胚胎干细胞在成体骨髓中长期存活的观察研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的胚胎干细胞(embryonic stem cells, ESCs)成为当今生命科学和生物技术研究的热点,这是因为它具有自我更新和多能分化的能力。多年来,“成体组织中是否存在胚胎干细胞”一直是一个很有争议的问题,很多实验室已报道可以从骨髓中分离出来ESCs样的多能干细胞,这些细胞都表达ESCs特异性标志Oct4基因,能像ESCs一样无限分裂繁殖,而且可以分化成几乎所有类型的细胞。但是对于这些原始细胞的来源和存在机制,人们并不清楚。因此,骨髓中是否存在ESCs就变成了一个很有争议的问题。
     之前的研究都是通过不同的方法从骨髓中分离、提取并培养这些ESCs样的干细胞,而我们首次通过相反的方式直接将小鼠ESCs注入到小鼠的骨髓腔里,通过观察ESCs在体内的分布生长情况,从而思考体内是否有这样一个适宜ESCs生存的环境。
     方法为了更好地追踪ESCs在体内的生长情况,我们新建两株带绿色荧光标记(green fluorescent protein, GFP)的ESCs——oGFP+ESCs(C57BL/6xOct4/EGFP转基因OG2小鼠);和aGFP+ESCs(129/SvxCAG/EGFP转基因C57BL/6-Tg小鼠)。oGFP+ESCs中GFP的表达受Oct4启动子调控,Oct4表达时GFP才表达;aGFP+ESCs中GFP的表达受chicken-beta-actin启动子调控,无论细胞分化与否,GFP均表达。为了避免了ESCs在体内其它器官的阻滞和损失,我们直接将ESCs通过胫骨注射植入到同源的C57BL/6小鼠(分照射组和非照射组)骨链腔内。
     移植后,通过双光子荧光显微镜来观察GFP+细胞在小鼠骨髓腔里存活的时间和状态。另在不同的时间点,我们从移植小鼠的骨髓腔里再次分离出这些GFP+的细胞,采用建立ES细胞系的方法使这些Oct4+-GFP+细胞建成稳定细胞系,并检测这些新建细胞系的功能。通过基因芯片检测来比较这些细胞系和母代ESCs在基因表达水平上的差异。另一方面,我们通过流式细胞学检测和体外培养的方式,分析移植的ESCs在骨髓这种造血微环境下的分化特征。
     结果通过双光子荧光显微镜观察发现100天后,骨髓中仍然存在Oct4-GFP表达的类似ESCs的细胞。大多数GFP+细胞位于移植侧胫骨的注射部位,也有少部分GFP+细胞位于注射骨对侧的骨髓腔内。在不同的时间点,我们从移植小鼠的骨髓腔里分离出这些Oct4+-GFP+的细胞,建成稳定细胞系。到目前为止,我们成功建立了8株细胞系,形态同ESCs。通过对这些Oct4+细胞系的功能性检测发现它们具有与ESCs相似的特性,但这种细胞系生成嵌合鼠的嵌合率很低,且不能生殖系传递。芯片结果显示这些细胞与母代ESCs比较,部分基因表达水平发生了改变,表观遗传学也发生了逐渐的变化。
     另外,移植的ESCs在骨髓腔内大多数分化成了非造血的细胞,形态多样,与骨质连接紧密,而仅有少部分分化成了造血细胞,并可进入血循环。结论1)我们的研究首次通过直接将ESCs移植入小鼠骨髓腔来证实了成体环境中长期存在ESCs的可能性,对以后干细胞的应用,在成体内的发育和分化研究具有一定的提示作用;
     2)由于原始的胚胎干细胞在体内可以长期存在,因此在使用ESCs或ESCs衍生物做细胞治疗或移植过程中,安全性一定要长期关注;
     3)胚胎干细胞在成体骨髓中存活一段时间后,逐渐发生了表观遗传学方面的变化,这种变化影响了ESCs的多能性改变,因此在以后细胞移植过程中,我们需要关注这些细胞表观遗传学方面的影响;
     4)ESCs在骨髓腔内存活一段时间后,大多数分化成了非造血细胞。我们的研究首次说明了ESCs在骨髓微环境的分化趋势,对ESCs的分化潜能和周围环境的关系以及ESCs的应用具有一定的提示作用。
Objective Embryonic stem cells (ESCs), derived from the inner cell mass of a blastocyst, possess two important characteristics:self-renewal and pluripotency toward all three embryonic germ layers. These remarkable characteristics make ESCs an attractive cell source in regenerative medicine. The important question regarding whether ESCs can survive in adult organs/tissues for the long-term remains unanswered. Bone marrow (BM) has been identified as a major source containing hematopoietic stem cells (HSCs) as well as non-hematopoietic stem cells, which include mesenchymal stem cells, multipotent adult progenitor cells, marrow-isolated adult multilineage inducible cells, and very small embryonic-like stem cells. Some of these non-hematopoietic bone-marrow-derived stem cells have been shown to have features in common with ESCs. Some express the Oct4 gene, and multipotent adult progenitor cells have been shown to differentiate into each of the three germ layer cells. However, the claim regarding the existence of ESC-like cells in adult tissues/organs such as bone marrow remains controversial largely due to the difficulty in isolating these extremely rare cells.
