用户名: 密码: 验证码:
MSCs多向分化潜能及对失神经肌萎缩的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分MSCs多向分化潜能(成骨、成肌以及神经定向诱导分化)的研究
     目的①探讨间充质干细胞(MSCs)体外诱导分化为骨样细胞、肌样细胞以及神经元(NCs)样/星形胶质样细胞的有效方法。②评价体外诱导分化的神经前体样细胞(NPCs)、神经元(NCs)样细胞以及星形胶质样细胞的体外存活和增殖特性,为神经细胞再生寻找良好的种子来源。
     方法参考有关文献,采用贴壁筛选法自大鼠骨髓中分离培养间充质干细胞,进行传代纯化,收集得到第5代(P5)间充质干细胞单细胞悬液(1×10~(5~6)/ml)。①成骨诱导分化方案:DMEM/10%FBS培养基+0.1μmol/l地塞米松+50ng/ml维生素C+2.16mg/mlβ-甘油磷酸钠。诱导分化2周以上。行Van Kossa银染色和茜素红染色。②成肌诱导分化方案:DMEM/20%FBS+3μmol/l5-氮胞苷(5-azayttidine)。诱导分化24h,培养2~3周以上。观察形成的肌样细胞及肌管样结构。③神经定向诱导分化方案:采用多因子、分阶段逐步诱导方法定向诱导MSCs分化为神经前体细胞(NPCs),进而分化为NCs样/星形胶质样细胞。即:MSCs以1×10~6/L置入6孔培养板中,加入生长培养基:DMEM/20%FBS培养基,24h后更换为诱导分化培养基:DMEM/F12+2%B27+40μg/L碱性成纤维生长因子(bFGF)+20μg/L表皮生长因子诱导分化培养基(EGF),培养10~20d,定向诱导MSCs分化为NPCs。下一个阶段更换诱导分化培养基为生长培养基:DMEM/5%FBS,分两组进行诱导:一组向神经元样细胞诱导分化:全反式维甲酸(RA)连续加样,首次浓度0.5μmol/L,以后每天半量加样;二组向星形胶质样细胞诱导分化:10μg/L血小板衍生生长因子BB(PDGF-BB)首次加样,以后每天半量加样,均连续诱导10~14d,制作两组细胞爬片均行纤维丝蛋白200(NF200)、胶质纤维酸性阳性蛋白(GFAP)免疫组织化学荧光染色。④神经元样/星形胶质样细胞共同培养:收集诱导分化的神经元样/星形胶质样细胞,各自浓度1×106/L,以1:1混合在DMEM/5%FBS培养基中共同培养。观察两组细胞生长情况。⑤应用活细胞计数试剂盒CCK-8检测单独诱导分化的NCs样/星形胶质样细胞的增殖及存活情况,绘制生长曲线及细胞存活曲线。
     结果①经DMEM/10%FBS培养基+0.1μmol/l地塞米松+50ng/ml维生素C+2.16mg/mlβ-甘油磷酸钠诱导培养2周以上,MSCs细胞聚集呈结节状,钙盐沉积,Van Kossa银染色法阳性。茜素红染色阳性。②经3μmol/l 5-氮胞苷诱导处理大鼠MSCs,24小时部分细胞胞体收缩变为瘦长形状, 1~2周后细胞相互形成肌管样结构。2周后可以观察到自发搏动。③bFGF+EGF连续诱导7d后MSCs分化为球团样的神经前体样细胞,巢蛋白(Nestin)表达阳性。RA、PDGF-BB分别连续诱导10d后MSCs逐步分化出神经元样、星形胶质样细胞,分别表达NF200、GFAP蛋白。④神经元样/星形胶质样细胞可以共同培养、生长,呈现各自的细胞形态。⑤活细胞计数试剂盒CCK-8检测显示:神经元样细胞早期生长较缓慢,具有一定的增殖能力,6~7d后达到高峰,此后增殖能力明显下降,细胞存活率明显下降,14~15d之后细胞存活率下降到7.6%以下,难以传代培养。星形胶质样细胞初期生长慢,5d后生长加速,进入对数生长期,7~8d后进入平台期,仍保持较高的细胞存活率。
     结论①MSCs是具有自我增殖和多向分化潜能的干细胞。可以诱导分化为骨样细胞、骨骼肌样细胞、神经元样细胞及星形胶质样细胞。②诱导分化的神经元样/星形胶质样细胞可以共同培养、生长。③单独诱导分化的神经元样细胞可继续存活13-15d后发生自然死亡,难以传代,星形胶质样细胞生长较稳定,具有较强的存活和增殖能力,可传代培养至3代以上。
     第二部分MSCs归巢性及对失神经肌萎缩的作用
     目的检测大鼠间充质干细胞(MSCs)经静脉移植后在周围神经损伤模型内的迁移、分布情况,评价其对轴突、靶器官的保护作用。
     方法制作80只大鼠坐骨神经损伤模型,收集P5代MSCs行BrdU(10μmol/L)掺和标记48h。经尾静脉注射BrdU标记的MSCs(2×10~7/ml)入体内。术后8周内评价坐骨神经指数(SFI)。术后神经断端、肌肉组织取材均行BrdU免疫组织化学荧光染色。肌肉组织行HE染色、TUNEL组织化学染色。Image-Pro Plus 5.0专业图像分析软件分析肌纤维横截面积并计算以及实验侧与正常侧肌纤维横截面积之比,分析肌细胞调亡数量。所得数据均用均数±标准差(x——±s)表示,结果用SPSS统计软件行t检验。
     结果MSCs体外可以被BrdU掺和标记,阳性标记率为62.5%。术后4、8周神经断端、肌肉组织内检测到BrdU阳性细胞。实验组SFI、肌纤维横截面积明显高于对照组,细胞凋亡数量低于对照组,均有统计学意义﹙4周P<0.05, 8周P<0.01﹚。
     结论MSCs静脉移植至体内能归巢到损伤的神经断端及肌肉组织,可以存活至少8周,具有促进轴突再生、延缓肌萎缩的作用。
     第三部分骨髓源性神经前体样细胞对失神经肌萎缩的作用
     目的探讨大鼠间充质干细胞(MSCs)诱导分化的神经前体样细胞(NPCs)移植在体内存活情况,评价其及对失神经肌萎缩的作用。
