新生大鼠耳蜗前体细胞分化的分子机制及微环境的初步研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
在成年哺乳动物耳蜗中,听觉毛细胞和神经元在损伤后不能自然再生,这是感音神经性聋的主要原因。近年来,成功从新生鼠耳蜗分离出耳蜗前体细胞,也有人称之为耳蜗干细胞,在体外培养的耳蜗前体细胞能分化为毛细胞和螺旋神经元样细胞。研究耳蜗前体细胞的分化机制及微环境将为诱导损伤后毛细胞和螺旋神经元再生提供依据。我们通过实验发现细胞周期调节分子cyclin A2和CRP2可能参与了耳蜗的发育以及前体细胞的增殖、分化。
     实验一新生大鼠耳蜗前体细胞的分离、培养
     目的对新生大鼠耳蜗前体细胞进行分离、培养。
     方法从P2大鼠耳蜗分离培养前体细胞,血清诱导分化后用免疫细胞化学染色鉴定多向分化潜能,通过扫描电镜观察分化细胞表面的超微结构,进一步了解分化细胞的特性。
     结果原代培养的细胞第3天即可见“细胞球”,随着培养天数的增加,可见细胞球体积增大。细胞球内绝大部分细胞呈nestin和BrdU阳性,表明其具有自我更新的能力。细胞球经诱导分化14 d后,对分化细胞行免疫荧光化学染色,发现部分细胞表达毛细胞标志物myosin VIIA,部分细胞表达星形胶质细胞标志物GFAP或者成熟神经元标志物NeuN,证明其具有多向分化潜能。扫描电镜下可见扁平的三角形或不规则形细胞,表面具有纤毛样结构,其纤毛顶端的结构与正常耳蜗毛细胞纤毛顶端相似,此外还可见双极神经元形态的细胞。
     结论本部分实验为研究耳蜗前体细胞的分化机制奠定基础。
     实验二cyclin A2在大鼠耳蜗组织发育和耳蜗前体细胞分化过程中的表达变化
     目的细胞周期分子cyclin A2具有调控G1/S转换和G2/M转换的双重作用。目前对于cyclin A2在内耳中的功能尚未见任何报道。我们拟通过检测cyclin A2在大鼠耳蜗组织发育及耳蜗前体细胞分化过程中的表达情况,探讨其是否参与了哺乳动物耳蜗的发育。
     方法一、cyclin A2在大鼠耳蜗组织发育过程中的表达情况:P2、P10和P42的SD大鼠取耳蜗组织,RT-PCR、Western blot和免疫组织化学染色的方法检测cyclin A2的表达。二、cyclin A2在大鼠耳蜗前体细胞分化过程中的表达情况:用RT-PCR、免疫细胞化学染色和Western blot的方法检测cyclin A2在未分化、分化6天及分化14天耳蜗前体细胞的表达情况。
     结果cyclin A2表达于P2大鼠耳蜗的螺旋缘,在P10后的耳蜗未见表达。在前体细胞的分化过程中cyclin A2的表达量逐渐下降。
     结论在大鼠耳蜗组织发育及耳蜗前体细胞分化的过程中cyclin A2表达量逐渐下降,这表明cyclin A2可能参与了大鼠出生后耳蜗的发育。
     实验三CRP2及CRIP2在大鼠耳蜗组织发育和耳蜗前体细胞分化过程中的表达变化
     目的我们实验室在以往研究中通过RT-PCR及Western blot首次证实LIM分子CRP2及CRIP2表达于大鼠的内耳。其中CRP2表达于胚胎期耳蜗,在成年耳蜗中无表达;而CRIP2在不同年龄耳蜗中(包括胚胎期)表达量无明显差异。为进一步探讨CRP2及CRIP2是否参与了哺乳动物耳蜗的发育,我们拟通过免疫组织化学和免疫细胞化学的方法检测CRP2及CRIP2在不同年龄大鼠耳蜗组织中的分布及耳蜗前体细胞分化前后的表达变化。
     方法一、CRP2及CRIP2在大鼠耳蜗组织发育过程中的表达情况:免疫组织化学染色的方法检测其在P2、P6和P10大鼠耳蜗中的表达。二、CRP2及CRIP2在大鼠耳蜗前体细胞分化过程中的表达情况:用免疫细胞化学染色的方法检测其在未分化和分化7天耳蜗前体细胞中的表达情况。
