⅤEffect of Intravenously Injected Marrow Mesenchymal Cells in Rat Infarcted Myocardium
     Objective: To investigate whether intravenously injected MSCs improved cardiac function in rat myocardial infarction model, and explore whether the combined application of MSCs and shRNA-TIMP-1 gene in the treatment of MI is feasible, as well as investigate stem cell homing mechanism for the treatment of MI.
     Methods: A commercially available self-inactivating lentiviral vector has been modified for the delivery of TIMP-1-siRNA into MSCs. MSCs from healthy rat were isolated, cultured and transduced with a lentiviral vector containing either TIMP-1-siRNA and green fluorescent protein (GFP; TIMP-1-siRNA /GFP vector) or GFP alone (control vector).
     MI model was induced by occluding the LAD in 71 SD rats (230-385g). After the establishment of MI model, survival 56 rats were randomly divided into 4 groups: control group (A group, n = 14), MSCs group (B group, n = 14), GFP-MSCs group (C group, n = 14), and TIMP-1-KD-MSCs group (D group, n = 14). One week later, intravenously injected MSCs or suspensions were performed.
     The echocardiographic studies were performed to measure cardiac function in the preoperative and postoperative 4 weeks. Intravenously injected MSCs three days later, four rats were killed randomly. TUNEL technology was performed to detect myocardial cell apoptosis. Four weeks later, triphenyltetrazolium chloride (TTC) staining was performed to compare infarct sizes (IS) in each group and assess the number of blood perfusion capillary in the infarct zone and infarct border zone. In addition, ELISA assay were performed to measure serum TGF-β1 content before MI and stem cell transplantation preoperative and postoperative for treatment of myocardial infarction. Finally, the number of MSCs homing to myocardial tissue was measured by flow cytometry.
     Results: Myocardial infarction was defined by echocardiography as any segmental wall motion abnormality. Left ventricular end-diastolic diameter (LVEDd) increase and contractility decreased significantly after MI (P<0.05). After intravenously injected MSCs, the cardiac function in B, C, D group improved compared with control group (P<0.05), the cardiac function improvement in D group was better than the other three groups (P<0.05). MSCs transplantation can prevent post-infarction ventricular dilatation, increased cardiac contractility.
     TUNEL test showed: Apoptotic myocytes of B group and C group were no significantly difference (P>0.05), but lower than the control group, higher than D group (P<0.05). Myocardial infarct size of D group, B group and C group were reduced significantly compared with the control group (P<0.05), Myocardial infarct size in D group was smaller than B group and C groups (P<0.05). Compared with the control group, B, C, D group infarct zone capillary density was significantly increased (P <0.05), the number of blood perfusion capillary of D group in the infarct zone was the most highest in B, C, D group (P <0.05). The serum TGF-β1 levels began to increase in post-infarction and gradually decreased after cells transplantation, the level of TGF-β1 of D group decreased significantly Compared with A,B, C group (P<0.05). The number of MSCs homing to myocardial tissue was measured by flow cytometry confirmed D group was significantly higher than the other three groups (P<0.05).
     Conclusion: MSCs may both migrate and differentiate extensively, might improve the cardiac function by attenuating contractile dysfunction and pathologic thinning in this model of left ventricular wall infarction. This improvement might result from myocardial regeneration and angiogenesis in injured hearts by engrafted cells. Some cytokines maybe play an important role in repairing the ischemic myocardium.
     TIMP-1 gene knockout increased the MMP-9 activity and promoted the invasive ability of MSCs, and increased the number of MSCs homing to ischemic myocardium, so that the quantitations of MSCs that participating in myocardial repair increased, and the therapeutic benefit was more powerful.
     Use of lentiviral vectors for delivery of TIMP-1-shRNA enhanced invasion and homing efficiency of intravenously injected MSCs in rat infarcted myocardium, so the strategies will be an effective approach for the treatment of MI.
引文
1. Abbas-Terki T,Blanco-Bose W,Deglon N,et al.Lentiviral-mediated RNA interferen ce. Hum Gene Ther, 2002, 13:2197-2201.
    2. Stewart SA,Dykxhoorn DM,Palliser D,et al.Lentivirus-delivered stable gene silencing by RNAi in primary cells. RNA, 2003, 9:493-501.
    3. Li MJ, Rossi JJ.Lentiviral vector delivery of recombinant small interfering RNA expression cassettes. Methods Enzymol 2005; 392:218-226.
    4. Rao DD, Vorhies JS, Senzer N, et al.siRNA vs. shRNA: similarities and differences. Adv Drug Deliv Rev, 2009, 61:746-759.
    5. Li M, Rossi JJ.Lentiviral vector delivery of siRNA and shRNA encoding genes into cultured and primary hematopoietic cells. Methods Mol Biol, 2008, 433:287-299.
    6. Chang LJ, Liu X,He J.Lentiviral siRNAs targeting multiple highly conserved RNA sequences of human immunodeficiency virus type 1. Gene Ther, 2005,12: 1133-1144.
    7. Zhang XY, La Russa VF, Bao L,et al.Lentiviral vectors for sustained transgene expression in human bone marrow-derived stromal cells. Mol Ther, 2002; 5:555-565.
    8. Downward J.RNA interference. BMJ, 2004, 328:1245-1248.
    9. Rossi JJ. Expression strategies for short hairpin RNA interference triggers. Hum Gene Ther, 2008, 19:313-317.
    10. Mittal V.Improving the efficiency of RNA interference in mammals. Nat Rev Genet, 2004, 5:355-365.
    1. Shankar P, Manjunath N, Lieberman J.The prospect of silencing disease using RNA interference. JAMA, 2005, 293:1367-1373.