     方法联合应用EGF、bFGF诱导MSCs分化为NPCs。行Nestin免疫组织化学荧光染色鉴定,并行5-溴脱氧尿苷(BrdU)掺和标记实验和单细胞克隆实验。建立90只大鼠坐骨神经损伤模型。BrdU掺和标记的NPCs以5×106/ml注入神经损伤断端。术后4、8周神经断端、肌肉组织取材,神经断端分别行Nestin、BrdU单标免疫荧光染色检验。肌肉组织行HE染色、TUNEL免疫组织化学染色。Image-Pro Plus 5.0专业图像分析软件分析肌纤维横截面积并计算以及实验侧与正常侧肌纤维横截面积之比。所得数据结果用SPSS统计软件行t检验。
     结果MSCs体外诱导分化可获得大量活性的NPCs,Nestin蛋白染色阳性,体外可以被BrdU掺和标记,阳性标记率为87.9%,单细胞克窿率为2.7%。移植术后4、8周实验组神经断端均检测到Nestin、BrdU阳性细胞。实验组肌纤维横截面积明显高于对照组,肌细胞调亡数量明显低于对照组,均有统计学意义﹙4周P<0.05,8周P<0.01﹚。
     结论骨髓源性NPCs移植至神经断端可以存活至少8周,具有延缓失神经肌萎缩、抑制失神经肌肉细胞调亡的作用。
     第四部分骨髓源性神经样细胞对失神经肌萎缩的作用
     目的探讨间充质干细胞(MSCs)诱导分化的神经元(NCs)样、星形胶质样细胞联合移植后在体内存活情况,评价其联合移植对失神经肌萎缩的作用。
     方法多因子、分阶段逐步诱导MSCs分化为NCs样、星形胶质样细胞。行NF200、GFAP免疫组织化学荧光染色鉴定。建立60只大鼠失神经骨骼肌模型。行细胞联合移植,以5×10~6/ml肌肉注射入失神经骨骼肌内。术后4、8周肌肉组织取材行HE染色、免疫组织化学染色检测NF200、GFAP阳性细胞。Image-Pro Plus 5.0专业图像分析软件分析肌纤维横截面积并计算以及实验侧与正常侧肌纤维横截面积之比。所得数据结果用SPSS统计软件行t检验。
     结果MSCs逐步诱导分化出NCs样、星形胶质样细胞。分别表达NF200、GFAP阳性蛋白。移植术后4、8周实验侧肌肉组织检测到NF200、GFAP阳性细胞。实验侧肌纤维横截面积明显高于对照组,有统计学意义(4周P<0.05,8周P<0.01)。
     结论骨髓源性NCs样、星形胶质样细胞可以体外大量获得,联合移植至肌肉内可以存活至少8周,可以增加失神经肌肉面积,有效改善失神经肌萎缩。
PartⅠThe multipotency of mesenchymal stem cells in rats.
     Objective To investigate multipotentcy of MSCs differentiation into osteoblasts,skeletal muscle, neuron-like cells and astrocyte-like cells in vitro and evaluate the proliferation and survival capacity of MSCs-differentiated neural progenitor cells, neuron-like cells and astrocyte-like cells in vitro.
     Methods Adult rat MSCs were isolated and cultured for 5 passages in vitro. Osteogenic differentiation was induced by plating the cells at 1×10~5cells/ml in DMEM medium containing 10%FBS,supplenmented with 0.1μmol/l dexamethasone,50ng/ml ascorbic acid, and 2.16mg/mlβ–glycerophosphate. Cells were cultured for 14days and the medium was changed twice a week. Skeletal muscle differentiation was induced at 1×10~5cells/ml by DMEM/20%FBS supplenmented with 3μmol/l 5-azayttidine for 24h, then cultured for 2~3weeks.Neural differentiation were selectively and progressively induced into neuron-like cells and astrocyte-like cells.by multi-cytokines, first DMEM/F12/2%B27 with EGF(20μg/L) and bFGF(40μg/L) for 10~20days, then DMEM/5%FBS with retinoic acid (0.5μmol/L) or PDGF-BB(10μg/L) for another 10~20days. The differentiated neuron-like cells and astrocyte-like cells were co-cultured at 1×10~5cells/ml at the ratio of 1:1 in DMEM medium containing 10%FBS. Immunohistochemistry stainings were selectively carried out for CD44,CD45,nestin,NF200 and GFAP. The cell proliferation and survival assays were made using cell counting Kit-8.