     In contrast to the previous studies that have examined this important yet unanswered question, we chose to approach it from the opposite direction by transplanting ESCs directly into the bone marrow to verify whether the bone marrow could provide a potential micro-environment for sustaining the pluripotent stem cells. A further rationale for carrying out our current study is the existing safety concerns regarding the potential of leftover or contaminated ESCs in therapeutic ESC products.
     Methods We approached this question by directly injecting mouse ESCs into syngeneic BM and then monitoring the survival and differentiation potential of the injected ESCs in BM based on the expression of GFP protein of the injected cells. To this end, an ESC line was derived from the Oct4-GFP transgenic mice in which GFP protein expression is driven by the Oct4 promoter, thereby indicating the status of pluripotency. As a control, an ESC line was also derived from the mice that constitutively express GFP that is driven by the chicken-β-Actin. The cells were injected via intra-tibia and then monitored at different time points after injection. The transplanted cells were visualized at several time points using two-photon microscopy to determine whether the bone marrow could sustain the pluripotent stem cells. Moreover, we further characterized the properties of the re-established cell lines recovered from the transplanted cells. To explore why the pluripotency of the re-derived cells are decreased, we compared the global gene-expression profiles of ESCs and the re-derived cell lines. To analyze the differentiation potential of ESCs after bone marrow transplantation, we examined the engrafted cells by flow cytometric analysis and the colony-forming unit-fibroblast assay.
     Results Despite the fact that some of the animals (28-29%in the irradiated hosts) developed tumors as expected, we intended to focus on the engrafted ESCs in the animals in which no tumor had occurred. We found that the Oct4+ cells could exist in recipient's BM for more than 100 days after transplantation as examined under the two-photon microscopy. Notably, most injected ESCs in BM differentiated into non-hematopoietic cells whereas few cells acquired hematopoietic cell surface markers. Interestingly, the engrafted cells were found in the contrary BM of the injected side as well as the blood circulation. We then harvested the Oct4+ cells in the BM at various time points after transplantation and attempted to re-establish the ESC lines. Eight ESC lines were recovered from a total of 141 irradiated recipients that were transplanted with the Oct4-driven GFP+ESCs. Those successfully recovered ESC lines from BM expressed known markers for embryonic pluripotency and were able to differentiate into three germ layers although germ line transmission is yet to be further defined. Gene expression profiling suggests time-dependent epigenetic alterations in the re-derived ESC lines as compared to their parental ESC line.
     Conclusions This study provides definitive evidence for long-term survival and differentiation of exogenous ESCs in adults. It has important implications for the understanding of stem cell development and maintenance, as well as for the safety evaluation of therapeutic ESC derivatives especially given the fact that rare un-differentiated ESCs can be contained in the final cellular products.