    2. Yano J, Hirabayashi K, Nakagawa S,et al.Antitumor activity of small interfering RNA/cationic liposome complex in mouse models of cancer. Clin Cancer Res, 2004, 10:7721-7726.
    3. Bogdanovich S, Krag TO,Barton ER,et al.Functional improvement of dystrophic muscle by myostatin blockade. Nature, 2002, 420:418-421.
    4. McManus MT, Sharp PA. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet, 2002, 3:737-747.
    5. Elbashir SM, Harborth J, Lendeckel W,et al.Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001; 411:494-498.
    6. Stewart SA,Dykxhoorn DM,Palliser D,et al.Lentivirus-delivered stable gene silencing by RNAi in primary cells. RNA, 2003, 9:493-501.
    7. Sui G,Soohoo C,Affar el B,et al.A DNA vector-based RNAi technology to suppress gene expression in mammalian cells. Proc Natl Acad Sci U S A, 2002, 99:5515- 5520.
    8. Scherr M, Eder M. Gene silencing by small regulatory RNAs in mammalian cells. Cell Cycle, 2007, 6:444-449.
    9. Rubinson DA,Dillon CP,Kwiatkowski AV,et al.A lentivirus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference. Nat Genet, 2003, 33:401-406.
    10. Tiscornia G, Singer O, Verma IM.Design and cloning of lentiviral vectors expressing small interfering RNAs. Nat Protoc, 2006, 1:234-240.
    11. Paddison PJ, Caudy AA, Bernstein E, et al.Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev, 2002,16:948-958.
    1. Schwarz EJ,Alexander GM,Prockop DJ, et al..Multipotential marrow stromal cells transduced to produce L-DOPA: engraftment in a rat model of Parkinson disease. Hum Gene Ther, 1999,10:2539-2549.
    2. Dobson KR, Reading L,Haberey M, et al.Scutt A.Centrifugal isolation of bone marrow from bone: an improved method for the recovery and quantitation of bone marrow osteoprogenitor cells from rat tibiae and femurae. Calcif Tissue Int, 1999, 65:411-413.
    3. Scutt A, Bertram P.Bone marrow cells are targets for the anabolic actions of prostaglandin E2 on bone: induction of a transition from nonadherent to adherent osteoblast precursors. J Bone Miner Res, 1995,10:474-487.
    4. Pittenger MF,Mackay AM,Beck SC,et al.Multilineage potential of adult human mesenchymal stem cells. Science, 1999, 284:143-147.
    5. Tropel P,Noel D,Platet N,et al.Isolation and characterisation of mesenchymal stem cells from adult mouse bone marrow. Exp Cell Res, 2004,295:395-406.
    6. Kitano Y,Radu A,Shaaban A, et al.Selection, enrichment, and culture expansion of murine mesenchymal progenitor cells by retroviral transduction of cycling adherent bone marrow cells. Exp Hematol, 2000,28:1460-1469.
    7. Lisignoli G,Remiddi G,Cattini L,et al.An elevated number of differentiated osteoblast colonies can be obtained from rat bone marrow stromal cells using a gradient isolation procedure. Connect Tissue Res, 2001, 42:49-58.
    8. Barry FP, Boynton RE,Haynesworth S, et al.The monoclonal antibody SH-2, raised against human mesenchymal stem cells, recognizes an epitope on endoglin (CD105). Biochem Biophys Res Commun, 1999,265:134-139.
    9. Vaquero J,Zurita M,Oya S,Santos M.Cell therapy using bone marrow stromal cells in chronic paraplegic rats: systemic or local administration? Neurosci Lett, 2006, 398:129-134.
    10. Flax JD, Aurora S,Yang C,et al.Engraftable human neural stem cells respond to developmental cues, replace neurons, and express foreign genes. Nat Biotechnol, 1998,16:1033-1039.
    11. Thomas T,Gori F,Khosla S,et al.Leptin acts on human marrow stromal cells to enhance differentiation to osteoblasts and to inhibit differentiation to adipocytes. Endocrinology, 1999, 140:1630-1638.
    12. Peister A, Mellad JA, Larson BL,et al.Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood, 2004,103:1662-1668.
    13. Conget PA, Minguell JJ.Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells. J Cell Physiol, 1999,181:67-73.
    14. De Ugarte DA,Alfonso Z,Zuk PA,et al.Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow. Immunol Lett, 2003, 89:267-270.
    15. Foster LJ,Zeemann PA,Li C,et al.Differential expression profiling of membrane proteins by quantitative proteomics in a human mesenchymal stem cell line undergoing osteoblast differentiation. Stem Cells, 2005, 23:1367-1377.
    16. Woodbury D, Schwarz EJ,Prockop DJ, et al.Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res, 2000, 61:364-370.
    17. Li CD, Zhang WY, Li HL,et al.Mesenchymal stem cells derived from human placenta suppress allogeneic umbilical cord blood lymphocyte proliferation. Cell Res, 2005, 15:539-547.
    18. Sun S,Guo Z,Xiao X,et al.Isolation of mouse marrow mesenchymal progenitors by a novel and reliable method. Stem Cells, 2003, 21:527-535.
    19. Sung JH, Yang HM,Park JB,et al.Isolation and characterization of mouse mesenchymal stem cells. Transplant Proc, 2008, 40:2649-2654.