     Results The MSCs showed a fibroblast-like morphology, and could self-renew and survive at least 15 passages. In the presence of selected cytokines MSCs were effectively differentiated into osteoblasts for 4 weeks. The cells morphology changed after treatment with 5-azacytidine for 1 week. MSCs connected with adjoining cells after one week, formed myotube-like structures, began spontaneously beating after two weeks, and beat synchronously after three weeks. MSCs were differentiated into neurospheres -like cells with EGF and bFGF for 7 days. Then neurospheres-like cells were differentiated into neuron-like cells with retinoic acid and astrocyte-like cells with PDGF-BB for 10~14 days, respectively positive for nestin, NF200 and GFAP immunohistochemistry stainings. The differentiated neuron-like cells and astrocyte-like cells could survive the next co-culture procedure.The CCK-8 assays demonstrated that the differentiated neuron-like cells could proliferate and survive until the next 13~15 days. However the astrocyte-like cells proliferated and survived better and could be cultured for at least 3 passages.
     Conclusions MSCs were self-renewing and mutipotent cells which maintain the capacity of differentiation into osteoblasts,skeletal muscle-like cells,neuron-like cells and astrocyte-like cells in vitro by multi-cytokines induction protocols. And the MSCs-derived astrocyte-like cells had a better proliferation and survival capacity than the neuron-like cells.
     PartⅡTherapeutic benefits of intravenous administration of mesenchymal stem cells on peripheral nerve regeneration in rats
     Objective To investigate the migration and distribution of rat bone mesenchymal stem cells in vivo after peripheral nerve injury and evaluate the beneficial effects on peripheral nerve regeneration and delaying denervation muscle atrophy.
     Methods Eighty female Sprague-Dawley rats were randomly divided into 2 groups, with 40 in each. All the sciatic nerves were transected and sutured with 8-0 nylon suture. The transplanted group received an infusion of BrdU-labelled MSCs at 2×10~7cells/ml cells/ml through the tail vein. Every week until 12 weeks after surgery the sciatic function index (SFI) was assessed by walking tract test. Sections were carried out for HE staining, BrdU and TUNEL immunohistochemical staining.
     Results BrdU-labeled cells were detected housing the nerve stumps and denervation muscles at 1 week and 8 weeks. MSCs transplantation group had a promotion of sciatic nerve functional recovery from weeks 4 to 12 and the number of denervation muscle apoptotic cells significantly decreased compared with that of control group at 8 weeks(P<0.05, P<0.01).
     Conclusions MSCs is capable in vivo of homing to the nerve stumps and denervation muscles after peripheral nerve injury via the blood circulation, and had therapeutic benefits on promoting peripheral nerve regeneration and delaying denervation muscle atrophy.
     PartⅢThe effect of neural progenitor cells differentiated from bone marrow mesenchymal stem cells on denervation muscle atrophy
     Objective To investigate the effect on delaying denervation muscle atrophy after peripheral nerve injury by transplantion of neural progenitor cells differentiated from bone marrow mesenchymal stem cells.
     Methods Isolated and purified MSCs were induced into neural progenitor cells by EGF (20ng/ml) and bFGF (40ng/ml) for 10~20d. Differentiated cells were analyzed by single cell clone technique and immunohistochemistry staining for nestin. Ninety female Sprague-Dawley rats were randomly divided into 2 groups, with 45 in each. All the sciatic nerves were transected and sutured with 8-0 nylon suture. Before transplantation the neural progenitor cells were labeled with BrdU for 48h and then were injected into the sciatic nerve stumps at 5×10~6cells/ml. At weeks 4, 8 the nerve stump frozen sections were carried out for BrdU immunofluorescence staining, and muscle paraffin sections were carried out for HE staining and TUNEL immunohistochemical staining. Muscle cross-sectional areas and apoptotic cells were measured by an image analyzer.
     Results MSCs were positive for CD44, however negative for CD45 and nestin. After 10~14d differentiation, neurospheres were observed and mostly of them were positive for nestin. The rate of label of BrdU was 87.9% and the single-clone formed rate was 2.7%.At weeks 4 and 8 after surgery BrdU positive cells were detected at the nerve stumps. Muscle cross-sectional areas of cell transplantation group significantly increased and the number of the denervation muscle apoptotic cells significantly decreased compared with that of control group (P<0.05, P<0.01).
     Conclusions MSCs can be efficiently differentiated into neural progenitor cells with EGF and bFGF. Differentiated neural stem cell-like cells can survive the next transplantation into nerve stumps and had beneficial effects on delaying denervation muscle atrophy after injury.
     PartⅣThe effect of differentiated neuron-like cells and astrocyte-like cells from bone marrow mesenchymal stem cells on denervation muscle atrophy
     Objective To investigate the survive of MSCs-differentiated neuron-like cells and astrocyte-like cells after co-transplantation in vivo and evaluate the beneficial effects on delaying denervation atrophy.
