用户名: 密码: 验证码:
MicroRNAs调控人脂肪源性干细胞成骨分化及ODN影响其调控的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本实验的目的是识别miRNAs是人脂肪源性干细胞(human Adipose–Derived Stem Cells,hADSCs)成骨分化的一个调控因子,探讨对成骨分化有调节作用的寡核苷酸(oligodeoxynucleotide,ODN)是否通过调控miRNAs的表达从而调节成骨分化。
     从人脂肪抽吸物中分离hADSCs,体外培养扩增及鉴定。对hADSCs成骨分化的效率进行评估。检测miRNA-196a、microRNA-133a在hADSCs成骨分化中的表达以及Runx2、HOXC8的蛋白表达。将ODN C2及C12加入hADSCs成骨诱导培养基中诱导hADSCs成骨分化,以成骨诱导培养基诱导的hADSCs作为对照。检测三组成骨能力的变化,miRNA-196a、microRNA-133a表达的变化,Runx2、HOXC8蛋白表达的变化。
     结果证明hADSCs有极强的向成骨细胞分化的能力。miRNA-196a的表达在hADSCs成骨分化的过程中增强,伴随着HOXC8蛋白表达降低。提示miRNA-196a在hADSCs成骨分化中有重要作用,它的作用是通过HOXC8这一靶点介导的。通过本实验,miRNA-133a对成骨分化是否有调控作用尚不十分明确。本实验结果显示ODN C2及C12对成骨有抑制作用,是通过下调miRNA-196a表达水平,增加HOXC8的蛋白表达来实现的。
     本实验研究证明miRNAs是hADSCs成骨分化的调控因子;首次将ODN应用于hADSCs的成骨分化过程中,显示其对hADSCs的成骨分化有抑制作用;并证明ODN C2及C12对成骨的抑制作用是通过下调miRNA-196a表达水平,增加HOXC8的蛋白表达实现的。为hADSCs应用于临床作为组织工程骨的种子细胞及探索对成骨分化有影响的药物开发提供新的思路。
Human Adipose–Derived Stem Cells (hADSCs) are excellent candidates for cell-based therapeutic strategies to regenerate injured tissue. Although hADSCs can be isolated from Adipose Tissue and directed to differentiate by means of an osteogenic pathway, the regulation of cell-fate determination is not well understood. MicroRNAs (miRNAs) are a class of small (~22 nt), noncoding RNAs that regulate gene expression at a post-transcriptional level. Recently, it has been shown that miRNAs have an influence on the complexity of the stemness state either by inhibiting the translation or by stimulating the degradation of target mRNAs. Luzi et al. reported that miR-26a inhibited osteogenic differentiation in hADSCs by targeting SMAD1. Oligodeoxynucleotides(ODNs)are a class of small (less than one hundred nucleotides), single strand DNAs that mimic bacterial or human DNA. DNA is a kind of biomacromolecule which is composed of nucleotides; almost all the genetic information determining biological features is stored in DNA molecule. The sequence character and modified style of DNA varies significantly between different species. Recent studies demonstrate that DNA has some special biological activities in addition to carrying and transmitting genetic information, for example, certain DNA with specific sequence and modification has stimulatory or inhibitory effect on the immune system. Some DNA which has specific sequence and modification can be used for the prevention and treatment of diseases. CpG ODN, upon TLR9 ligation, modulates osteoclastogenesis via interactions with osteoclast precursors or induction of osteoblasts osteoclastogenic activity. Some CpG ODNs affect the proliferation of hADSCs and decrease the osteogenic differentiation. Recently, the members of our research group have shown that ODNs with special sequence characteristic can significantly promote the differentiation of rat BMSCs to osteoblasts. The aim of this study is to determine whether microRNAs serve as regulators of the osteogenic differentiation of hADSCs and whether the osteogenic differentiation of hADSCs is affected by ODNs through its regulating of microRNAs expression. This study includes four aspects:
     1. Cell cultures and osteogenic differentiation of hADSCs
     hADSCs were isolated from adipose tissue obtained from the subcutaneous abdominal depot undergoing elective abdominoplasty in accordance with a protocol approved by the Institutional Review Board for human studies. The isolation procedure was according to the methods described in the previous studies of Zuk’. All the cells used in the experiments were in three passages. The hADSCs were cultured and amplified in vitro and were identified by cell phenotype detection. Multi-lineage differentiation potential was assessed by histological staining such as oil red O for lipid accumulation and alizarin red S for mineralization nodules during adipogenic and osteogenic induction. Osteogenic differentiation induction group was divided into five subgroups: 0, 7, 14, 21 and 28 d groups. Alizarin red S staining, Van Gieson collagen fiber staining and ColⅠimmunofluorescence were performed to detect the expression of ColⅠ. FV Viewer 1.7 software was employed to analyze the date. The positive stainning of oil red O was found 14 d after adipogenic induction. For the osteogenic induction groups, the calcified nodules and the collagen fiber red staining was found in 21 d group. The expression of ColⅠwas found in 21 d group for most cells and was found strongly positive expression in 28 d group. Compared with 0 d group, the expression of ColⅠin osteogenic induction group was significantly increased from 7 d to 28 d(P<0.05). This aspect of the study has shown that osteogenic induction of hADSCs, most of the cells have osteogenic capability and the hADSCs have outstanding capability of osteoinductive and osteogenesis. hADSCs may be useful in future clinical cell-based therapy for bone regeneration engineering.
     2. The expression of microRNAs and osteoblastic marker genes of hADSCs during osteogenic differentiation
     Osteogenic differentiation of hADSCs was induced for 28 days. MiRNAs were prepared at the indicated times and relative real-time PCR were performed to detect the miRNAs. U6 was used as a loading control and for normalization. The relative quantities of the amplified products were determined with an image analyzer. The data represent the mean±SE of the relative ratio of the miRNAs signal of the corresponding samples (n = 3). The expression of Runx2 and ColⅠmRNA was evaluated after the induction by simultaneous monitoring the expression of genes involved in osteoblastic differentiation by relative real-time RT-PCR, normalized toβ-actin. Relative levels are expressed setting 0 time to 1. MiR-196a expression was increased and miR-133a expression up-regulated to 7 d, then decreased during osteogenic differentiation of hADSCs. Runx2 mRNA expression was increased and had a significant increase at 7 d during hADSCs differentiation. ColⅠmRNA had two peak at 3 d and 14 d, and had a significant decrease at 7 d during hADSCs differentiation. Our data indicate that miR-196a plays an important role in hADSCs osteogenic differentiation and miR-133a may also play a role in hADSCs osteogenic differentiation. The osteoblasts differentiated from hADSCs have osteoblastic potential to express Runx2 and ColⅠmRNA.