    20. Guo Z,Li H,Li X,et al.In vitro characteristics and in vivo immunosuppressive activity of compact bone-derived murine mesenchymal progenitor cells. Stem Cells, 2006, 24:992-1000.
    21. Fukuda K. Reprogramming of bone marrow mesenchymal stem cells into cardiomyocytes. C R Biol, 2002, 325:1027-1038.
    1. Fishbein MC, Maclean D, Maroko PR.Experimental myocardial infarction in the rat: qualitative and quantitative changes during pathologic evolution. Am J Pathol, 1978, 90:57-70.
    2. Selye H,Bajusz E,Grasso S,Mendell P.Simple techniques for the surgical occlusion of coronary vessels in the rat. Angiology, 1960, 11:398-407.
    3. Ytrehus K, Liu Y, Tsuchida A, et al.Rat and rabbit heart infarction: effects of anesthesia, perfusate, risk zone, and method of infarct sizing. Am J Physiol, 1994, 267:H2383-2390.
    4. Miya Y,Kanda T,Tamura J,Sumino H,Kurabayashi M.A new murine model of coronary artery thrombosis and role of interleukin-8 in the development of coronary thrombosis. Res Commun Mol Pathol Pharmacol, 2000, 108:108-115.
    5. Ikeda Y, Umemura K.A characterization of myocardial infarction induced by thrombotic occlusion and a comparison with mechanical ligation of the rat coronary artery. Methods Find Exp Clin Pharmacol, 2001, 23:23-28.
    6. Kellar RS, Landeen LK, Shepherd BR, et al.Scaffold-based three-dimensional human fibroblast culture provides a structural matrix that supports angiogenesis in infarcted heart tissue. Circulation, 2001, 104:2063-2068.
    7. Shake JG, Gruber PJ, Baumgartner WA, et al.Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg, 2002, 73:1919-1925; discussion 1926.
    8. Kumashiro H,Kusachi S,Moritani H,et al.Establishment of a long-surviving murine model of myocardial infarction: qualitative and quantitative conventional microscopic findings during pathological evolution. Basic Res Cardiol, 1999, 94:78-84.
    9. Li RK, Weisel RD, Mickle DA,et al.Autologous porcine heart cell transplantation improved heart function after a myocardial infarction. J Thorac Cardiovasc Surg, 2000, 119:62-68.
    10. Moainie SL,Gorman JH, 3rd,Guy TS,et al.An ovine model of postinfarction dilated cardiomyopathy. Ann Thorac Surg, 2002, 74:753-760.
    11. Johns TN, Olson BJ. Experimental myocardial infarction. I. A method of coronary occlusion in small animals. Ann Surg, 1954, 140:675-682.
    12. Pfeffer MA, Pfeffer JM, Fishbein MC,et al.Myocardial infarct size and ventricular function in rats. Circ Res, 1979, 44:503-512.
    13. Rivard AL,Simura KJ,Mohammed S,et al.Rat intubation and ventilation for surgical research. J Invest Surg, 2006, 19:267-274.
    14. Degabriele NM, Griesenbach U, Sato K,et al.Critical appraisal of the mouse model of myocardial infarction. Exp Physiol, 2004, 89:497-505.
    15. Bhindi R,Witting PK,McMahon AC,Khachigian LM,Lowe HC.Rat models of myocardial infarction. Pathogenetic insights and clinical relevance. Thromb Haemost, 2006, 96:602-610.
    1. Engers R,Springer E,Michiels F, et al.Rac affects invasion of human renal cell carcinomas by up-regulating tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2 expression. J Biol Chem, 2001, 276:41889-41897.
    2. Engers R,Springer E,Kehren V,et al.Rac upregulates tissue inhibitor of metalloproteinase-1 expression by redox-dependent activation of extracellular signal-regulated kinase signaling. FEBS J, 2006, 273:4754-4769.
    3. Kraitchman DL,Tatsumi M,Gilson WD,et al.Dynamic imaging of allogeneic mesenchymal stem cells trafficking to myocardial infarction. Circulation, 2005, 112:1451-1461.
    4. Li HF,Huang CH,Ai LS, et al.Mutagenesis of the fusion peptide-like domain of hepatitis C virus E1 glycoprotein: involvement in cell fusion and virus entry. J Biomed Sci, 2009, 16:89.
    5. Murakami S, Nagaya N,Itoh T,et al.C-type natriuretic peptide attenuates bleomycin-induced pulmonary fibrosis in mice. Am J Physiol Lung Cell Mol Physiol, 2004, 287:L1172-1177.
    6. Nagaya N, Fujii T,Iwase T,et al.Intravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesis. Am J Physiol Heart Circ Physiol, 2004, 287:H2670-2676.
    7. Frangogiannis NG, Smith CW, Entman ML.The inflammatory response in myocardial infarction. Cardiovasc Res,,2002, 53:31-47.
    8. Menon LG,Picinich S,Koneru R,et al.Differential gene expression associated with migration of mesenchymal stem cells to conditioned medium from tumor cells or bone marrow cells. Stem Cells, 2007, 25:520-528.
    9. Lapidot T, Petit I. Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp Hematol, 2002, 30:973-981.
    10. Ries C,Egea V,Karow M,et al.MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood, 2007, 109:4055-4063.
    11. De Becker A,Van Hummelen P,Bakkus M,et al.Migration of culture-expanded human mesenchymal stem cells through bone marrow endothelium is regulated by matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-3. Haematologica, 2007, 92:440-449.
    12. Ho IA, Chan KY,Ng WH,et al.Matrix metalloproteinase 1 is necessary for the migration of human bone marrow-derived mesenchymal stem cells toward human glioma. Stem Cells, 2009, 27:1366-1375.