     Methods Isolated and purified rMSCs were selectively and progressively induced into neuron-like cells and astrocyte-like cells by multi-cytokine, EGF and bFGF first, followed by retinoic acid or PDGF. Immunohistochemistry staining was carried out for CD44, CD45, nestin, NF200 and GFAP. Sixty female Sprague-Dawley rat sciatic nerve transection models were made and then randomly divided into 2 groups, with 25 in each. In the experimental group, differentiated neuron-like cells and astrocyte-like cells were co-injected into the denervated skeletal muscle at 5×10~6cells/ml. At weeks 4 and 8 after cell transplantation muscle frozen sections were carried out for NF200 and GFAP immunofluorescence staining and muscle paraffin sections were carried out for HE staining Muscle cross-sectional areas were measured by an image analyzer.
     Results MSCs were effectively differentiated into neurospheres -like cells , then neuron-like cells and astrocyte-like cells ,respectively positive for nestin , NF200 and GFAP. At 4 ,8 weeks after transplantation NF200 and GFAP -positive cells were detected in denervated skeletal muscle .Muscle cross-sectional areas of cell transplantation group were significantly increased,compared with that of the control group. There was a statistical significance (P < 0.05, P < 0.01). Conclusions Differentiated neuron-like cells and astrocyte-like cells can be acquired in vitro by effective induction protocols. After co-transplantation into the denervated skeletal muscle cells can survive in vivo and have therapeutic benefits on delaying denervation atrophy.
引文
1 Mckay R.1997.Stem cells in the nervous system.Science,276:66-71.
    2 Grove JE, Bruscia E, Krause DS. Plasticity of bone marrow-derived stem cells. Stem Cells,2004;22(4):487-500.
    3 Sanchez-Ramos JR.. Neural cells derived from adult bone marrow and umbilical cord blood. J Neurosci Res,2002, 69(6):880-893.
    4 Azizi S,Stokes D,Augelli B,etc.Prockop after injection into neonatal mouse brains.Proc Natl Acad Sci USA.1999;96:10711-10716.
    5 Rismanchi N, Floyd CL, Berman RF, et al. Cell death and long-term maintenance of neuron-like state after differentiation of rat bone marrow stromal cells: a comparison of protocols. Brain Res. 2003 ,Nov 21;991(1-2):46-55.
    6 Aoki S, Toda S, Ando T, et al. Bone marrow stromal cells, preadipocytes, and dermal fibroblasts promote epidermal regeneration in their distinctive fashions. Mol Biol Cell,2004, 15(10):4647-4657.
    7 Fiedler J, Roderer G, Gunther KP, etc.BMP-2, BMP-4, and PDGF-bb stimulate chemotactic migration of primary human mesenchymal progenitor cells. J Cell Biochem. 2002;87(3):305-312.
    8 Reyes M,Verfaillie CM.2001.Characterization of multipotent adult progenitor cells, a subpopulation of mesenchymal stem cells. Ann N Y Acad Sci,2001,938:231-233.
    9 Makino S, Fukuda K, Miyoshi S, etc.Cardiomyocytes can be generated from marrow stromal cells in vitro.J Clin Invest. 1999,103(5):697-705.
    10 Fukuda K.Application of mesenchymal stem cells for the regeneration of cardiomyocyte and its use for cell transplantation therapy. Hum Cell, 2003,16(3):83-94.
    11 Westerlund U, Moe MC, Varghese M,etc. Stem cells from the adult human brain develop into functional neurons in culture. Exp Cell Res, 2003,289(2):378-383.
    12 Corti S, Locatelli F, Strazzer S,etc. Modulated generation of neuronal cells from bonemarrow by expansion and mobilization of circulating stem cells with in vivo cytokine treatment. Exp Neurol. 2002, 177(2):443-452.
    13 Kabos P, Ehtesham M, Kabosova A,etc.Generation of neural progenitor cells from whole adult bone marrow. Exp Neurol. 2002 ,178(2):288-93.
    14 Friedenstein AJ, Piatetzky-Shapiro II, Petrakova KV. Osteogenesis in transplants of bone marrow cells.J Embryol Exp Morpho, 1966,16(3):381-390.
    15 Gordon MY, Blacket NM.1998. Reconstruction of the hematopoietic system after stem cell transplantation.Cell Transplant, 1998 ,7(4):339-344.
    16 Sanchez-Ramos JR,.Neural cells derived from adult bone marrow and umbilical cord blood. J Neurosci Res, 2002, 69(6):880-893.
    17 Herzog EL,Chai L,Krause DS. Plasticity of marrow-derived stem cells.Blood,2003,102(10):3483-3493.
    18 Ying QL, Nichols J, Evans EP, etc.Changing potency by spontaneous fusion. Nature.,2002,416(6880):545-548.
    19 Masuya M, Drake CJ, Fleming PA,etc.Hematopoietic origin of glomerular mesangial cells.Blood,2003,101(6):2215-2218.
    20 Mokry J, Nemecek S. Immunohistochemical detection of intermediate filament Nestin.Acta Medica (Hradec Kralove). 1998;41(2):73-80.
    21 Aoki S, Toda S, Ando T, et al. Bone marrow stromal cells, preadipocytes, and dermal fibroblasts promote epidermal regeneration in their distinctive fashions. Mol Biol Cell,2004,15(10):4647-4657.
    22 Seri B, Garcia-Verdugo JM, McEwen BS,et al. Astrocytes give rise to new neurons in the adult mammalian hippocampus.J Neurosci,2001, 21(18):7153-7160.