     3. The protein expression of targets of microRNAs and osteoblastic markers of hADSCs during osteogenic differentiation
     Total protein extracts were prepared at the indicated times. Runx2 and HOXC8 proteins expression were evaluated by Western blotting analysis. GAPDH was used as the standard protein. The protein expression of Runx2, served as markers for osteogenic differentiation, was observed at all time-points in osteo-induced hADSCs. Furthermore, Runx2 basal expression was observed in noninduced hADSCs. Runx2 proteins expression was increased and had a significant increase at 7 d during hADSCs differentiation. HOXC8 protein expression was down-regulated during hADSCs osteogenic differentiation, whereas expression of miR-196a was complementary to that of HOXC8. Our data indicate that miR-196a plays an important role in hADSCs osteogenic differentiation and proliferation, which may be mediated through its predicted target, HOXC8.
     4. The effect of ODN C2 and ODN C12 on microRNAs modulation of hADSCs osteogenic differentiation
     To investigate the effect of ODN addition during osteogenic differentiation induction, ODN C2, ODN C12 was added to the osteogenic medium and hADSCs osteogenic induction were performed in three groups: ODN C2 added to osteogenic medium( I+C2) group, ODN C12 added to osteogenic medium( I+C12) group, osteogenic medium(I )group. The effect was evaluated by means of histological observation, genomic and protein measurement. Alizarin red S staining and ColⅠimmunofluorescence were used as well as direct observation. The expression of Runx2 and ColⅠmRNA were evaluated by relative real-time RT-PCR. ALP proteins expression was evaluated by ALP Substrate measurement and Runx2 proteins expression was evaluated by Western blotting analysis. MiRNAs were prepared at the indicated times of the three groups and relative real-time PCR were performed to detect miR-196a and miR-133a expression. HOXC8 protein expressions were evaluated by Western blotting analysis. The calcified nodules and the ColⅠimmunofluorescence of I+C2, I+C12 group were less than I group. The Runx2 and ColⅠmRNA of I+C2, I+C12 group were less than I group. The miR-196a and miR-133a expression of I+C2, I+C12 group were less than I group in 3, 7, 14, 21 d. The HOXC8 protein expression of I+C2, I+C12 group were more than I group. ODN C2, ODN C12 promoted the proliferation of hADSCs and decreased the osteogenic differentiation. ODN C2, ODN C12 decreased the expression of miR-196a and miR-133a during hADSCs differentiation. ODN C2, ODN C12 down-regulate the osteogenic differentiation of hADSCs by repressing the expression of miR-196a and thereby releasing the target HOXC8 protein expression.
     Taken together, our results suggest that hADSCs cultured in vitro have outstanding capability of osteoinductive and osteogenesis. HADSCs may be useful in future clinical cell-based therapy for bone regeneration engineering. Our data indicate that miR-196a plays a role in hADSC osteogenic differentiation, which may be mediated through its predicted target, HOXC8. MiR-133a plays a role in hADSCs differentiation. ODN C2, ODN C12 promoted the proliferation of hADSCs and decreased the osteogenic differentiation. ODN C2, ODN C12 down-regulate the osteogenic differentiation of hADSCs by repressing the expression of miR-196a and thereby releasing the target HOXC8 protein expression.
     The innovation of this research is that the expression of miR-196a and HOXC8 has been monitored consecutively during 28 days, a complete period of hADSCs osteogenic differentiation. The first time that ODN C2, ODN C12 has been added to the osteogenic medium to investigate the influence on hADSCs osteogenic differentiation and miRNAs expression. Our results suggest that miRNAs functions as a regulator of osteogenic differentiation by repressing target gene expression. It is suggested that some drugs, like ODN with special sequence, inhibited osteogenic differentiation by regulating miRNAs expression. Elucidation of the molecular mechanisms guiding human adipose–derived stem cells (hADSCs) differentiation and obtaining drugs regulated this process are of extreme importance for improving the treatment of bone-related diseases such as alveolar bone defects, maxillofacial defects, periodontitis and osteoporosis.
引文
[1]Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998 Nov 6;282(5391):1145-1147.
    [2]Hwang WS, Ryu YJ, Park JH, et al. Evidence of a pluripotent human embryonic stem cell line derived from a cloned blastocyst. Science. 2004 Mar 12;303(5664):1669-1674.
    [3]Maitra A, Arking DE, Shivapurkar N, et al. Genomic alterations in cultured human embryonic stem cells. Nat Genet. 2005 Oct;37(10):1099-1103.
    [4]Fathi A, Pakzad M, Taei A, et al. Comparative proteome and transcriptome analyses of embryonic stem cells during embryoid body-based differentiation. Proteomics. 2009 Nov;9(21):4859-4870.
    [5]Valbuena D, Sanchez-Luengo S, Galan A, et al. Efficient method for slow cryopreservation of human embryonic stem cells in xeno-free conditions. Reprod Biomed Online. 2008 Jul;17(1):127-135.
    [6]Almeida-Porada G, Porada C, Zanjani ED. Adult stem cell plasticity and methods of detection. Rev Clin Exp Hematol. 2001 Mar;5(1):26-41.
    [7]Toma JG, Akhavan M, Fernandes KJ, et al. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol. 2001 Sep;3(9):778-784.
    [8]Barria E, Mikels A, Haas M. Maintenance and self-renewal of long-term reconstituting hematopoietic stem cells supported by amniotic fluid. Stem Cells Dev. 2004 Oct;13(5): 548-562.
    [9]Prusa AR, Marton E, Rosner M, et al. Neurogenic cells in human amniotic fluid. Am J Obstet Gynecol. 2004 Jul;191(1):309-314.
    [10]De Gemmis P, Lapucci C, Bertelli M, et al. A real-time PCR approach to evaluate adipogenic potential of amniotic fluid-derived human mesenchymal stem cells. Stem Cells Dev. 2006 Oct;15(5):719-728.
    [11]De Coppi P, Callegari A, Chiavegato A, et al. Amniotic fluid and bone marrow derived mesenchymal stem cells can be converted to smooth muscle cells in the cryo-injured rat bladder and prevent compensatory hypertrophy of surviving smooth muscle cells. J Urol. 2007 Jan;177(1):369-376.
    [12]Kolambkar YM, Peister A, Soker S, et al. Chondrogenic differentiation of amniotic fluid-derived stem cells. J Mol Histol. 2007 Oct;38(5):405-413.
    [13]Perin L, Giuliani S, Jin D, et al. Renal differentiation of amniotic fluid stem cells. Cell Prolif. 2007 Dec;40(6):936-948.