    1. Ries C, Egea V,Karow M,et al.MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood, 2007, 109:4055-4063.
    2. Beltrami AP, Urbanek K, Kajstura J, et al.Evidence that human cardiac myocytes divide after myocardial infarction. N Engl J Med, 2001, 344:1750-1757.
    3. Yau TM, Tomita S, Weisel RD,et al.Beneficial effect of autologous cell transplantation on infarcted heart function: comparison between bone marrow stromal cells and heart cells. Ann Thorac Surg, 2003, 75:169-176; discussion 176-167.
    4. Anversa P, Leri A, Kajstura J, Nadal-Ginard B.Myocyte growth and cardiac repair. J Mol Cell Cardiol, 2002, 34:91-105.
    5. Assmus B,Schachinger V,Teupe C,et al.Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI). Circulation, 2002, 106:3009-3017.
    6. Makino S, Fukuda K, Miyoshi S, et al.Cardiomyocytes can be generated from marrow stromal cells in vitro. J Clin Invest, 1999, 103:697-705.
    7. Reyes M,Dudek A,Jahagirdar B,et al.Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest, 2002, 109: 337-346.
    8. Toma C,Pittenger MF,Cahill KS, et al.Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2002; 105:93-98.
    9. Wang JS,Shum-Tim D,Galipeau J,et al.Marrow stromal cells for cellular cardiomyoplasty: feasibility and potential clinical advantages. J Thorac Cardiovasc Surg, 2000,120: 999-1005.
    10. Al-Khaldi A,Eliopoulos N,Martineau D,et al.Postnatal bone marrow stromal cells elicit a potent VEGF-dependent neoangiogenic response in vivo. Gene Ther, 2003, 10:621-629.
    11. Al-Khaldi A, Al-Sabti H, Galipeau J, et al.Therapeutic angiogenesis using autologous bone marrow stromal cells: improved blood flow in a chronic limb ischemia model. Ann Thorac Surg, 2003, 75:204-209.
    12. Shake JG, Gruber PJ, Baumgartner WA, et al.Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg, 2002, 73:1919-1925; discussion 1926.
    13. Asahara T, Murohara T, Sullivan A, et al.Isolation of putative progenitor endothelial cells for angiogenesis. Science, 1997, 275:964-967.
    14. Boomsma RA,Swaminathan PD,Geenen DL.Intravenously injected mesenchymal stem cells home to viable myocardium after coronary occlusion and preserve systolic function without altering infarct size. Int J Cardiol, 2007, 122:17-28.
    15. Soeki T,Tamura Y,Shinohara H,et al.Serial changes in serum VEGF and HGF in patients with acute myocardial infarction. Cardiology, 2000, 93:168-174.
    16. Ma J,Ge J,Zhang S,et al.Time course of myocardial stromal cell-derived factor 1 expression and beneficial effects of intravenously administered bone marrow stem cells in rats with experimental myocardial infarction. Basic Res Cardiol, 2005, 100:217-223.
    17. Lapidot T, Petit I.Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp Hematol, 2002, 30:973-981.
    18. De Becker A, Van Hummelen P, Bakkus M,et al.Migration of culture-expanded human mesenchymal stem cells through bone marrow endothelium is regulated by matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-3. Haematologica, 2007, 92:440-449.
    19. Ho IA, Chan KY, Ng WH, et al.Matrix metalloproteinase 1 is necessary for the migration of human bone marrow-derived mesenchymal stem cells toward human glioma. Stem Cells, 2009, 27:1366-1375.
    20. Papapetropoulos A, Fulton D, Mahboubi K,et al.Angiopoietin-1 inhibits endothelial?cell apoptosis via the Akt/survivin pathway. J Biol Chem, 2000, 275:9102-9105.
    21. Donovan M, Cotter TG.. Control of mitochondrial integrity by Bcl-2 family members and caspase-independent cell death. Biochim Biophys Acta, 2004, 1644: 133-147.
    22. Tsai RY,Kittappa R,McKay RD.Plasticity, niches, and the use of stem cells. Dev Cell, 2002, 2:707-712. ?
    1. Toma C, Pittenger MF, Cahill KS, et al.Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation, 2002, 105: 93-98.
    2. Liechty KW, MacKenzie TC, Shaaban AF, et al.Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep.Nat Med,2000,6:1282-1286.
    3. Pochampally RR, Neville BT, Schwarz EJ, et al.Rat adult stem cells (marrow stromal cells) engraft and differentiate in chick embryos without evidence of cell fusion. Proc Natl Acad Sci U S A, 2004, 101: 9282-9285.
    4. Pittenger MF, Martin BJ.Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res, 2004, 95(1):19-20.
    5 Mangi AA, Noiseux N, Kong D, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med, 2003,9: 195-201.
    6. Tang YL, Tang Y, Zhang YC, et al. Improved graft mesenchymal stem cell survival in ischemic heart with a hypoxia-regulated heme oxygenase-1 vector. J Am Coll Cardiol, 2005, 46:1339-1350.
    7. Young LS, Searle PF, Onion D, Mautner V. Viral gene therapy strategies: from basic science to clinical application[J]. Pathol, 2006, 08:299-318.
    8. Rodrigues T, Carrondo MJ, Alves PM, et al.Purification of retroviral vectors for clinical application: biological implications and technological challenges [J]. Biotechnol, 2007, 127: 520-541.
    9. Leibowitz G, Beattie GM, Kafri T, et al.Gene transfer to human pancreatic endocrine cells using viral vectors.Diabetes, 1999, 48:745-753.