     结果免疫组化结果显示,CRP2主要表达于螺旋神经节,并且在耳蜗发育过程中,其阳性细胞数逐渐减少。CRIP2主要表达于耳蜗组织中的螺旋神经节、螺旋缘和Corti器,而且在P2、P6和P10各组中阳性细胞数量无明显差异。另外,在P2组前庭的感觉上皮部位也可见CRIP2和CRP2的表达。免疫细胞化学染色显示,CRIP2和CRP2均可表达于未分化的前体细胞,在胞核和胞质中均呈阳性表达,CRIP2可表达于分化7天细胞的胞质。分化细胞未见CRP2的表达。
     结论CRP2可能参与了耳蜗的发育以及毛细胞和螺旋神经元的增殖;CRIP2在耳蜗不同发育时期以及前体细胞分化前后均稳定表达,表明其可能在维持耳蜗细胞基本功能中发挥一定作用。
     实验四耳蜗前体细胞分化的微环境对神经干细胞分化影响的初步研究
     目的耳蜗前体细胞向毛细胞和螺旋神经元定向分化的具体机制尚不清楚,前体细胞所处的微环境在分化过程中起重要作用。为探讨微环境在细胞分化中的作用,我们在体外将GFP标记的神经干细胞和耳蜗前体细胞按比例混合,并诱导其分化,观察耳蜗前体细胞分化的微环境对神经干细胞分化的影响。
     方法将耳蜗前体细胞和GFP标记的神经干细胞按10:1的比例混合后接种于同一培养皿中,诱导分化后进行贴壁培养。21天后采用免疫荧光的方法对分化细胞进行鉴定。一抗采用抗myosin VIIA(毛细胞标志物)和抗peripherin(神经元标志物)。
     结果耳蜗前体细胞分化的微环境促进神经干细胞的存活,并诱导其向myosin VIIA阳性细胞及双极神经元形态细胞分化。
     结论耳蜗前体细胞的成功分离、培养为研究其分化的机制提供了一个极佳的体外模型,对于耳蜗前体细胞分化的微环境需要更加深入的研究。
     实验五干细胞向雏鸡和成年豚鼠耳蜗移植方法的建立
     目的研究耳蜗前体细胞分化的机制,目的是诱导外源性干细胞定向分化为听觉毛细胞及神经元,或刺激体内内源性耳蜗前体细胞再生,从而修复受损的听觉细胞。将外源性干细胞或治疗基因表达载体(如腺病毒等)植入耳蜗是治疗的前提,为此我们建立了干细胞向动物耳蜗移植的方法。
     方法雏鸡耳蜗的干细胞移植是经外耳道剥离鼓膜,暴露中耳腔的蜗窗,再经蜗窗膜造孔注入干细胞。成年豚鼠耳蜗鼓阶的干细胞移植是行耳后切口暴露听泡,在耳蜗底转近镫骨动脉处钻孔,并注入GFP标记的干细胞。中阶的移植是经耳蜗第三转的血管纹外侧壁注入干细胞。细胞移植1天后,分离雏鸡和豚鼠的耳蜗作冰冻切片,在荧光显微镜下观察移植细胞的分布。
     结果移植后的干细胞可分布于雏鸡耳蜗及豚鼠耳蜗的鼓阶、中阶。
     结论成功建立了干细胞向雏鸡和成年豚鼠耳蜗移植的方法,为进一步将干细胞应用于耳聋的治疗及机制研究奠定基础。
In adult mammalian cochlea, the hair cells cannot regenerate spontaneously after damaged. This is the major cause of permanent hearing loss. Cochlear stem cells have been isolated from the early postnatal rat and mouse in previous studies. These stem cells cultured in vitro can differentiate into neurons, astrocytes, hair cells and supporting cells. Differentiation of cochlear stem cells is critical to the development of cochlea tissue. Study of cochlear stem cells may help to learn more about cochlear development.