    [14]Laughlin MJ. Umbilical cord blood for allogeneic transplantation in children and adults. Bone Marrow Transplant. 2001 Jan;27(1):1-6.
    [15]Hurlbut W, Doerflinger R. Can a morally acceptable way be found to obtain embryonic stem cells? Origins. 2004 Dec 16;34(27):429-433.
    [16]Bianchi DW, Zickwolf GK, Weil GJ, et al. Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc Natl Acad Sci U S A. 1996 Jan 23;93(2):705-708.
    [17]Gang EJ, Jeong JA, Hong SH, et al. Skeletal myogenic differentiation of mesenchymal stem cells isolated from human umbilical cord blood. Stem Cells. 2004; 22(4):617-624.
    [18]Warnke PH, Springer IN, Wiltfang J, et al. Growth and transplantation of a custom vascularised bone graft in a man. Lancet. 2004 Aug 28-Sep 3;364(9436):766-770.
    [19]Young HE, Black AC, Jr. Adult stem cells. Anat Rec A Discov Mol Cell Evol Biol. 2004 Jan;276(1):75-102.
    [20]Parker GC, Anastassova-Kristeva M, Broxmeyer HE, et al. Stem cells: shibboleths of development. Stem Cells Dev. 2004 Dec;13(6):579-584.
    [21]Pompilio G, Cannata A, Peccatori F, et al. Autologous peripheral blood stem cell transplantation for myocardial regeneration: a novel strategy for cell collection and surgical injection. Ann Thorac Surg. 2004 Nov;78(5):1808-1812.
    [22]Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997 Apr 4;276(5309):71-74.
    [23]Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001 Apr;7(2):211-228.
    [24]Katz AJ, Llull R, Hedrick MH, et al. Emerging approaches to the tissue engineering of fat. Clin Plast Surg. 1999 Oct;26(4):587-603, viii.
    [25]An C, Cheng Y, Yuan Q, et al. IGF-1 and BMP-2 induces differentiation of adipose-derived mesenchymal stem cells into chondrocytes-like cells. Ann Biomed Eng. Apr;38(4):1647- 1654.
    [26]Madonna R, Willerson JT, Geng YJ. Myocardin a enhances telomerase activities in adipose tissue mesenchymal cells and embryonic stem cells undergoing cardiovascular myogenic differentiation. Stem Cells. 2008 Jan;26(1):202-211.
    [27]Farre-Guasch E, Marti-Pages C, Hernandez-Alfaro F, et al. Buccal Fat Pad, an Oral Access Source of Human Adipose Stem Cells with Potential for Osteochondral Tissue Engineering: An In Vitro Study. Tissue Eng Part C Methods. Mar 2.
    [28]Li J, Gao JH, Lu F, et al. [Experimental study of the effect of adipose tissue derived stem cells on the survival rate of free fat transplantation]. Zhonghua Zheng Xing Wai Ke Za Zhi. 2009 Mar;25(2):129-133.
    [29]De Ugarte DA, Morizono K, Elbarbary A, et al. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs. 2003;174(3):101-109.
    [30]Rodriguez AM, Elabd C, Amri EZ, et al.The human adipose tissue is a source of multipotent stem cells. Biochimie. 2005 Jan;87(1):125-128.
    [31]Estes BT, Wu AW, Guilak F. Potent induction of chondrocytic differentiation of human adipose-derived adult stem cells by bone morphogenetic protein 6. Arthritis Rheum. 2006 Apr;54(4):1222-1232.
    [32]Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002 Dec;13(12):4279-4295.
    [33]Peptan IA, Hong L, Mao JJ. Comparison of osteogenic potentials of visceral and subcutaneous adipose-derived cells of rabbits. Plast Reconstr Surg. 2006 Apr 15;117(5): 1462-1470.
    [34]Gimble JM, Guilak F. Differentiation potential of adipose derived adult stem (ADAS) cells. Curr Top Dev Biol. 2003;58:137-160.
    [35]Schaffler A, Buchler C. Concise review: adipose tissue-derived stromal cells--basic and clinical implications for novel cell-based therapies. Stem Cells. 2007 Apr;25(4):818-827.
    [36]Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007 May 11;100(9):1249-1260.
    [37]Shi C, Zhu Y, Su Y, et al. Stem cells and their applications in skin-cell therapy. Trends Biotechnol. 2006 Jan;24(1):48-52.
    [38]Amoh Y, Li L, Katsuoka K, et al. Multipotent nestin-positive, keratin-negative hair-follicle bulge stem cells can form neurons. Proc Natl Acad Sci U S A. 2005 Apr 12; 102 (15): 5530-5534.
    [39]Taylor G, Lehrer MS, Jensen PJ, et al. Involvement of follicular stem cells in forming not only the follicle but also the epidermis. Cell. 2000 Aug 18;102(4):451-461.
    [40]Mao JJ, Giannobile WV, Helms JA, et al. Craniofacial tissue engineering by stem cells. J Dent Res. 2006 Nov;85(11):966-979.
    [41]Seo BM, Miura M, Gronthos S, et al. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet. 2004 Jul 10-16;364(9429):149-155.
    [42]Thomson JA, Marshall VS. Primate embryonic stem cells. Curr Top Dev Biol. 1998; 38:133-165.
    [43]Morrison SJ, White PM, Zock C, et al. Prospective identification, isolation by flow cytometry, and in vivo self-renewal of multipotent mammalian neural crest stem cells. Cell. 1999 Mar 5;96(5):737-749.
    [44]Dieterlen-Lievre F. [Lineage-switching by pluripotent cells derived from adults]. J Soc Biol. 2001;195(1):39-46.
    [45]Bailey AS, Jiang S, Afentoulis M, et al. Transplanted adult hematopoietic stems cells differentiate into functional endothelial cells. Blood. 2004 Jan 1;103(1):13-19.
    [46]Woodbury D, Schwarz EJ, Prockop DJ, et al. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res. 2000 Aug 15;61(4):364-370.
    [47]Bjornson CR, Rietze RL, Reynolds BA, et al. Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science. 1999 Jan 22;283 (5401): 534-537.
    [48]Dezawa M, Kanno H, Hoshino M, et al. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest. 2004 Jun;113(12):1701-1710.
    [49]Yanai S, Adachi Y, Shi M, et al. Adult bone marrow cells can differentiate into hemopoietic cells and endothelial cells but not into other lineage cells in normal growth and normal life. Int J Hematol. Mar;91(2):213-218.
    [50]Hori Y, Nakamura T, Kimura D, et al. Experimental study on tissue engineering of the small intestine by mesenchymal stem cell seeding. J Surg Res. 2002 Feb;102(2):156-160.