    10. Yamamoto A.M, Chernajovsky Y, Lepault F, et al.The activity of immunoregulatory T cells mediating active tolerance is potentiated in nonobese diabetic mice by an IL-4-based retroviral gene therapy.Immunol, 2001, 166: 4973-4980.
    11. Chan J, Clements W, Field J, Nasa Z, et al. Transplantation of bone marrow genetically engineered to express proinsulin II protects against autoimmune insulitis in NOD mice.Gene Med, 2006, 8:1281-1290.
    12. Lu Y, Wang Z, Zhu M.Human bone marrow mesenchymal stem cells transfected with human insulin genes can secrete insulin stably.Ann.Clin,2006,36:127-136.
    13. Ting-De Ravin SS, Kennedy DR, Naumann N, et al.Correction of canine X-linked severe combined immunodeficiency by in vivo retroviral gene therapy. Blood, 2006, 107:3091-3097.
    14. Suter SE,Gouthro TA,OM alley T,et al.Marking of peripheral Tlymphocytes by retroviral transduction and transplantation of CD34(+)cells in a canine X-linked severe combined immunodeficiency model. VetImmunol Immunopathol, 2007, 117: 183-196.
    15. Robinet E, Fehse B, Ebeling S, et al.Improving the ex vivo retroviral-mediated suicide-gene transfer process in T lymphocytes to preserve immune function. Cytotherapy, 2005, 7:150–157.
    16. Yanagita S, Hori T, Matsubara Y, et al. Retroviraltransduction of acute myeloid leukaemia-derived dendritic cells with OX40 ligand augments their antigen presenting activity. Haematol, 2004, 124: 454-462.
    17. Gersbach CA, Le Doux JM, Guldberg RE, et al.Inducible regulation of Runx2-stimulated osteogenesis. Gene Ther, 2006, 13:873-882.
    18. Dai J, Rabie AB, Hagg U, Xu R.Alternative gene therapy strategies for the repair of craniofacial bone defects. Curr Gene Ther, 2004, 4:469-485.
    19. Oreffo RO, Virdi AS, Triffitt JT.Retroviral marking of human bone marrow fibroblasts: in vitro expansion and localization in calvarial sites after subcutaneous transplantation in vivo[J].Cell Physiol,2001,186:201–209.
    20. Daga A, Muraglia A, Quarto R, et al.Enhanced engraftment of EPO-transduced human bone marrow stromal cells transplanted in a 3D matrix in non-conditioned NOD/SCID mice. Gene Ther, 2002, 9:915–921.
    21. Adachi AN, Sato K, Usas A, et al. Muscle derived, cell based ex vivo gene therapy for treatment of full thickness articular cartilage defects[J]. Rheumatol, 2002,29: 1920-1930.
    22. Watson DJ, Longhi L, Lee EB, et al.Genetically modified NT2N human neuronal cells mediate long-term gene expression as CNS grafts in vivo and improve functional cognitiveoutcome following experimental traumatic brain injury. Exp Neurol, 2003, 62:368–380.
    23. Kong D, Melo LG, Mangi AA, et al. Enhanced inhibition of neointimal hyperplasia by genetically engineered endothelial progenitor Cells. Circulation, 2004, 109:1769- 1775.
    24. Griese DP, Achatz S, Batzlsperger CA, et al. Vascular gene delivery of anticoagulants by transplantation of retrovirally-transduced endothelial progenitor cells. Cardiovasc Res,2003, 58:469-477.
    25. Hu J, Takatoku M, Sellers SE, et al. Analysis of origin and optimization of expansion and transduction of circulating peripheral blood endothelial progenitor cells in the rhesus macaque model. Hum Gene Ther, 2002,13:2041-2050.
    26 .Ikezawa M, Cao B, Qu Z, et al.Dystrophin delivery in dystrophin-deficient DMDmdx skeletal muscle by isogenic muscle-derived stem cell transplantation.Hum Gene Ther, 2003, 14: 1535-1546.
    27. Pruchnic R, Yokoyama T, Lee JY, et al.Muscle derived cell mediated ex vivo gene transfer to the lower urinary tract: comparison of viral vectors. Urol Res, 2002,30: 310-316.
    28. Fleury S, Simeoni E, Zuppinger C, et al. Multiply attenuated, self- inactivating lentiviral vectors efficiently deliver and express genes for extended periods of time in adult rat cardiomyocytes in vivo. Circulation, 2003, 107: 2375-2382.
    29. Stitz J, Muhlebach MD, Blomer U, et al. A novel lentivirus vector derived from apathogenic simian immunodeficiency virus. Virology, 2001, 291: 191-197.
    30. Oh TK, Li MZ, Kim ST. Gene therapy for diabetes mellitus in rats by intramuscularinjection of lentivirus containing insulin gene. Diabetes Res Clin Pract, 2006, 71:233-240.
    31. Murata H, Hruz PW, Mueckler M.The mechanism of insulin resistance caused by HIV protease inhibitor therapy[J]. Biol Chem, 2000, 275:20251-20254.
    32. Kobinger GP, Louboutin JP, Barton ER, et al. Correction of the dystrophic phenotype by in vivo targeting of muscle progenitor cells.Hum Gene Ther,2003,15,1441-1449.
    33. Zhou BY, Ye Z, Chen G, et al. Cheng L. Inducible and reversible transgene expression in human stem cells after efficient and stable gene transfer. Stem Cells, 2007, 25:779-789.