引文
[1]Takahashi K, Mitsui K, Yamanaka S. Role of ERas in promoting tumour-like properties in mouse embryonic stem cells. Nature.2003,423:541-545
    [2]Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell.2006,126:663-676
    [3]Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science.2007,318:1917-1920
    [4]Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell.2007,131:861-872
    [5]Kucia M, Halasa M, Wysoczynski M, et al. Morphological and molecular characterization of novel population of CXCR4+ SSEA-4+ Oct-4+ very small embryonic-like cells purified from human cord blood:preliminary report. Leukemia. 2007,21:297-303
    [6]Beltrami AP, Cesselli D, Bergamin N, et al. Multipotent cells can be generated in vitro from several adult human organs (heart, liver, and bone marrow). Blood. 2007,110:3438-3446
    [7]Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol.2000,109:235-242
    [8]Yu H, Fang D, Kumar SM, et al. Isolation of a novel population of multipotent adult stem cells from human hair follicles. Am J Pathol.2006,168:1879-1888
    [9]Ling TY, Kuo MD, Li CL, et al. Identification of pulmonary Oct-4+stem/progenitor cells and demonstration of their susceptibility to SARS coronavirus (SARS-CoV) infection in vitro. ProcNatl Acad Sci USA.2006,103:9530-9535
    [10]Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM. Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol.2002,30:896-904
    [11]Johnson J, Bagley J, Skaznik-Wikiel M, et al. Oocyte generation in adult mammalian ovaries by putative germ cells in bone marrow and peripheral blood. Cell.2005,122:303-315
    [12]Nayernia K, Lee JH, Drusenheimer N, et al. Derivation of male germ cells from bone marrow stem cells. Lab Invest.2006,86:654-663
    [13]Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970,3:393-403
    [14]Jiang Y, Jahagirdar BN, Reinhardt RL, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature.2002,418:41-49
    [15]D'Ippolito G, Diabira S, Howard GA, Menei P, Roos BA, Schiller PC. Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential. J Cell Sci. 2004,117:2971-2981
    [16]Kucia M, Reca R, Campbell FR, et al. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+stem cells identified in adult bone marrow. Leukemia. 2006,20:857-869
    [17]Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature.1981,292:154-156
    [18]Pannetier M, Feil R. Epigenetic stability of embryonic stem cells and developmental potential. Trends Biotechnol.2007,25:556-562
    [19]Schoonjans L, Kreemers V, Danloy S, Moreadith RW, Laroche Y, Collen D. Improved generation of germline-competent embryonic stem cell lines from inbred mouse strains. Stem Cells.2003,21:90-97
    [20]Xie Y, Yin T, Wiegraebe W, et al. Detection of functional haematopoietic stem cell niche using real-time imaging. Nature.2009,457:97-101
    [21]Zhan Y, Zhao Y. Hematopoietic stem cell transplant in mice by intra-femoral injection. Methods Mol Biol.2008,430:161-169
    [22]Lo Celso C, Fleming HE, Wu JW, et al. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature.2009,457:92-96
    [23]Haylock DN, Williams B, Johnston HM, et al. Hemopoietic stem cells with higher hemopoietic potential reside at the bone marrow endosteum. Stem Cells.2007,25:1062-1069
    [24]Erdo F, Buhrle C, Blunk J, et al. Host-dependent tumorigenesis of embryonic stem cell transplantation in experimental stroke. J Cereb Blood Flow Metab.2003,23:780-785
    [25]Menard C, Hagege AA, Agbulut O, et al. Transplantation of cardiac-committed mouse embryonic stem cells to infarcted sheep myocardium:a preclinical study. Lancet. 2005,366:1005-1012
    [26]Acharya MM, Christie LA, Lan ML, et al. Rescue of radiation-induced cognitive impairment through cranial transplantation of human embryonic stem cells. Proc Natl Acad Sci USA.2009,106:19150-19155
    [27]Reik W, Walter J. Genomic imprinting:parental influence on the genome. Nat Rev Genet.2001,2:21-32