    [51]Lee RH, Kim B, Choi I, et al. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem. 2004; 14(4-6):311-324.
    [52]Muschler GF, Nitto H, Boehm CA, et al. Age- and gender-related changes in the cellularity of human bone marrow and the prevalence of osteoblastic progenitors. J Orthop Res. 2001 Jan;19(1):117-125.
    [53]Aust L, Devlin B, Foster SJ, et al. Yield of human adipose-derived adult stem cells from liposuction aspirates. Cytotherapy. 2004;6(1):7-14.
    [54]Bacigalupo A, Tong J, Podesta M, et al. Bone marrow harvest for marrow transplantation: effect of multiple small (2 ml) or large (20 ml) aspirates. Bone Marrow Transplant. 1992 Jun;9(6):467-470.
    [55]Nishimori M, Yamada Y, Hoshi K, et al. Health-related quality of life of unrelated bone marrow donors in Japan. Blood. 2002 Mar 15;99(6):1995-2001.
    [56]Boquest AC, Shahdadfar A, Brinchmann JE, et al. Isolation of stromal stem cells fromhuman adipose tissue. Methods Mol Biol. 2006;325:35-46.
    [57]Follmar KE, Decroos FC, Prichard HL, et al. Effects of glutamine, glucose, and oxygen concentration on the metabolism and proliferation of rabbit adipose-derived stem cells. Tissue Eng. 2006 Dec;12(12):3525-3533.
    [58]Guilak F, Lott KE, Awad HA, et al. Clonal analysis of the differentiation potential of human adipose-derived adult stem cells. J Cell Physiol. 2006 Jan;206(1):229-237.
    [59]Gronthos S, Franklin DM, Leddy HA, et al. Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol. 2001 Oct;189(1):54-63.
    [60]Strem BM, Hicok KC, Zhu M, et al. Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med. 2005 Sep;54(3):132-141.
    [61]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 Oct 31;89(2-3):267-270.
    [62]Silva WA, Jr., Covas DT, et al. The profile of gene expression of human marrow mesenchymal stem cells. Stem Cells. 2003;21(6):661-669.
    [63]Mitchell JB, McIntosh K, Zvonic S, et al. Immunophenotype of human adipose- derived cells: temporal changes in stromal-associated and stem cell-associated markers. Stem Cells. 2006 Feb;24(2):376-385.
    [64]Kern S, Eichler H, Stoeve J, et al. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006 May;24(5): 1294- 1301.
    [65]Nakagami H, Morishita R, Maeda K, et al. Adipose tissue-derived stromal cells as a novel option for regenerative cell therapy. J Atheroscler Thromb. 2006 Apr;13(2):77-81.
    [66]Lin Y, Wang T, Wu L, et al. Ectopic and in situ bone formation of adipose tissue- derived stromal cells in biphasic calcium phosphate nanocomposite. J Biomed Mater Res A. 2007 Jun 15;81(4):900-910.
    [67]Cowan CM, Shi YY, Aalami OO, et al. Adipose-derived adult stromal cells heal critical- size mouse calvarial defects. Nat Biotechnol. 2004 May;22(5):560-567.
    [68]Huang JI, Beanes SR, Zhu M, et al. Rat extramedullary adipose tissue as a source of osteochondrogenic progenitor cells. Plast Reconstr Surg. 2002 Mar;109(3):1033-1041; discussion 1042-1033.
    [69]Li X, Yao J, Wu L, et al. Osteogenic Induction of Adipose-derived Stromal Cells: Not a Requirement for Bone Formation In Vivo. Artif Organs. 2009 Oct 11.
    [70]Lendeckel S, Jodicke A, Christophis P, et al. Autologous stem cells (adipose) andfibrin glue used to treat widespread traumatic calvarial defects: case report. J Craniomaxillofac Surg. 2004 Dec;32(6):370-373.
    [71]Katz AJ, Tholpady A, Tholpady SS, et al. Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hADAS) cells. Stem Cells. 2005 Mar;23(3): 412-423.
    [72]Winter A, Breit S, Parsch D, et al. Cartilage-like gene expression in differentiated human stem cell spheroids: a comparison of bone marrow-derived and adipose tissue-derived stromal cells. Arthritis Rheum. 2003 Feb;48(2):418-429.
    [73]Dragoo JL, Samimi B, Zhu M, et al. Tissue-engineered cartilage and bone using stem cells from human infrapatellar fat pads. J Bone Joint Surg Br. 2003 Jul;85(5):740-747.
    [74]Shieh SJ, Vacanti JP. State-of-the-art tissue engineering: from tissue engineering to organ building. Surgery. 2005 Jan;137(1):1-7.
    [75]Bacou F, el Andalousi RB, Daussin PA, et al. Transplantation of adipose tissue- derived stromal cells increases mass and functional capacity of damaged skeletal muscle. Cell Transplant. 2004;13(2):103-111.
    [76]Hong L, Peptan IA, Colpan A, et al. Adipose tissue engineering by human adipose-derived stromal cells. Cells Tissues Organs. 2006;183(3):133-140.
    [77]Rangappa S, Entwistle JW, Wechsler AS, et al. Cardiomyocyte-mediated contact programs human mesenchymal stem cells to express cardiogenic phenotype. J Thorac Cardiovasc Surg. 2003 Jul;126(1):124-132.
    [78]Zhu Y, Liu T, Song K, et al. Adipose-derived stem cell: a better stem cell than BMSC. Cell Biochem Funct. 2008 Aug;26(6):664-675.
    [79]Zhu Y, Liu T, Song K, et al. ADSCs differentiated into cardiomyocytes in cardiac microenvironment. Mol Cell Biochem. 2009 Apr;324(1-2):117-129.
    [80]Strem BM, Zhu M, Alfonso Z, et al. Expression of cardiomyocytic markers on adipose tissue-derived cells in a murine model of acute myocardial injury. Cytotherapy. 2005;7(3): 282-291.
    [81]Cao Y, Sun Z, Liao L, et al. Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo. Biochem Biophys Res Commun. 2005 Jul 1;332(2):370-379.
    [82]Miranville A, Heeschen C, Sengenes C, et al. Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation. 2004 Jul 20;110(3):349-355.
    [83]Rehman J, Traktuev D, Li J, et al. Secretion of angiogenic and antiapoptotic factorsby human adipose stromal cells. Circulation. 2004 Mar 16;109(10):1292-1298.
    [84]Casteilla L, Planat-Benard V, Cousin B, et al. Plasticity of adipose tissue: a promising therapeutic avenue in the treatment of cardiovascular and blood diseases? Arch Mal Coeur Vaiss. 2005 Sep;98(9):922-926.