    23 Jiang Y, Henderson D, Blackstad M, et al.Neuroectodermal differentiation from mouse multipotent adult progenitor cells. Proc Natl Acad Sci U S A,2003 ,100 Suppl 1:11854-11860.
    24 Bartholomew A, Sturgeon C, Siatskas M,et al. Mesenchymal stem cells suppresslymphocyte proliferation in vitro and prolong skin graft survival in vivo.Exp Hematol,2002 , 30(1):42-48.
    
    1. Bittira B, Shum-Tim D, Al-Khaldi A, et al. Mobilization and homing of bone marrow stromal cells in myocardial infarction. Eur J Cardiothorac Surg,2003,24(3):393-398.
    2. Koc ON, Lazarus HM. Mesenchymal stem cells: heading into the clinic.Bone Marrow Transplant,2001,27(3):235-239.
    3. Chen J, Li Y, Wang L,et al. Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats.J Neurol Sci,2001,89(1-2):49-57.
    4. Li Y,Chen J,Chen XG, et al. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functionalrecovery.Neurology,2002,59(4):514-523.
    5. Berry MF, Engler AJWoo YJ, et al.Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance. Am J Physiol Heart Circ Physiol, 2006,290(6):H2196-2203.
    6. Koc ON, Day J, Nieder M, et al.Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH).Bone Marrow Transplant,2002 ,30(4):215-2.
    7. Akiyama Y, Radtke C,Honmou O, et al.Remyelination of the spinal cord following intravenous delivery of bone marrow cells. Glia, 2002,39(3):229-236.
    8.沈宁江,朱家恺.坐骨神经功能指数在神经功能评价中的应用.中华显微外科杂志,1993,16(4):284-287.
    9. Son BR, Marquez-Curtis LA, Kucia M, et al.Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal-derived factor-1-CXCR4 and hepatocyte growth factor-c-met axes and involves matrix metalloproteinases.Stem Cells, 2006 ,24(5):1254-1264.
    10. Abbott JD, Huang Y, Liu D, et al.Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation,2004,110(21):3300-3305.
    11. Orlic D, Kajstura J, Chimenti S, et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci USA,2001, 28;98(18):10344-10349.
    12. Lopez Ponte A, Marais E, Gallay N,et al. The in vitro migration capacity of human bone marrow mesenchymal stem cells: Comparison of chemokine and growth factor chemotactic activities.Stem Cells, 2007,29; [Epub ahead of print]
    13. Caplan AI,Dennis JE. Mesenchymal stem cells as trophic mediators.J Cell Biochem,2006 98(5):1076-1084.
    14. Kamihata H, Matsubara H, Nishiue T, et al.Improvement of collateral perfusion and regional function by implantation of peripheral blood mononuclear cells into ischemic hibernating myocardium.Arterioscler Thromb Vasc Biol,2002,22(11):1804-1810.
    15. Fukuda K.Reprogramming of bone marrow mesenchymal stem cells into cardiomyocytes. C R Biol,2002,325(10):1027-1038.
    16. Chedrawy EG, Wang JS, Nguyen DM, et al.Incorporation and integration of implanted myogenic and stem cells into native myocardial fibers: anatomic basis for functional improvements.J Thorac Cardiovasc Surg,2002 ,124(3):584-590.
    17. Jiang XX,Zhang Y,Liu B,et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells.Blood,2005 ,105(10):4120-4126.
    1. Gottlieb DI. Large-scale sources of neural stem cells. Annu Rev.Neurosci, 200225:381-407.
    2. Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cellsof the adult mammalian central nervous system.Science,1992,255(5052): 1707-1710.
    3. Weiss S, Dunne C, Hewson J,et al. Multipotent CNS stem cells are present in the adult mammalian spinal cord and ventricular neuroaxis.J Neurosci,1996 ,16(23):7599-7609.
    4. Kabos P, Ehtesham M, Kabosova A, et al.Generation of neural progenitor cells from whole adult bone marrow. Exp Neurol,2002 ,178(2):288-293.
    5. Mezey E, Chandross KJ, Harta G, et al. Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow.Science., 2000,290(5497):1779-1782.
    6. Taupin P.Neural progenitor and stem cells in the adult central nervous system. Ann Acad Med Singapore,2006,35(11):814-820.
    7. Heine W, Conant K,Griffin JW,et al.Transplanted neural stem cells promote axonalregeneration through chronically denervated peripheral nerves.Exp Neurol,2004 , 189(2):231-240.
    8.王德华,徐建光,姜浩,等.神经干细胞移植再支配失神经骨骼肌的实验研究.中华手外科杂志,2004,20(2):96-99.
    9. Corti S, Locatelli F, Strazzer S, et al. Modulated generation of neuronal cells from bone marrow by expansion and mobilization of circulating stem cells with in vivo cytokine treatment.Exp Neuro,2002,177(2):443-452.
    10. Johansson CB,Momma S,Clarke DL,et al.Identification of a neural stem cell in the adult mammalian central nervous system.Cell,1999,96(1):25-34.
    11. Hulspas R, Tiarks C, Reilly J, et,al.In vitro cell density-dependent clonal growth of EGF-responsive murine neural progenitor cells under serum-free conditions.Exp Neurol,1997 148(1):147-156.