    [28]Yamazaki Y, Mann MR, Lee SS, et al. Reprogramming of primordial germ cells begins before migration into the genital ridge, making these cells inadequate donors for reproductive cloning. Proc Natl Acad Sci USA.2003,100:12207-12212
    [29]Horii T, Kimura M, Morita S, Nagao Y, Hatada I. Loss of genomic imprinting in mouse parthenogenetic embryonic stem cells. Stem Cells.2008,26:79-88
    [30]Delaval K, Feil R. Epigenetic regulation of mammalian genomic imprinting. Curr Opin Genet Dev.2004,14:188-195
    [31]Kobayashi H, Suda C, Abe T, Kohara Y, Ikemura T, Sasaki H. Bisulfite sequencing and dinucleotide content analysis of 15 imprinted mouse differentially methylated regions (DMRs):paternally methylated DMRs contain less CpGs than maternally methylated DMRs. Cytogenet Genome Res.2006,113:130-137
    [32]Mager J, Montgomery ND, de Villena FP, Magnuson T. Genome imprinting regulated by the mouse Polycomb group protein Eed. Nat Genet.2003,33:502-507
    [33]Fournier C, Goto Y, Ballestar E, et al. Allele-specific histone lysine methylation marks regulatory regions at imprinted mouse genes. EMBO J.2002,21:6560-6570
    [34]Ratajczak MZ, Machalinski B, Wojakowski W, Ratajczak J, Kucia M. A hypothesis for an embryonic origin of pluripotent Oct-4(+) stem cells in adult bone marrow and other tissues. Leukemia.2007,21:860-867
    [35]Shin DM, Zuba-Surma EK, Wu W, et al. Novel epigenetic mechanisms that control pluripotency and quiescence of adult bone marrow-derived Oct4(+) very small embryonic-like stem cells. Leukemia.2009,23:2042-2051
    [36]McCullough KA, Waits CA, Garimella R, Tague SE, Sipe JB, Anderson HC. Immunohistochemical localization of bone morphogenetic proteins (BMPs) 2,4,6, and 7 during induced heterotopic bone formation. J Orthop Res.2007,25:465-472
    [37]Ying QL, Nichols J, Chambers I, Smith A. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell.2003,115:281-292
    [38]Ying QL, Wray J, Nichols J, et al. The ground state of embryonic stem cell self-renewal. Nature.2008,453:519-523
    [39]Davey RE, Zandstra PW. Spatial organization of embryonic stem cell responsiveness to autocrine gp130 ligands reveals an autoregulatory stem cell niche. Stem Cells. 2006,24:2538-2548
    [40]Shen J, Qu CK. In vitro hematopoietic differentiation of murine embryonic stem cells. Methods Mol Biol.2008,430:103-118
    [41]Cantarelli E, Melzi R, Mercalli A, et al. Bone marrow as an alternative site for islet transplantation. Blood.2009,114:4566-4574
    [42]Anjos-Afonso F, Bonnet D. Nonhematopoietic/endothelial SSEA-1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood.2007,109:1298-1306
    [43]Dexter TM, Allen TD, Lajtha LG Conditions controlling the proliferation of haemopoietic stem cells in vitro. J Cell Physiol.1977,91:335-344
    [44]Verfaillie CM. Soluble factor(s) produced by human bone marrow stroma increase cytokine-induced proliferation and maturation of primitive hematopoietic progenitors while preventing their terminal differentiation. Blood.1993,82:2045-2053
    [45]Wilson A, Trumpp A. Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol. 2006,6:93-106
    [46]Paris DB, Stout TA. Equine embryos and embryonic stem cells:Defining reliable markers of pluripotency. Theriogenology.2010
    [47]Cao F, Lin S, Xie X, et al. In vivo visualization of embryonic stem cell survival, proliferation, and migration after cardiac delivery. Circulation.2006,113:1005-1014
    [48]Keirstead HS, Nistor G, Bernal G, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci.2005,25:4694-4705
    [49]Fujikawa T, Oh SH, Pi L, Hatch HM, Shupe T, Petersen BE. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathol.2005,166:1781-1791
    [50]Crisostomo PR, Abarbanell AM, Wang M, Lahm T, Wang Y, Meldrum DR. Embryonic stem cells attenuate myocardial dysfunction and inflammation after surgical global ischemia via paracrine actions. Am J Physiol Heart Circ Physiol.2008,295:H 1726-1735
    [51]Zhang F, Pasumarthi KB. Embryonic stem cell transplantation:promise and progress in the treatment of heart disease. BioDrugs.2008,22:361-374