    34. Shi Q, Wilcox DA, Fahs SA, et al. Montgomery, Lentivirus-mediated platelet-derived factor VIII gene therapy in murine haemophilia A. J Thromb Haemost, 2007,5:352-361.
    35. Zubler RH. Ex vivo expansion of haematopoietic stem cells and gene therapy development. Swiss Med Wkly, 2006, 136: 795-799.
    36. Bienemann AS, Martin-Rendon E, Cosgrave AS, et al.Long-term replacement of a mutated nonfunctional CNS gene: reversal of hypothalamic diabetes insipidus using an EIAV-based lentiviral vector expressing arginine vasopressin. Mol Ther, 2003, 7:588-596.
    37. Hughes SM, Moussavi-Harami F, Sauter SL, et al.Viral mediated gene transfer to mouse primary neural progenitor cells. Mol Ther, 2002, 5: 16-24.
    38. Buchet D, Serguera C, Zennou V, et al. Long-term expression of beta-glucuronidase by genetically modified human neural progenitor cells grafted into the mouse central nervous system. Mol Cell Neurosci, 2002, 19: 389-401.
    39. Jakobsson J, Nielsen TT, Staflin K, et al. Efficient transduction of neurons using Ross River glycoprotein-pseudotyped lentiviral vectors. Gene Ther, 2006, 13: 966-973.
    40. Geraerts M, Eggermont K, Hernandez-Acosta P, et al. Lentiviral vectors mediateefficient and stable gene transfer in adult neural stem cells in vivo. Hum Gene Ther, 2006, 17:635-650.
    41. Cenciarelli C, Budoni M, Mercanti D, et al.In vitro analysis of mouse neural stem cells genetically modified to stably express human NGF by a novel multigenic viral expression system. Neurol Res, 2006, 28:505-512.
    42. Vroemen M, Weidner N, Blesch A. Loss of gene expression in lentivirus- and retrovirus-transduced neural progenitor cells is correlated to migration and differentiation in the adult spinal cord. Exp Neurol, 2005, 195: 127-139.
    43. Strappe PM, Hampton DW, Brown D, et al. Identification of unique reciprocal and non reciprocal cross packaging relationships between HIV-1, HIV-2and SIV reveals an efficient SIV/HIV-2 lentiviral vector system with highly favourable features for in vivo testing and clinical usage. Retrovirology, 2005, 2:55-57.
    44. Duale H, Kasparov S, Paton JF, et al.Differences in transductional tropism of adenoviral and lentiviral vectors in the rat brainstem. Exp Physiol, 2005, 90:71-78.
    45. Fodor A, Harel C, Fodor L, et al.Adult rat liver cells transdifferentiated with lentiviral IPF1 vectors reverse diabetes in mice: an ex vivo gene therapy approach. Diabetologia, 2007, 50:121-130.
    46. He Z, Wang F, Kumagai-Braesch M, et al.Longterm gene expression and metabolic control exerted by lentivirustransduced pancreatic islets. Xenotransplantation, 2006, 13:195-203.
    47. Kobayashi N, Arata T, Okitsu T, et al. Transduction of human islets with the lentiviral vector. Transplant Proc, 2004, 36:2203-2204.
    48. Barbu AR, Bodin B, Welsh M, et al.A perfusion protocol for highly efficient transduction of intact pancreatic islets of Langerhans. Diabetologia, 2006, 49: 2388 -2391.
    49. Arata T, Okitsu T, Fukazawa T, et al. Maintenance of glucose-sensitive insulin secretion of cryopreserved human islets with University of Wisconsin solution and ascorbic acid-2 glucoside. Artif Organs, 2004, 28:529-536.
    50. Mizuguchi H, Hayakawa T.Targeted adenovirus vectors. Hum Gene Ther, 2004, 15: 1034-1044.
    51. Takahashi R, Ishihara H, Takahashi K, et al. Efficient and controlled gene expression in mouse pancreatic islets by arterial delivery of tetracycline- inducible adenoviral vectors [J].Mol Endocrinol, 2007, 38: 127- 136.
    52. Liu Y, Rabinovitch A, Suarez-Pinzon W, et al. Expression of the bcl-2 gene from a defective HSV-1 amplicon vector protects pancreatic beta- cells from apoptosis. Hum Gene Ther, 1996, 7:1719-1726.
    53. Carter BJ. Adeno-associated virus vectors in clinical trials. Hum Gene Ther, 2005, 16: 541-550.
    54. Wilson J.M.Adeno-associated virus and lentivirus pseudotypes for lungdirected gene therapy. Proc Am Thorac Soc, 2004, 1: 309-314.
    55. Grieger JC, Choi VW, Samulski RJ, et al. Production and characterization of adeno-associated viral vectors. Nat Protoc, 2006, 1: 1412-1428.
    56. Grieger JC, Samulski RJ. Adeno-associated virus as a gene therapy vector:vector development, production and clinical applications. Adv Biochem Eng Biotechnol, 2005, 99:119-145.
    57. Duan D, Yan Z, Yue Y, Engelhardt JF. Structural analysis of adenoassociated virus transduction intermediates. Virology, 1999, 261: 8-14.
    58. Grieger JC, Samulski RJ. Packaging capacity of adeno-associated virus serotypes:impact of larger genomes on infectivity and postentry steps.J Virol, 2005, 79: 9933- 9944.
    59. Rehman KK, Wang Z, Bottino R, et al. Efficient gene delivery to human and rodent islets with double-stranded (ds) AAV-based vectors. Gene Ther, 2005,12: 1313- 1323.