     Microenvironment is critical to differentiation of stem cells. But little is known about microenvironment of cochlear stem cell differentiation. Microenvironment of differentiation consists of all molecules in molecular network. We need to know more molecules involved in this network. Cell cycle exit participates in stem cell differentiation. Several cell cycle regulators, including cyclin A2 and CRP2, may be involved in this network.
     Hence, we isolated cochlear stem cells from newborn rats and induced differentiation of these cells. The level of cyclin A2 and CRP2 were assessed in stem cells and differentiated cells for better understanding of cochlea development mechanism. Then we cultured neural stem cells in the microenvironment of cochlear stem cell differentiation to see the effect of microenvironment on neural stem cell differentiation.
     1. Isolation and culture of cochlear stem cells from newborn rat
     Stem cells were isolated from the P2 rat cochlea tissues. Stem cells were incubated at 37°C in 5% CO2 atmosphere. The culture medium consisting of DMEM/F12 supplemented with B27, N2, EGF and FGF-2. For cell expansion and testing for self-renewal, we plated the cells at very low density to ensure that cell spheres were formed from single cells. For analysis of cell differentiation, we transferred cell spheres into 6-well dishes with poly-l-lysine treated coverslips and filled with 10% FCS. After 16 h, we replaced the medium with serum-free DMEM/F12 supplemented with N2 and B27. Half of the medium was replaced every second day. The differentiated cells were identified after 14 days in culture by immunocytochemistry. The cell spheres and differentiated cells were fixed with 4% paraformaldehyde, and incubated at 4°C overnight with primary antibodies: mouse anti-nestin monoclonal antibody, mouse anti-BrdU monoclonal antibody, rabbit anti-myosin VIIA (hair cell marker) polyclonal antibody, rabbit anti-GFAP (astrocyte marker) polyclonal antibody, mouse anti-NeuN (neuron marker) monoclonal antibody. Cy3 conjugated second antibodies were used to detect primary antibodies. Hoechst 33342 was used to visualize nuclei. For BrdU detection, stem cells were incubated with 10μM BrdU in the culture medium for 24 h, fixed for 30 min, treated with 2 N HCl for 30 min before antibody incubation. The differentiated cells cultured for 21 d were analyzed by SEM
     We found sphere-forming capacity of isolated cells after 7 days in culture. These cells were further identified with nestin and BrdU antibodies. The stem cell-derived differentiated cells expressed myosin VIIA, GFAP and NeuN. SEM was performed in order to determine the shape and surface structure of hair cells. Hair cells in culture were identified with stereocilia of cell surface. We found that hair cells with stereocilia were flat and irregularly shaped. We also found bipolar neuron-like cells.
     2. Decreased level of cyclin A2 in rat cochlea development and cochlear stem cell differentiation
     In order to detect the distribution of cyclin A2 protein in the rat cochlea, the immunohistochemical staining method was used in our study. P2 (postnatal day 2), P10 and adult (P42) rats were deeply anesthetized and exsanguinated, then decapitated. The cochlea tissues were excised carefully and then fixed in 4% paraformaldehyde in PBS at 4°C overnight. The P2 samples were decalcified for 1 day. The P10 and P42 samples were decalcified for 10 days. All samples were then immersed in 30% sucrose in PBS for 1 day and embedded carefully in optimal cutting temperature (OCT) compound. Sections (10μm thick) were cut on a cryostat at -27°C and mounted on poly-l-lysine treated glass slides. The immunohistochemistry of cyclin A2 in the rat cochlea was performed by streptavidin–biotin complex (SABC) staining. The primary antibody was mouse anti-cyclin A monoclonal antibody, and the second antibody was biotinylated horse anti-mouse IgG. For negative control, an equivalent dilution of normal mouse IgG was used in place of the primary antibody. We used immunocytochemistry to detect cyclin A2 protein expression in differentiated cells and cell spheres.