    [52]McKinney-Freeman S, Daley G. Derivation of hematopoietic stem cells from murine embryonic stem cells. J Vis Exp.2007:162
    [53]Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem.1997,64:278-294
    [54]Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO, Pittenger MF. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng.1998,4:415-428
    [55]Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science.1999,284:143-147
    [56]Short B, Brouard N, Driessen R, Simmons PJ. Prospective isolation of stromal progenitor cells from mouse BM. Cytotherapy.2001,3:407-408
    [57]Short B, Brouard N, Occhiodoro-Scott T, Ramakrishnan A, Simmons PJ. Mesenchymal stem cells. Arch Med Res.2003,34:565-571
    [58]Phinney DG, Kopen G, Isaacson RL, Prockop DJ. Plastic adherent stromal cells from the bone marrow of commonly used strains of inbred mice:variations in yield, growth, and differentiation. J Cell Biochem.1999,72:570-585
    [59]Anjos-Afonso F, Siapati EK, Bonnet D. In vivo contribution of murine mesenchymal stem cells into multiple cell-types under minimal damage conditions. J Cell Sci. 2004,117:5655-5664
    [1]Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970,3:393-403
    [2]Phinney DG, Prockop DJ. Concise review:mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair--current views. Stem Cells. 2007,25:2896-2902
    [3]Badillo AT, Redden RA, Zhang L, Doolin EJ, Liechty KW. Treatment of diabetic wounds with fetal murine mesenchymal stromal cells enhances wound closure. Cell Tissue Res.2007,329:301-311
    [4]Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng.2001,7:211-228
    [5]Williams JT, Southerland SS, Souza J, Calcutt AF, Cartledge RG. Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes. Am Surg.1999,65:22-26
    [6]In't Anker PS, Scherjon SA, Kleijburg-van der Keur C, et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood. 2003,102:1548-1549
    [7]Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol.2000,109:235-242
    [8]Bieback K, Kluter H. Mesenchymal stromal cells from umbilical cord blood. Curr Stem Cell Res Ther. 2007,2:310-323
    [9]Miyahara Y, Nagaya N, Kataoka M, et al. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med.2006,12:459-465
    [10]Kondo T, Johnson SA, Yoder MC, Romand R, Hashino E. Sonic hedgehog and retinoic acid synergistically promote sensory fate specification from bone marrow-derived pluripotent stem cells. Proc Natl Acad Sci USA.2005,102:4789-4794
    [11]Li H, Fu X, Ouyang Y, Cai C, Wang J, Sun T. Adult bone-marrow-derived mesenchymal stem cells contribute to wound healing of skin appendages. Cell Tissue Res. 2006,326:725-736
    [12]Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells.2007,25:2648-2659
    [13]Javazon EH, Keswani SG, Badillo AT, et al. Enhanced epithelial gap closure and increased angiogenesis in wounds of diabetic mice treated with adult murine bone marrow stromal progenitor cells. Wound Repair Regen.2007,15:350-359
    [14]Herdrich BJ, Lind RC, Liechty KW. Multipotent adult progenitor cells:their role in wound healing and the treatment of dermal wounds. Cytotherapy.2008,10:543-550
    [15]Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004,363:1439-1441
    [16]Le Blanc K. Mesenchymal stromal cells:Tissue repair and immune modulation. Cytotherapy.2006,8:559-561
    [17]Stagg J. Immune regulation by mesenchymal stem cells:two sides to the coin. Tissue Antigens.2007,69:1-9
    [18]Morrison SJ, Weissman IL. The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype. Immunity.1994,1:661-673
    [19]Osawa M, Hanada K, Hamada H, Nakauchi H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science. 1996,273:242-245
    [20]Kiel MJ, Yilmaz OH, Iwashita T, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell.2005,121:1109-1121
    [21]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
    [22]Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, Prockop DJ. 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
    [23]Morikawa S, Mabuchi Y, Kubota Y, et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med.2009,206:2483-2496