    12. Longo FM, Manthorpe M, Skaper SD, et,al. Neuronotrophic activities accumulate in vivo within silicone nerve regeneration chambers.Brain Res,1983 ,261(1):109-116.
    13. Tews DS, Goebel HH, Schneider I, et,al. DNA-fragmentation and expression ofapoptosis-related proteins in experimentally denervated and reinnervated rat facial muscle.Neuropathol Appl Neurobiol,1997, 23(2):141-149.
    14. Kawada H, Takizawa S, Takanashi T, Administration of hematopoietic cytokines in the subacute phase after cerebral infarction is effective for functional recovery facilitating proliferation of intrinsic neural stem/progenitor cells and transition of bone marrow-derived neuronal cells.Circulation, 2006,113(5):701-710.
    15. Hori J, Ng TF, Shatos M, et,al. Neural progenitor cells lack immunogenicity and resist destruction as allografts. Stem Cells, 2003,21(4): 405-416.
    1 Jiang G,Gu Y. The observation of transplanted embryonic motoneurons in the denervated muscles of adult rats.Chin Med J (Engl),1998 ,111(1):63-66.
    2 Craff MN,Zeballos JL,Johnson TS, et al.Embryonic stem cell-derived motor neurons preserve muscle after peripheral nerve injury. Plast Reconstr Surg,2007,119(1):235-245.
    3 Ortiz-Gonzalez XR, Keene CD, Verfaillie CM, et al.Neural induction of adult bone marrow and umbilical cord stem cells.Curr Neurovasc Res,2004,1(3):207-213.
    4 Krabbe C, Zimmer J, Meyer M. Neural transdifferentiation of mesenchymal stem cells--a critical review.APMIS.,2005,113(11-12):831-844.
    5 Neggy Rismanchi, Candace L ,Floyd, Robert F,et al. Cell death and long-term maintenance of neuron-like state after differentiation of rat bone marrow stromal cells: a comparison of protocols. Brain Res,2003, 991(1-2):46-55.
    6 Seri B, Garcia-Verdugo JM, McEwen BS,et al. Astrocytes give rise to new neurons in the adult mammalian hippocampus.J Neurosci, 2001,21(18):7153-60.
    7 Jiang Y, Henderson D, Blackstad M, et al.Neuroectodermal differentiation from mouse multipotent adult progenitor cells.Proc Natl Acad Sci U S A,2003,11854-11860.
    8 Sunderland S. Factors influencing the development and severity of the changes in denervated muscle . Nerve injuries and their repair. Churchill Livingstone, London, 1991, 247-257.
    9 Erb DE, Mora RJ, Bunge RP.Reinnervation of adult rat gastrocnemius muscle by embryonic motoneurons transplanted into the axotomized tibial nerve.Exp Neurol,1993,124(2):372-376.
    10姜广良,顾玉东,张丽银.胚胎运动神经元失神经骨骼肌内种植防治肌萎缩的研究.中华实验外科杂志,1998,15:364-36
    11 Ridet JL Malhotra SK, Privat A, et al.Reactive astrocytes: cellular and molecular cues to biological function Trends Neurosci,1997,20(12):570-577.
    1王拴科,洪光祥,王同光,等.细胞外ATP防治失神经肌肉萎缩的实验研究.中华手外科杂志,2002,18(1):43-45.
    2 Liu J, Lamb D, Chou MM, et al. Nerve Growth Factor-mediated Neurite Outgrowth via Regulation of Rab5.Mol Biol Cell,2007;18(4):1375-1384.
    3 Horwitz EM. Stem cell plasticity: the growing potential of cellular therapy. Arch Med Res,2003 ,34(6):600-606.
    4 Vieyra DS, Jackson KA, Goodell MA. Plasticity and tissue regenerative potential of bone marrow-derived cells.Stem Cell Rev,2005,1(1):65-69.
    5 Friedenstein AJ, Piatetzky-Shapiro II, Petrakova KV.Osteogenesis in transplants of bone marrow cells.J Embryol Exp Morphol,1966 ,16(3):381-390.
    6 Piersma AH, Brockbank KG, Ploemacher RE, et al.Characterization of fibroblastic stromal cells from murine bone marrow.Exp Hematol,1985,13(4):237-243.
    7 Owen M. Marrow stromal stem cells.J Cell Sci Suppl,1988,10:63-76.
    8 Azizi SA,Stokes D, Augelli BJ, et al. Engraftment and migration of human bone marrow stromal cells implanted in the brains of albino rats--similarities to astrocyte grafts.Proc Natl Acad Sci U S A., 1998 ,95(7):3908-3913.
    9王廷华,羊惠君,John W.McDonald.主编.干细胞理论与技术.第1版.北京:科学出版社,2005.
    10 Pittenger MF, Mackay AM, Beck SC, et al.Multilineage potential of adult human mesenchymal stem cells.Science,1999 ,284(5411):143-147.
    11 Satomura K, Derubeis AR, Fedarko NS, et al. Receptor tyrosine kinase expression in human bone marrow stromal cells.J Cell Physiol.,1998,177(3):426-438.
    12 Dawn B, Bolli R. Adult bone marrow-derived cells: regenerative potential, plasticity, and tissue commitment.Basic Res Cardiol.,2005 ,100(6):494-503.