    [85]Seo MJ, Suh SY, Bae YC, et al. Differentiation of human adipose stromal cells into hepatic lineage in vitro and in vivo. Biochem Biophys Res Commun. 2005 Mar 4;328(1):258-264.
    [86]Kim S, Moustaid-Moussa N. Secretory, endocrine and autocrine/paracrine function of the adipocyte. J Nutr. 2000 Dec;130(12):3110S-3115S.
    [87]Neuhuber B, Gallo G, Howard L, et al. Reevaluation of in vitro differentiation protocols for bone marrow stromal cells: disruption of actin cytoskeleton induces rapid morphological changes and mimics neuronal phenotype. J Neurosci Res. 2004 Jul 15;77(2): 192-204.
    [88]Lu H, Nishi M, Matsuda K, et al. Estrogen reduces the neurite growth of serotonergic cells expressing estrogen receptors. Neurosci Res. 2004 Sep;50(1):23-28.
    [89]Ashjian PH, Elbarbary AS, Edmonds B, et al. In vitro differentiation of human processed lipoaspirate cells into early neural progenitors. Plast Reconstr Surg. 2003 May;111(6):1922-1931.
    [90]Aluigi MG, Coradeghini R, Guida C, et al. Pre-adipocytes commitment to neurogenesis 1: preliminary localisation of cholinergic molecules. Cell Biol Int. 2009 May;33(5): 594-601.
    [91]Kang SK, Putnam LA, Ylostalo J, et al. Neurogenesis of Rhesus adipose stromal cells. J Cell Sci. 2004 Aug 15;117(Pt 18):4289-4299.
    [92]Romanov YA, Darevskaya AN, Merzlikina NV, et al. Mesenchymal stem cells from human bone marrow and adipose tissue: isolation, characterization, and differentiation potentia- lities. Bull Exp Biol Med. 2005 Jul;140(1):138-143.
    [93]Timper K, Seboek D, Eberhardt M, et al. Human adipose tissue-derived mesenchymal stem cells differentiate into insulin, somatostatin, and glucagon expressing cells. Biochem Biophys Res Commun. 2006 Mar 24;341(4):1135-1140.
    [94]陈小红,刘鲁川,金岩,等.脂肪来源干细胞诱导分化为成牙本质样细胞的体外培养研究.实用口腔医学杂志. 2008;24(1):9-12.
    [95]Kim WS, Park BS, Sung JH, et al. Wound healing effect of adipose-derived stem cells: a critical role of secretory factors on human dermal fibroblasts. J Dermatol Sci. 2007Oct;48(1):15-24.
    [96]Kilroy GE, Foster SJ, Wu X, et al. Cytokine profile of human adipose-derived stem cells: expression of angiogenic, hematopoietic, and pro-inflammatory factors. J Cell Physiol. 2007 Sep;212(3):702-709.
    [97]Sadat S, Gehmert S, Song YH, et al. The cardioprotective effect of mesenchymal stem cells is mediated by IGF-I and VEGF. Biochem Biophys Res Commun. 2007 Nov 23;363(3): 674-679.
    [98] Kim Y, Kim H, Cho H, et al. Direct comparison of human mesenchymal stem cells derived from adipose tissues and bone marrow in mediating neovascularization in response to vascular ischemia. Cell Physiol Biochem. 2007;20(6):867-876.
    [99] Mizuno H, Itoi Y, Kawahara S, et al. In vivo adipose tissue regeneration by adipose- derived stromal cells isolated from GFP transgenic mice. Cells Tissues Organs. 2008; 187(3):177-185.
    [100]Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993 Dec 3;75(5):843-854.
    [101]Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993 Dec 3;75(5):855-862.
    [102]Reinhart BJ, Slack FJ, Basson M, et al. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 2000 Feb 24;403(6772): 901- 906.
    [103]Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004 Jan 23;116(2):281-297.
    [104]Tsuchiya S, Okuno Y, Tsujimoto G. MicroRNA: biogenetic and functional mecha- nisms and involvements in cell differentiation and cancer. J Pharmacol Sci. 2006 Aug; 101(4): 267-270.
    [105]Xu P, Vernooy SY, Guo M, et al. The Drosophila microRNA Mir-14 suppresses cell death and is required for normal fat metabolism. Curr Biol. 2003 Apr 29;13(9):790-795.
    [106]Brennecke J, Hipfner DR, Stark A, et al. bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell. 2003 Apr 4;113(1):25-36.
    [107]Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006 Apr;6(4):259-269.
    [108]Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005 Jan14;120(1):15-20.
    [109]Xie X, Lu J, Kulbokas EJ, et al. Systematic discovery of regulatory motifs in human promoters and 3' UTRs by comparison of several mammals. Nature. 2005 Mar 17;434(7031):338-345.
    [110]Couzin J. Breakthrough of the year. Small RNAs make big splash. Science. 2002 Dec 20;298(5602):2296-2297.
    [111]Lim LP, Glasner ME, Yekta S, et al. Vertebrate microRNA genes. Science. 2003 Mar 7;299(5612):1540.
    [112]Ruvkun G. Molecular biology. Glimpses of a tiny RNA world. Science. 2001 Oct 26;294(5543):797-799.
    [113]Lau NC, Lim LP, Weinstein EG, et al. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001 Oct 26;294(5543):858-862.
    [114]Reinhart BJ, Weinstein EG, Rhoades MW, et al. MicroRNAs in plants. Genes Dev. 2002 Jul 1;16(13):1616-1626.
    [115]Lagos-Quintana M, Rauhut R, Lendeckel W, et al. Identification of novel genes coding for small expressed RNAs. Science. 2001 Oct 26;294(5543):853-858.
    [116]Hutvagner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme complex. Science. 2002 Sep 20;297(5589):2056-2060.
    [117]Pasquinelli AE, Hunter S, Bracht J. MicroRNAs: a developing story. Curr Opin Genet Dev. 2005 Apr;15(2):200-205.
    [118]Yi R, Qin Y, Macara IG, et al. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003 Dec 15;17(24):3011-3016.
    [119]Lund E, Guttinger S, Calado A, et al. Nuclear export of microRNA precursors. Science. 2004 Jan 2;303(5654):95-98.
    [120]Dennis C. The brave new world of RNA. Nature. 2002 Jul 11;418(6894):122-124.
    [121]Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit strand bias. Cell. 2003 Oct 17;115(2):209-216.
    [122]Oakley EJ, Van Zant G. Unraveling the complex regulation of stem cells: implications for aging and cancer. Leukemia. 2007 Apr;21(4):612-621.
    [123]Lee YS, Nakahara K, Pham JW, et al. Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways. Cell. 2004 Apr 2;117(1):69-81.