    [24]Reyes M, Lund T, Lenvik T, Aguiar D, Koodie L, Verfaillie CM. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood. 2001,98:2615-2625
    [25]Jiang Y, Jahagirdar BN, Reinhardt RL, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature.2002,418:41-49
    [26]Zeng L, Rahrmann E, Hu Q, et al. Multipotent adult progenitor cells from swine bone marrow. Stem Cells.2006,24:2355-2366
    [27]Ulloa-Montoya F, Kidder BL, Pauwelyn KA, et al. Comparative transcriptome analysis of embryonic and adult stem cells with extended and limited differentiation capacity. Genome Biol.2007,8:R163
    [28]Qu-Petersen Z, Deasy B, Jankowski R, et al. Identification of a novel population of muscle stem cells in mice:potential for muscle regeneration. J Cell Biol.2002,157:851-864
    [29]Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM. Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol.2002,30:896-904
    [30]Seaberg RM, Smukler SR, Kieffer TJ, et al. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol.2004,22:1115-1124
    [31]Bickenbach JR, Stern MM, Grinnell KL, Manuel A, Chinnathambi S. Epidermal stem cells have the potential to assist in healing damaged tissues. J Investig Dermatol Symp Proc. 2006,11:118-123
    [32]Jiang Y, Henderson D, Blackstad M, Chen A, Miller RF, Verfaillie CM. Neuroectodermal differentiation from mouse multipotent adult progenitor cells. Proc Natl Acad Sci USA.2003,100 Suppl 1:11854-11860
    [33]Keene CD, Ortiz-Gonzalez XR, Jiang Y, Largaespada DA, Verfaillie CM, Low WC. Neural differentiation and incorporation of bone marrow-derived multipotent adult progenitor cells after single cell transplantation into blastocyst stage mouse embryos. Cell Transplant.2003,12:201-213
    [34]Yasuhara T, Hara K, Maki M, et al. Intravenous grafts recapitulate the neurorestoration afforded by intracerebrally delivered multipotent adult progenitor cells in neonatal hypoxic-ischemic rats. J Cereb Blood Flow Metab.2008,28:1804-1810
    [35]Yasuhara T, Matsukawa N, Yu G, et al. Transplantation of cryopreserved human bone marrow-derived multipotent adult progenitor cells for neonatal hypoxic-ischemic injury: targeting the hippocampus. Rev Neurosci.2006,17:215-225
    [36]Chen CW, Boiteau RM, Lai WF, Barger SW, Cataldo AM. sAPPalpha enhances the transdifferentiation of adult bone marrow progenitor cells to neuronal phenotypes. Curr Alzheimer Res.2006,3:63-70
    [37]Tang Y, Yasuhara T, Hara K, et al. Transplantation of bone marrow-derived stem cells:a promising therapy for stroke. Cell Transplant.2007,16:159-169
    [38]Pelacho B, Nakamura Y, Zhang J, et al. Multipotent adult progenitor cell transplantation increases vascularity and improves left ventricular function after myocardial infarction. J Tissue Eng Regen Med.2007,1:51-59
    [39]Serafini M, Dylla SJ, Oki M, et al. Hematopoietic reconstitution by multipotent adult progenitor cells:precursors to long-term hematopoietic stem cells. J Exp Med. 2007,204:129-139
    [40]Ross JJ, Hong Z, Willenbring B, et al. Cytokine-induced differentiation of multipotent adult progenitor cells into functional smooth muscle cells. J Clin Invest. 2006,116:3139-3149
    [41]Highfill SL, Kelly RM, O'Shaughnessy MJ, et al. Multipotent adult progenitor cells can suppress graft-versus-host disease via prostaglandin E2 synthesis and only if localized to sites of allopriming. Blood.2009,114:693-701
    [42]Kuci S, Wessels JT, Buhring HJ, et al. Identification of a novel class of human adherent CD34-stem cells that give rise to SCID-repopulating cells. Blood.2003,101:869-876
    [43]Young HE, Steele TA, Bray RA, et al. Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors. Anat Rec.2001,264:51-62
    [44]Toma JG, Akhavan M, Fernandes KJ, et al. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol.2001,3:778-784
    [45]D'Ippolito G, Diabira S, Howard GA, Menei P, Roos BA, Schiller PC. Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential. J Cell Sci. 2004,117:2971-2981