    60. Flotte T, Agarwal A, Wang J, et al. Efficient ex vivo transduction of pancreatic islet cells with recombinant adeno-associated virus vectors. Diabetes, 2001, 50: 515-520.
    61. Goudy K, Song S, Wasserfall C, et al.Adeno-associated virus vector-mediated IL-10 gene delivery prevents type 1 diabetes in NOD Mice. Proc Natl Acad, 2001, 98: 13913-13918.
    62. Zhang N, Clement N, Chen D, et al.Transduction of pancreatic islets with pseudotypedadeno-associated virus: effect of viral capsid and genome conversion. Transplantation, 2005, 80:683-690.
    63. Lai Y, Yue Y, Liu M, et al.Efficient in vivo gene expression by trans-splicing adenoassociated viral vectors. Nat Biotechnol,2005,23:1435-1439.
    64. Duan D, Yue Y, Yan Z, et al.A new dual-vector approach to enhance recombinant adeno-associated virus-mediated gene expression through intermolecular cis activation. Nat Med, 2000, 6:595-598.
    65. Benhamou PY, Moriscot C, Prevost P, et al.Standard- ization of procedure for efficient ex vivo gene transfer into porcine pancreatic islets with cationic liposomes. Transplantation, 1997, 93:1798-1803.
    66. Chae HY, Lee BW, Oh SH, Ahn YR, et al. Effective glycemic control achieved by transplanting non-viral cationic liposome-mediated VEGF-transfected islets in streptozotocin-induced diabetic mice. Exp Mol Med, 2005, 37:513-523.
    67. Mahato RI, Henry J, Narang AS, et al.Cationic lipid and polymer-based gene delivery to human pancreatic islets. Mol Ther, 2003, 7: 89-100.
    68. Saldeen J, Sandler S, Bendtzen K, et al.Liposome-mediated transfer of IL-1 receptor antagonist gene to dispersed islet cells does not prevent recurrence of disease in syngeneically transplanted NOD mice. Cytokine, 2000, 12: 405-408.
    69. Prudhomme GJ, Draghia-Akli R. Wang Q.Plasmid-based gene therapy of diabetes mellitus. Gene Ther, 2007, 14: 553-564.
    70. Prudhomme GJ, Glinka Y, Khan AS, et al. Electroporation-enhanced nonviral gene transfer for the prevention or treatment of immunological, endocrine and neoplastic diseases. Curr Gene Ther, 2006, 6: 243-273.
    71. Odorico JS, Kaufman DS,Thomson JA. Multilineage differentiation from humanembryonic stem cell lines. Stem Cells, 2001, 19(3): 193-204.
    72. Bobbert M. Ethical questions concerning research on human embryos, embryonic stem cells and chimeras. Biotechnol, 2006, 1:1352-1369.
    73. Wadman M. Congress and Bush set to clash on stem cells again. Nature, 2007, 445: 134-135.
    74. Ying QL,Nichols J,Evans EP,et a1.Changing potency by spontaneous fusion. Nature,2002,416:545-548.
    75. Wang X,Willenbring H,Akkari Y,et a1.Cell fusion is the principal source of bone marow derived hepatocytes.Nature,2003,422:897-901.
    76. Hurwitz DR, Kirchgesser M, Merrill W, et al.Systemic delivery of human growth hormone or human factor IX in dogs by reintroduced genetically modified autologous bone marrow stromal cells[J].Hum Gene Ther,1997,8(2):137-156.
    77. Niagara MI, Haider HK, Jiang S, et al. Pharmacologically preconditioned skeletal myoblasts are resistant to oxidative stress and promote angio- myogenesis via release of paracrine factors in the infracted heart.Circ Res,2007,100:545-555.
    78. Suzuki K, Smolenski RT, Jayakumar J, et al.Heat shock treatment enhances graft cell survival in skeletal myoblast transplantation to the heart.Circulation, 2000, 102 (3): 216-221.
    79. Meuillet J, Mahadevan D, Vankayalapati H, et al. Specific inhibition of the Akt1 pleckstrin homology domain by D-3-deoxy- phosphatidyl-myo-inositol analogues. Mol Cancer Ther, 2003, 2:389-399.
    80. Otterbein LE, Choi AM. Heme oxygenase: colors of defense against cellular Stress. Am J Physiol, 2000, 279:1029-1037.
    81. Tang YL, Zhao Q, Zhang YC, et al. Autologous mesenchymal stem cell transplantation induce VEGF and neovascularization in ischemic myocardium. Regul Pept,2004,117:3-10.
    82. Lange C, Bassler P, Lioznov MV, et al. Hepatocytic gene expression in cultured rat mesenchymal stem cells.Transplant Proc, 2005, 37:276-279.
    83. Tang YL, Zhao Q, Qin X, et al. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Ann Thorac Surg, 2005, 80:229-236.
    84. Lei Y, Haider HKh, Shujia J, et al.Therapeutic angiogenesis. Devising new strategies based on past experiences.Basic Res Cardiol, 2004, 99:121-132.
    85. Chakrabarti SK, Mirmira RG. Transcription factors direct the development and function of pancreatic beta cells.Trends Endocrinol Metab, 2003, 14:78-84.
    86. Ahlgren U, Jonsson J, Jonsson L, et al. Beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the beta-cell phenotype and maturity onset diabetes. Genes Dev, 1998, 12:1763-1768.
    87. Wang J, Elghazi L, Parker SE, et al. The concerted activities of Pax4 and Nkx2.2 are essential to initiate pancreatic beta-cell differentiation. Dev Biol, 2004, 266:178-189.