    [124]Bernstein E, Kim SY, Carmell MA, et al. Dicer is essential for mouse development. Nat Genet. 2003 Nov;35(3):215-217.
    [125]Kanellopoulou C, Muljo SA, Kung AL, et al. Dicer-deficient mouse embryonic stemcells are defective in differentiation and centromeric silencing. Genes Dev. 2005 Feb 15;19(4):489-501.
    [126]Murchison EP, Partridge JF, Tam OH, et al. Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci U S A. 2005 Aug 23;102(34):12135-12140.
    [127]Cobb BS, Nesterova TB, Thompson E, et al. T cell lineage choice and differentiation in the absence of the RNase III enzyme Dicer. J Exp Med. 2005 May 2;201(9):1367-1373.
    [128]Chen C, Ridzon D, Lee CT, et al. Defining embryonic stem cell identity using differentiation-related microRNAs and their potential targets. Mamm Genome. 2007 May;18(5):316-327.
    [129]Morin RD, O'Connor MD, Griffith M, et al. Application of massively parallel sequencing to microRNA profiling and discovery in human embryonic stem cells. Genome Res. 2008 Apr;18(4):610-621.
    [130]Callis TE, Chen JF, Wang DZ. MicroRNAs in skeletal and cardiac muscle development. DNA Cell Biol. 2007 Apr;26(4):219-225.
    [131]Naguibneva I, Ameyar-Zazoua M, Polesskaya A, et al. The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nat Cell Biol. 2006 Mar;8(3):278-284.
    [132]Rao PK, Kumar RM, Farkhondeh M, et al. Myogenic factors that regulate expression of muscle-specific microRNAs. Proc Natl Acad Sci U S A. 2006 Jun 6;103(23): 8721-8726.
    [133]Kim HK, Lee YS, Sivaprasad U, et al. Muscle-specific microRNA miR-206 promotes muscle differentiation. J Cell Biol. 2006 Aug 28;174(5):677-687.
    [134]Schoolmeesters A, Eklund T, Leake D, et al. Functional profiling reveals critical role for miRNA in differentiation of human mesenchymal stem cells. PLoS One. 2009;4(5):e5605.
    [135]Iorio MV, Ferracin M, Liu CG, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005 Aug 15;65(16):7065-7070.
    [136]Visone R, Pallante P, Vecchione A, et al. Specific microRNAs are downregulated in human thyroid anaplastic carcinomas. Oncogene. 2007 Nov 29;26(54):7590-7595.
    [137]Lee YS, Kim HK, Chung S, et al. Depletion of human micro-RNA miR-125b reveals that it is critical for the proliferation of differentiated cells but not for the down-regulation of putative targets during differentiation. J Biol Chem. 2005 Apr 29;280(17):16635-16641.
    [138]Scott GK, Goga A, Bhaumik D, et al. Coordinate suppression of ERBB2 and ERBB3 by enforced expression of micro-RNA miR-125a or miR-125b. J Biol Chem. 2007 Jan 12;282(2):1479-1486.
    [139]Sempere LF, Freemantle S, Pitha-Rowe I, et al. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol. 2004;5(3):R13.
    [140]Mizuno Y, Yagi K, Tokuzawa Y, et al. miR-125b inhibits osteoblastic differentiation by down-regulation of cell proliferation. Biochem Biophys Res Commun. 2008 Apr 4;368(2):267-272.
    [141]Goff LA, Boucher S, Ricupero CL, et al. Differentiating human multipotent mesenchymal stromal cells regulate microRNAs: prediction of microRNA regulation by PDGF during osteogenesis. Exp Hematol. 2008 Oct;36(10):1354-1369.
    [142]Oskowitz AZ, Lu J, Penfornis P, et al. Human multipotent stromal cells from bone marrow and microRNA: regulation of differentiation and leukemia inhibitory factor expression. Proc Natl Acad Sci U S A. 2008 Nov 25;105(47):18372-18377.
    [143]Hicok KC, Du Laney TV, Zhou YS, et al. Human adipose-derived adult stem cells produce osteoid in vivo. Tissue Eng. 2004 Mar-Apr;10(3-4):371-380.
    [144]Luzi E, Marini F, Sala SC, et al. Osteogenic differentiation of human adipose tissue-derived stem cells is modulated by the miR-26a targeting of the SMAD1 transcription factor. J Bone Miner Res. 2008 Feb;23(2):287-295.
    [145]Kim J, Johnson K, Chen HJ, et al. Drosophila Mad binds to DNA and directly mediates activation of vestigial by Decapentaplegic. Nature. 1997 Jul 17;388(6639):304-308.
    [146]Shi X, Yang X, Chen D, et al. Smad1 interacts with homeobox DNA-binding proteins in bone morphogenetic protein signaling. J Biol Chem. 1999 May 7;274(19):13711-13717.
    [147]Yang X, Ji X, Shi X, et al. Smad1 domains interacting with Hoxc-8 induce osteoblast differentiation. J Biol Chem. 2000 Jan 14;275(2):1065-1072.
    [148]Kim YJ, Bae SW, Yu SS, et al. miR-196a regulates proliferation and osteogenic differentiation in mesenchymal stem cells derived from human adipose tissue. J Bone Miner Res. 2009 May;24(5):816-825.
    [149]Li Z, Hassan MQ, Volinia S, et al. A microRNA signature for a BMP2-induced osteoblast lineage commitment program. Proc Natl Acad Sci U S A. 2008 Sep 16;105(37):13906-13911.
    [150]Li Z, Hassan MQ, Jafferji M, et al. Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J Biol Chem. 2009 Jun 5;284(23): 15676- 15684.
    [151]Palmieri A, Pezzetti F, Brunelli G, et al. Peptide-15 changes miRNA expression inosteoblast-like cells. Implant Dent. 2008 Mar;17(1):100-108.
    [152]Annalisa P, Furio P, Ilaria Z, et al. Anorganic bovine bone and a silicate-based synthetic bone activate different microRNAs. J Oral Sci. 2008 Sep;50(3):301-307.
    [153]Tokunaga T, Yamamoto H, Shimada S, et al. Antitumor activity of deoxyribonucleic acid fraction from Mycobacterium bovis BCG. I. Isolation, physicochemical characterization, and antitumor activity. J Natl Cancer Inst. 1984 Apr;72(4):955-962.
    [154]Krieg AM, Yi AK, Matson S, et al. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. 1995 Apr 6;374(6522):546-549.
    [155]Hemmi H, Takeuchi O, Kawai T, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000 Dec 7;408(6813):740-745.
    [156]Hornung V, Rothenfusser S, Britsch S, et al. Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J Immunol. 2002 May 1;168(9):4531-4537.