    [46]D'Ippolito G, Howard GA, Roos BA, Schiller PC. Sustained stromal stem cell self-renewal and osteoblastic differentiation during aging. Rejuvenation Res.2006,9:10-19
    [47]Tatard VM, D'Ippolito G, Diabira S, et al. Neurotrophin-directed differentiation of human adult marrow stromal cells to dopaminergic-like neurons. Bone.2007,40:360-373
    [48]Kucia M, Reca R, Campbell FR, et al. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-l(+)Oct-4+stem cells identified in adult bone marrow. Leukemia. 2006,20:857-869
    [49]Kucia M, Halasa M, Wysoczynski M, et al. Morphological and molecular characterization of novel population of CXCR4+ SSEA-4+ Oct-4+ very small embryonic-like cells purified from human cord blood:preliminary report. Leukemia. 2007,21:297-303
    [50]Ratajczak MZ, Machalinski B, Wojakowski W, Ratajczak J, Kucia M. A hypothesis for an embryonic origin of pluripotent Oct-4(+) stem cells in adult bone marrow and other tissues. Leukemia.2007,21:860-867
    [51]Kucia M, Zhang YP, Reca R, et al. Cells enriched in markers of neural tissue-committed stem cells reside in the bone marrow and are mobilized into the peripheral blood following stroke. Leukemia.2006,20:18-28
    [52]Paczkowska E, Kucia M, Koziarska D, et al. Clinical evidence that very small embryonic-like stem cells are mobilized into peripheral blood in patients after stroke. Stroke. 2009,40:1237-1244
    [53]Wojakowski W, Tendera M, Kucia M, et al. Mobilization of bone marrow-derived Oct-4+ SSEA-4+ very small embryonic-like stem cells in patients with acute myocardial infarction. J Am Coll Cardiol.2009,53:1-9
    [54]Machalinska A, Baumert B, Kuprjanowicz L, Wiszniewska B, Karczewicz D, Machalinski B. Potential application of adult stem cells in retinal repair--challenge for regenerative medicine. Curr Eye Res.2009,34:748-760
    [55]Zuba-Surma EK, Wu W, Ratajczak J, Kucia M, Ratajczak MZ. Very small embryonic-like stem cells in adult tissues-potential implications for aging. Mech Ageing Dev. 2009,130:58-66
    [56]Anjos-Afonso F, Bonnet D. Nonhematopoietic/endothelial SSEA-1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood.2007,109:1298-1306
    [57]Kucia M, Wysoczynski M, Ratajczak J, Ratajczak MZ. Identification of very small embryonic like (VSEL) stem cells in bone marrow. Cell Tissue Res.2008,331:125-134
    [58]Nagasawa T. A chemokine, SDF-1/PBSF, and its receptor, CXC chemokine receptor 4, as mediators of hematopoiesis. Int J Hematol.2000,72:408-411
    [59]Taichman R, Reilly M, Verma R, Ehrenman K, Emerson S. Hepatocyte growth factor is secreted by osteoblasts and cooperatively permits the survival of haematopoietic progenitors. Br J Haematol.2001,112:438-448
    [60]Kucia M, Wu W, Ratajczak MZ. Bone marrow-derived very small embryonic-like stem cells:their developmental origin and biological significance. Dev Dyn.2007,236:3309-3320
    [61]Sakihama H, Masunaga T, Yamashita K, et al. Stromal cell-derived factor-1 and CXCR4 interaction is critical for development of transplant arteriosclerosis. Circulation. 2004,110:2924-2930
    [62]LaBarge MA, Blau HM. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell. 2002,111:589-601
    [63]Kucia M, Dawn B, Hunt G, et al. Cells expressing early cardiac markers reside in the bone marrow and are mobilized into the peripheral blood after myocardial infarction. Circ Res.2004,95:1191-1199
    [64]Wojakowski W, Tendera M, Michalowska A, et al. Mobilization of CD34/CXCR4+, CD34/CD117+, c-met+stem cells, and mononuclear cells expressing early cardiac, muscle, and endothelial markers into peripheral blood in patients with acute myocardial infarction. Circulation.2004,110:3213-3220
    [65]Gomperts BN, Belperio JA, Rao PN, et al. Circulating progenitor epithelial cells traffic via CXCR4/CXCL12 in response to airway injury. J Immunol.2006,176:1916-1927
    [66]Aranda P, Agirre X, Ballestar E, et al. Epigenetic signatures associated with different levels of differentiation potential in human stem cells. PLoS One.2009,4:e7809