    13 Simmons PJ, Gronthos S, Zannettino A, et al. Isolation, characterization and functional activity of human marrow stromal progenitors in hemopoiesis.Prog Clin Biol Res,1994,389:271-280.
    14 Gronthos S, Zannettino AC, Hay SJ, et al. Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow.J Cell Sci,2003 ,116(Pt 9):1827-1835.
    15 Ciulla MM, Lazzari L, Pacchiana R,et al.Homing of peripherally injected bone marrow cells in rat after experimental myocardial injury. Haematologica,2003,88(6):614-621.
    16 Fiedler J, Roderer G, Gunther KP, et al.BMP-2, BMP-4, and PDGF-bb stimulate chemotactic migration of primary human mesenchymal progenitor cells. J Cell Biochem,2002,87(3):305-312.
    17 Reyes M, Lund T, Lenvik T, et al. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells.Blood,2001 ,98(9):2615-2625.
    18 Friedenstein AJ, Piatetzky-Shapiro II, et al.Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol,1966,16(3):381-390
    19 Jaiswal N, Haynesworth SE, Caplan AI, et al.Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitro.J Cell Biochem. 1997 ,64(2):295-312.
    20 Brodke D, Pedrozo HA, Kapur TA, et al. Bone grafts prepared with selective cell retention technology heal canine segmental defects as effectively as autograft.J Orthop Res.2006 ,24(5):857-866.
    21 Barry F, Boynton RE, Liu B, et al. Chondrogenic differentiation of mesenchymal stem cells from bone marrow: differentiation-dependent gene expression of matrix components.Exp Cell Res,2001 ,268(2):189-200.
    22 Bosnakovski D, Mizuno M, Kim G, et al. Chondrogenic differentiation of bovine bone marrow mesenchymal stem cells (MSCs) in different hydrogels: influence of collagen type II extracellular matrix on MSC chondrogenesis.Biotechnol Bioeng,2006,93(6):1152-1163.
    23 Mackay AM, Beck SC, Murphy JM, et al. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow.Tissue Eng,1998,4(4):415-428.
    24 Robinson D, Nevo Z. Articular Cartilage Chondrocytes are more Advantageous for Generating Hyaline-like Cartilage than Mesenchymal Cells Isolated from Microfracture Repairs.Cell Tissue Bank,2001,2(1):23-30.
    25 Schutze N, Noth U, Schneidereit J, et al. Differential expression of CCN-family members in primary human bone marrow-derived mesenchymal stem cells during osteogenic, chondrogenic and adipogenic differentiation.Cell Commun Signal,2005 ,3(1):5.
    26 Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine.Muscle Nerve,1995,18(12):1417-1426.
    27 Makino S, Fukuda K, Miyoshi S, et al. Cardiomyocytes can be generated from marrow stromal cells in vitro.J Clin Invest,1999 ,103(5):697-705.
    28 Negishi Y, Kudo A, Obinata A,et al. Multipotency of a bone marrow stromal cell line, TBR31-2, established from ts-SV40 T antigen gene transgenic mice. Biochem Biophys Res Commun, 2000, 268(2):450-455
    29 Ferrari G, Cusella-De Angelis G, Coletta M, et al. Muscle regeneration by bone marrow-derived myogenic progenitors.Science,1998,279(5356):1528-1530.
    30 Wang JS, Shum-Tim D, Galipeau J, et al. Marrow stromal cells for cellular cardiomyoplasty: feasibility and potential clinical advantages. Thorac Cardiovasc Surg,2000,120(5):999-1005.
    31 Kopen GC, Prockop DJ, Phinney DG. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci U S A,1999,;96(19):10711-10716.
    32 Woodbury D, Schwarz EJ, Prockop DJ, et al. Adult rat and human bone marrow stromal cells differentiate into neurons.J Neurosci Res, 2000 ,61(4):364-370.
    33 Sanchez-Ramos J, Song S, Cardozo-Pelaez F, et al. Adult bone marrow stromal cells differentiate into neural cells in vitro.Exp Neurol,2000 ,164(2):247-256.
    34 Jiang Y, Jahagirdar BN, Reinhardt RL,et al.Pluripotency of mesenchymal stem cells derived from adult marrow. Nature,2002 ,418(6893):41-49.
    35 Brazelton TR, Rossi FM, Keshet GI,et al. From marrow to brain: expression of neuronal phenotypes in adult mice.Science,2000 ,290(5497):1775-1779.
    36 Hematti P, Sloand E, Carvallo C, et al. Absence of donor-derived keratinocyte stem cells in skin tissues cultured from patients after mobilized peripheral blood hematopoietic stem cell transplantation.Exp Hematol,2002,30:943-949.
    37 Badiavas EV, Abedi M, Butmarc J, et al.Participation of bone marrow derived cells in cutaneous wound healing. J Cell Physiol, 2003,196:245-250.
    38 Petersen BE, Bowen WC, Patrene KD, et al.Bone marrow as a potential source of hepaticoval cells. Science,1999,284:1168-1170.
    39 Schwartz RE, Reyes M, Koodie L, et al. Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells.J Clin Invest,2002,109:1291-1302.
    40 Myozaki M, Kamiyasu M, Miura T ,et al.Induction of autoimmune hepatitis and autoantibodies to liver antigens by neonatal thymectomy in mice.Clin Exp Immunol ,1996 ,104(1):133-143.