    [157]Yi AK, Krieg AM. Rapid induction of mitogen-activated protein kinases by immune stimulatory CpG DNA. J Immunol. 1998 Nov 1;161(9):4493-4497.
    [158]Choudhury BK, Wild JS, Alam R, et al. In vivo role of p38 mitogen-activated protein kinase in mediating the anti-inflammatory effects of CpG oligodeoxynucleotide in murine asthma. J Immunol. 2002 Nov 15;169(10):5955-5961.
    [159]Yi AK, Yoon JG, Krieg AM. Convergence of CpG DNA- and BCR-mediated signals at the c-Jun N-terminal kinase and NF-kappaB activation pathways: regulation by mitogen-activated protein kinases. Int Immunol. 2003 May;15(5):577-591.
    [160]Yi AK, Yoon JG, Yeo SJ, et al. Role of mitogen-activated protein kinases in CpG DNA-mediated IL-10 and IL-12 production: central role of extracellular signal-regulated kinase in the negative feedback loop of the CpG DNA-mediated Th1 response. J Immunol. 2002 May 1;168(9):4711-4720.
    [161]Rutz M, Metzger J, Gellert T, et al. Toll-like receptor 9 binds single-stranded CpG-DNA in a sequence- and pH-dependent manner. Eur J Immunol. 2004 Sep;34(9): 2541- 2550.
    [162]Gursel I, Gursel M, Yamada H, et al. Repetitive elements in mammalian telomeres suppress bacterial DNA-induced immune activation. J Immunol. 2003 Aug 1;171(3):1393- 1400.
    [163]Jurk M, Vollmer J. Therapeutic applications of synthetic CpG oligodeoxynucleotides as TLR9 agonists for immune modulation. BioDrugs. 2007;21(6):387-401.
    [164]Hartmann G, Krieg AM. Mechanism and function of a newly identified CpG DNAmotif in human primary B cells. J Immunol. 2000 Jan 15;164(2):944-953.
    [165]Halperin SA, Van Nest G, Smith B, et al. A phase I study of the safety and immunogenicity of recombinant hepatitis B surface antigen co-administered with an immunostimulatory phosphorothioate oligonucleotide adjuvant. Vaccine. 2003 Jun 2;21(19-20):2461-2467.
    [166]Mason KA, Ariga H, Neal R, et al. Targeting toll-like receptor 9 with CpG oligodeoxynucleotides enhances tumor response to fractionated radiotherapy. Clin Cancer Res. 2005 Jan 1;11(1):361-369.
    [167]Jahrsdorfer B, Wooldridge JE, Blackwell SE, et al. Immunostimulatory oligodeoxy nucleotides induce apoptosis of B cell chronic lymphocytic leukemia cells. J Leukoc Biol. 2005 Mar;77(3):378-387.
    [168]Robinson DS, Hamid Q, Ying S, et al. Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma. N Engl J Med. 1992 Jan 30;326(5):298-304.
    [169]Nakajima H, Iwamoto I, Yoshida S. Aerosolized recombinant interferon-gamma prevents antigen-induced eosinophil recruitment in mouse trachea. Am Rev Respir Dis. 1993 Oct;148(4 Pt 1):1102-1104.
    [170]Sur S, Lam J, Bouchard P, et al. Immunomodulatory effects of IL-12 on allergic lung inflammation depend on timing of doses. J Immunol. 1996 Nov 1;157(9):4173-4180.
    [171]Hussain I, Kline JN. CpG oligodeoxynucleotides in asthma. Curr Opin Investig Drugs. 2001 Jul;2(7):914-918.
    [172]Halpern MD, Pisetsky DS. In vitro inhibition of murine IFN gamma production by phosphorothioate deoxyguanosine oligomers. Immunopharmacology. 1995 Feb;29(1):47-52.
    [173]Pisetsky DS, Reich CF. Inhibition of murine macrophage IL-12 production by natural and synthetic DNA. Clin Immunol. 2000 Sep;96(3):198-204.
    [174]Krieg AM, Love-Homan L, Yi AK, et al. CpG DNA induces sustained IL-12 expression in vivo and resistance to Listeria monocytogenes challenge. J Immunol. 1998 Sep 1;161(5):2428-2434.
    [175]Hartmann G, Weeratna RD, Ballas ZK, et al. Delineation of a CpG phosphorothioate oligodeoxynucleotide for activating primate immune responses in vitro and in vivo. J Immunol. 2000 Feb 1;164(3):1617-1624.
    [176]Kamstrup S, Verthelyi D, Klinman DM. Response of porcine peripheral blood mononuclear cells to CpG-containing oligodeoxynucleotides. Vet Microbiol. 2001 Feb 26;78(4):353-362.
    [177]Rankin R, Pontarollo R, Gomis S, et al. CpG-containing oligodeoxynucleotidesaugment and switch the immune responses of cattle to bovine herpesvirus-1 glycoprotein D. Vaccine. 2002 Jul 26;20(23-24):3014-3022.
    [178]Jones TR, Obaldia N, 3rd, et al. Synthetic oligodeoxynucleotides containing CpG motifs enhance immunogenicity of a peptide malaria vaccine in Aotus monkeys. Vaccine. 1999 Aug 6;17(23-24):3065-3071.
    [179]Nichani AK, Kaushik RS, Mena A, et al. CpG oligodeoxynucleotide induction of antiviral effector molecules in sheep. Cell Immunol. 2004 Jan;227(1):24-37.
    [180]Wernette CM, Smith BF, Barksdale ZL, et al. CpG oligodeoxynucleotides stimulate canine and feline immune cell proliferation. Vet Immunol Immunopathol. 2002 Jan 15;84(3-4): 223-236.
    [181]Verthelyi D, Ishii KJ, Gursel M, et al. Human peripheral blood cells differentially recognize and respond to two distinct CPG motifs. J Immunol. 2001 Feb 15;166(4):2372-2377.
    [182]Leifer CA, Verthelyi D, Klinman DM. Heterogeneity in the human response to immunostimulatory CpG oligodeoxynucleotides. J Immunother. 2003 Jul-Aug;26(4):313-319.
    [183]Agrawal S, Temsamani J, Galbraith W, et al. Pharmacokinetics of antisense oligonucleotides. Clin Pharmacokinet. 1995 Jan;28(1):7-16.
    [184]Gursel I, Gursel M, Ishii KJ, et al. Sterically stabilized cationic liposomes improve the uptake and immunostimulatory activity of CpG oligonucleotides. J Immunol. 2001 Sep 15;167(6):3324-3328.
    [185]Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol. 2002;20:709-760.