    88. Drukker M, Katz G, Urbach A, et al. Characterization of the expression of MHC proteins in human embryonic stem cells. Proc Natl Acad, 2002,99:9864-9869.
    89. Tang DQ, Lu S, Sun YP, et al. Reprogramming liver-stem WB cells into functional insulin-producing cells by persistent expression of Pdx1-and Pdx1-VP16 mediated by lentiviral vectors. Lab Invest,2006,86:83-93.
    90. Tang X, Cai PQ, Lin YQ, et al.Genetic engineering neural stem cell modified by lentivirus for repair of spinal cord injury in rats. Chin Med, 2006, 21:120-24.
    91. Cai PQ, Tang X, Lin YQ, et al.The experimental study of genetic engineering human neural stem cells mediated by lentivirus to express multigene. Chin J Traumatol, 2006, 9:43-49.
    92. Blits B, Kitay BM, Farahvar A, et al. Lentiviral vector-mediated transduction of neural progenitor cells before implantation into injured spinal cord and brain to detect their migration, deliver neurotrophic factors and repair tissue. Restor Neurol Neurosci, 2005, 23:313- 324.
    93. Brazelton TR, Rossi FM, Keshet GI, et al.From marrow to brain: expression of neuronal phenotypes in adult mice.Science, 2000, 290:1775-1779.
    94. Baker AH, Sica V, Work LM, et al.Brain protection using autologous bone marrow cell, metalloproteinase inhibitors, and metabolic treatment in cerebral ischemia. Proc Natl Acad, 2007, 104: 3597-3602.
    95. Kobune M, Kawano Y, Ito Y, et al. Telomerized human multipotent mesenchymal cells can differentiate into hematopoietic and cobblestone area-supporting cells. Exp Hematol, 2003, 31:715-722.
    96. Jiang XR, Jimenez G, Chang E, et al.Telomerase expression in human somatic cells does not induce changes associated with a transformed phenotype. Nat Genet, 1999, 21:111-114.
    97. Honma T, Honmou O, Iihoshi S, et al. Intravenous infusion of Immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Exp Neurol, 2006, 199: 56-66.
    98. Zhao LX,Zhang J,Cao F,et a1.Modification of the Brain-derived neurotrophic factor gene:a portal to transfotin mesenchymal stem cells into advantageous engineering cells for neuroregenemtion and neuroprotection.Experimental Neurology,2004, 190 (2): 396-406.
    99. Kurozumi K,Nakamura K,Tamiya T,et a1.BDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion mode1.Molecular Therapy,2004,2(9):189-197
    100. Schwarz EJ, Alexander GM, Prockop, DJ, et al. Multipotential marrow stromal cells transduced to produce L-DOPA:engraftment in a rat model of Parkinson disease. Hum Gene Ther, 1999, 10: 2539-2549
    101. Fisher LJ, Raymon HK, Gage FH. Cells engineered to produce acetylcholine: the rapeutic potential for Alzheimer's disease. Ann NY Acad, 1993, 695:278-284.
    102. Tuszynski MH, Thal L, Pay M, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med, 2005, 11: 551-555.
    103. Gazit D, Turgeman G, Kelley P, et al.Engineered pluripotent mesenchymal cells integrate and differentiate in regenerating bone: a novel cell-mediated gene therapy.Gene Med, 1999, 1: 121-133.
    104. Moutsatsos IK, Turgeman G, Zhou S, et al.Exogenously regulated stem cell-mediated gene therapy for bone regeneration. Mol Ther, 2001, 3: 449-461.
    105. Hasharoni A, Zilberman Y, Turgeman G, et al. Murine spinal fusion induced by engineered mesenchymal stem cells that conditionally express bone morphogenetic protein-2. Neurosurg Spine, 2005, 3:47-52.
    106. Hung SC, Deng WP, Yang WK, et al. Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging. Clin Cancer Res, 2005,11: 7749 - 7756.
    107. Lokshin A, Mayotte JE, Levitt ML. Mechanism of interferon betainduced squamous differentiation and programmed cell death in human non-small-cell lung cancer cell lines. Natl Cancer Inst, 1995,87: 206-212.
    108. Studeny M, Marini FC, Champlin RE, et al. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res, 2002,62(13):3603-3608.
    109. Studeny M, Marini FC, Dembinski JL, et al.Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. Natl Cancer Inst, 2004, 96(21):1593-1603.
    110. Nakamizo A, Marini F, Amano T, et al.Human bone marrow- derived mesenchymal stem cells in the treatment of gliomas. Cancer Res, 2005, 65:3307-3318.
    111. Stagg J, Lejeune L, Paquin A, et al. Marrow stromal cells for interleukin- 2 delivery in cancer immunotherapy. Hum Gene Ther, 2004, 15:597-608.
    112. Pittenger MF.Mackay AM, Beck SC,et a1.Multilineage potential of adult human mesenchymal stem cells[J].Science,1999,284(5411):143-147.
    113. Colter DC,Class R,Digiolamo CM,et a1.Rapid expansion of recycling stem cells in cultures of plastic adherent cells from human bone marrow[J].Proc Natl Acad,2000,97(9)3213-3218.
    114. Moayeri M, Hawley TS, Hawley RG. Correction of murine hemophilia A by hematopoietic stem cell gene therapy. Mol Ther, 2005, 12:1034-1042.
    115. Gangadharan B, Parker ET, Ide LM, et al.Highlevel expression of porcine factor VIII from genetically modified bone marrow-derived stem cells. Blood , 2006,107:3859-3864.