    41 Avital I, Inderbitzin D, Aoki T, et al. Isolation, characterization, and transplantation of bone marrow-derived hepatocyte stem cells.Biochem Biophys Res Commun,2001,288(1):156-164.
    42 Kotton DN, Ma BY, Cardoso WV, et al. Bone marrow-derived cells as progenitors of lung alveolarepithelium. Development,2001,128:5181-5188.
    43 Kale S, Karihaloo A, Clark PR,et al..Bone marrow stem cells contribute to repair of the ischemically injured renal tubule. J Clin Invest,2003,12:42-49.
    44 Ianus A, Holz GG, Theise ND,et al. In vivo derivation of glucose-competentpancreaticendocrine cells from bone marrow without evidence of cell fusion. J. Clin. Invest,2003,111:843-850.
    45 Krause DS, Theise ND, Collector MI, et al. Multiorgan, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell,2001,105:369-377.
    46 Koc ON, Day J, Nieder M, et al. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH).Bone Marrow Transplant,2002 ,30(4):215-222.
    47 Jennische E, Hansson HA. Regenerating skeletal muscle cells express insulin-like growth factor I.Acta Physiol Scand,1987,130(2):327-332.
    48 Schakman O, Thissen JP. Gene therapy with anabolic growth factors to prevent muscle atrophy.Curr Opin Clin Nutr Metab Care,2006 ,9(3):207-213.
    49 Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators.J Cell Biochem,2006 ,98(5):1076-1084.
    50 Stein TP, Wade CE. Protein turnover in atrophying muscle: from nutritional intervention to microarray expression analysis.Curr Opin Clin Nutr Metab Care,2003,6(1):95-102.
    51 Sunderland S. Factors influencing the development and severity of the changes in denervated muscle . Nerve injuries and their repair. Churchill Livingstone, London,1991,247-257.
    52 Kraft GH. Are fibrillation potentials and positive sharp waves the same? No. Muscle Nerve. 1996 Feb;19(2):216-20.
    53 Tews DS, Goebel HH, Schneider I, et,al. DNA-fragmentation and expression of apoptosis-related proteins in experimentally denervated and reinnervated rat facial muscle.Neuropathol Appl Neurobiol,1997, 23(2):141-149.
    54 Kamihata H, Matsubara H, Nishiue T, et al.Improvement of collateral perfusion and regional function by implantation of peripheral blood mononuclear cells into ischemic hibernating myocardium.Arterioscler Thromb Vasc Biol,2002,22(11):1804-1810.
    55 Berry MF, Engler AJWoo YJ, et al.Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance. Am J Physiol Heart Circ Physiol, 2006,290(6):H2196-2203.
    56 Chedrawy EG, Wang JS, Nguyen DM, et al.Incorporation and integration of implantedmyogenic and stem cells into native myocardial fibers: anatomic basis for functional improvements.J Thorac Cardiovasc Surg,2002 ,124(3):584-590.
    57 Park KI, Himes BT, Stieg PE, et al. Neural stem cells may be uniquely suited for combined gene therapy and cell replacement: Evidence from engraftment of Neurotrophin-3-expressing stem cells in hypoxic-ischemic brain injury.Exp Neurol,2006 ,199(1):179-190.
    58 Wislet-Gendebien S, Hans G, Leprince P,et al. Plasticity of cultured mesenchymal stem cells: switch from Nestin-positive to excitable neuron-like phenotype.Stem Cells,2005 ,23(3):392-402.
    59 Carvey PM, Ling ZD, Sortwell CE,et al. A clonal line of mesencephalic progenitor cells converted to dopamine neurons by hematopoietic cytokines: a source of cells for transplantation in Parkinson's disease.Exp Neurol,2001,171(1):98-108.
    60 Lang EM, Asan E, Plesnila N,et al. Motoneuron survival after C7 nerve root avulsion and replantation in the adult rabbit: effects of local ciliary neurotrophic factor and brain-derived neurotrophic factor application.Plast Reconstr Surg,2005,115(7):2042-2050.
    61 Park CH, Minn YK, Lee JY, et al. In vitro and in vivo analyses of human embryonic stem cell-derived dopamine neurons.J Neurochem,2005 ,92(5):1265-1276.
    62 Erb DE,Mora RJ,Bunge RP.Reinnervation of adult rat gastrocnemius muscle by embryonic motoneurons transplanted into the axotomized tibial nerve.Exp Neurol. 1993 ,Dec;124(2):372-6.
    63 Jiang G,Gu Y. The observation of transplanted embryonic motoneurons in the denervated muscles of adult rats.Chin Med J (Engl),1998 ,111(1):63-66.
    64王欢,顾玉东,徐建光,等.不同方法的感觉神经(元)营养失神经骨骼肌实验研究的疗效比较.中华手外科杂志,2000,16(1):49-52.
    65 Jiang Y, Henderson D, Blackstad M, et al.Neuroectodermal differentiation from mouse multipotent adult progenitor cells.Proc Natl Acad Sci U S A. 2003 ,Sep 30;100 Suppl1:11854-60.
    66 Herzog EL,Chai L,Krause DS. Plasticity of marrow-derived stem cells.Blood,2003,102(10):3483-3493.

© 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