    [186]Verthelyi D, Klinman DM. Immunoregulatory activity of CpG oligonucleotides in humans and nonhuman primates. Clin Immunol. 2003 Oct;109(1):64-71.
    [187]Heikenwalder M, Polymenidou M, Junt T, et al. et al. Lymphoid follicle destruction and immunosuppression after repeated CpG oligodeoxynucleotide administration. Nat Med. 2004 Feb;10(2):187-192.
    [188]Zou W, Schwartz H, Endres S, et al. CpG oligonucleotides: novel regulators of osteoclast differentiation. FASEB J. 2002 Mar;16(3):274-282.
    [189]Zou W, Amcheslavsky A, Bar-Shavit Z. CpG oligodeoxynucleotides modulate the osteoclastogenic activity of osteoblasts via Toll-like receptor 9. J Biol Chem. 2003 May 9;278(19):16732-16740.
    [190]Amcheslavsky A, Hemmi H, Akira S, et al. Differential contribution of osteoclast- and osteoblast-lineage cells to CpG-oligodeoxynucleotide (CpG-ODN) modulation of osteoclastogenesis. J Bone Miner Res. 2005 Sep;20(9):1692-1699.
    [191]Amcheslavsky A, Zou W, Bar-Shavit Z. Toll-like receptor 9 regulates tumor necrosis factor-alpha expression by different mechanisms. Implications for osteoclastogenesis. J Biol Chem. 2004 Dec 24;279(52):54039-54045.
    [192]Amcheslavsky A, Bar-Shavit Z. Interleukin (IL)-12 mediates the anti- osteocla- stogenic activity of CpG-oligodeoxynucleotides. J Cell Physiol. 2006 Apr;207(1): 244-250.
    [193]Amcheslavsky A, Bar-Shavit Z. Toll-like receptor 9 ligand blocks osteoclast differentiation through induction of phosphatase. J Bone Miner Res. 2007 Aug;22(8): 1301- 1310.
    [194]Chang JH, Chang EJ, Kim HH, et al. Enhanced inhibitory effects of a novel CpG motif on osteoclast differentiation via TREM-2 down-regulation. Biochem Biophys Res Commun. 2009 Nov 6;389(1):28-33.
    [195]Hwa Cho H, Bae YC, Jung JS. Role of toll-like receptors on human adipose-derived stromal cells. Stem Cells. 2006 Dec;24(12):2744-2752.
    [196]申玉芹,孙新华,于丽,等.不同CpG ODN对大鼠骨髓间充质干细胞增殖作用的研究.口腔医学研究. 2009;25(6):723-725.
    [197]申玉芹,孙新华,于丽,等.小寡核苷酸对大鼠骨髓间充质干细胞向成骨细胞分化的影响.吉林大学学报(医学版). 2010;36(2):336-339.
    [198]Liu Q, Cen L, Zhou H, et al. The role of the extracellular signal-related kinase signaling pathway in osteogenic differentiation of human adipose-derived stem cells and in adipogenic transition initiated by dexamethasone. Tissue Eng Part A. 2009 Nov;15(11): 3487-3497.
    [199]Tzaphlidou M. The role of collagen in bone structure: an image processing approach. Micron. 2005;36(7-8):593-601.
    [200]张赛霞,晖黎,陈东风,等. Van Gieson胶原纤维染色方法的探索.实用医技杂志. 2007;14(12):1580-1581.
    [201]Yalvac ME, Rizvanov AA, Kilic E, et al. Potential role of dental stem cells in the cellular therapy of cerebral ischemia. Curr Pharm Des. 2009;15(33):3908-3916.
    [202]Tobita M, Uysal AC, Ogawa R, et al. Periodontal tissue regeneration with adipose-derived stem cells. Tissue Eng Part A. 2008 Jun;14(6):945-953.
    [203]Quarles LD, Yohay DA, Lever LW, et al. Distinct proliferative and differentiated stages of murine MC3T3-E1 cells in culture: an in vitro model of osteoblast development. J Bone Miner Res. 1992 Jun;7(6):683-692.
    [204]van Rooij E, Liu N, Olson EN. MicroRNAs flex their muscles. Trends Genet. 2008Apr;24(4):159-166.
    [205]Makita N, Suzuki M, Asami S, et al. Two of four alternatively spliced isoforms of RUNX2 control osteocalcin gene expression in human osteoblast cells. Gene. 2008 Apr 30;413(1-2):8-17.
    [206]Ducy P, Starbuck M, Priemel M, et al. A Cbfa1-dependent genetic pathway controls bone formation beyond embryonic development. Genes Dev. 1999 Apr 15;13(8):1025-1036.
    [207]Stein GS, Lian JB, van Wijnen AJ, et al. Runx2 control of organization, assembly and activity of the regulatory machinery for skeletal gene expression. Oncogene. 2004 May 24;23(24):4315-4329.
    [208]Celil AB, Hollinger JO, Campbell PG. Osx transcriptional regulation is mediated by additional pathways to BMP2/Smad signaling. J Cell Biochem. 2005 Jun 1;95(3):518-528.
    [209]Zhang X, Aubin JE, Inman RD. Molecular and cellular biology of new bone formation: insights into the ankylosis of ankylosing spondylitis. Curr Opin Rheumatol. 2003 Jul;15(4):387-393.
    [210]Okazaki K, Sandell LJ. Extracellular matrix gene regulation. Clin Orthop Relat Res. 2004 Oct(427 Suppl):S123-128.
    [211]Li X, Nie S, Chang C, et al. Smads oppose Hox transcriptional activities. Exp Cell Res. 2006 Apr 1;312(6):854-864.
    [212]Kang M, Bok J, Deocaris CC, et al. Hoxc8 represses BMP-induced expression of Smad6. Mol Cells. 2009 Dec 7.
    [213]Yueh YG, Gardner DP, Kappen C. Evidence for regulation of cartilage differentiation by the homeobox gene Hoxc-8. Proc Natl Acad Sci U S A. 1998 Aug 18;95(17):9956-9961.
    [214]Wan M, Shi X, Feng X, et al. Transcriptional mechanisms of bone morphogenetic protein-induced osteoprotegrin gene expression. J Biol Chem. 2001 Mar 30;276(13):10119- 10125.
    [215]Hullinger TG, Pan Q, Viswanathan HL, et al. TGFbeta and BMP-2 activation of the OPN promoter: roles of smad- and hox-binding elements. Exp Cell Res. 2001 Jan 1;262(1):69-74.
    [216]Liu Z, Shi W, Ji X, et al. Molecules mimicking Smad1 interacting with Hox stimulate bone formation. J Biol Chem. 2004 Mar 19;279(12):11313-11319.

© 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