     We used RT-PCR to detect cyclin A2 mRNA expression in cochlea tissues, cochlear stem cells and differentiated cells. The cochlea tissues were excised from P0, P2, P14 and P42 rats respectively. The differentiated cells cultured for 6 or 14 days and cell spheres were collected for further assay. Total RNA was extracted. Then cDNA was amplified by PCR. The cyclin A2 PCR product was sequenced.
     Western blotting was used to detect cyclin A2 protein expression in cochlear stem cells, differentiated cells cultured for 14 days, and cochlea tissues (P0 and P14). Each protein sample was electrophoresed onto 12% SDS-polyacrylamide gel and transferred onto the nitrocellulose membranes. The membranes were incubated with mouse anti-cyclin A monoclonal antibody. The membranes were then incubated with the HRP-conjugated goat anti-mouse IgG. The membranes were also probed with anti-β-actin as internal control. The reaction product was visualized with enhanced chemiluminescence.
     With immunohistochemical staining, cyclin A2 was observed localizing in the spiral limbus of the P2 rat cochlea, but not in the cochlea of P10, P42 and the negative control. Cyclin A2 mRNA and protein levels in cultured cells fell down after differentiation of cochlear stem cells. Cyclin A2 level in cochlea tissues decreased from newborn to adult. The sequence of PCR product was determined to be identical to the originally published sequence of rat cyclin A2.
     3. Expression of CRP2 and CRIP2 in rat cochlea development and cochlear stem cell differentiation
     In order to detect the distribution of CRP2 and CRIP2 proteins in the rat cochlea of P2, P6 and P10, the immunohistochemical staining method was used. We used immunocytochemistry to detect CRP2 and CRIP2 proteins in differentiated cells and cell spheres.
     CRP2 protein level fell down during the development of cochlea tissues and differentiation of cochlear stem cells. While we found no difference of CRIP2 protein level in cochlea development and cochlear stem cell differentiation.
     4. Microenvironment of cochlear stem cell differentiation induces neural stem cells into myosin VIIA-positive cells and bipolar neuron-like cells
     Cocultures were performed on poly-L-lysine-coated coverslips in 6-well plates mixed with 1×10~5 CPCs and 1×10~4 NPCs per well. Cell spheres of NSCs and CPCs were dissociated by 0.025% trypsin and mechanical procedures. Then dissociated cells were mixed and allowed to attach for 16 h in wells filled with DMEM/F12 containing 10% FCS. After the cells were attached, we replaced the medium with serum-free DMEM/ F12 supplemented with N2 and B27 solutions. Half of the medium was replaced every second day. Cocultured cells were analyzed after 21 d by immunocytochemistry. Primary antibodies were as follows: rabbit anti-myosin VIIA polyclonal antibody, rabbit anti-peripherin polyclonal antibody. Cy3 conjugated second antibodies were used to detect primary antibodies. Hoechst 33342 was used to visualize nuclei. For negative controls, 1×10~5 NPCs were dissociated and cultured in 6-well plates with the same medium and methods as above.
     After 21 d in coculture, about 8% differentiated GFP-labeled NSCs expressed myosin VIIA. Myosin VIIA-positive cells were flat and irregularly shaped. About 1% differentiated NSCs expressed peripherin, a marker of peripheral neuron. Some of them were bipolar neuron-like cells. We also observed that spontaneous neural cell apoptosis which occurred in adherent cultures was delayed in the presence of differentiated CPCs. After 21 d in culture, no differentiated NPC was alive in negative controls, while the number of living differentiated NSCs among cocultured cells was about 8×103 per well.
     5. Methods for transplantation of stem cells into the cochlea of chickens and mature guinea pigs
     NSCs labeled with GFP were transplanted into chicken's cochlea via cochlear window after exposure of middle ear cavity. For cell transplantation into cochlear perilymphatic space of guinea pigs, NSCs were inoculated via a small hole drilled on the bony wall of scala tympani in the basal turn of the cochlea. For cell transplantation into scala media, a small hole was made to penetrate through the stria vascularis in the third turn of the cochlea. NSCs were then injected into the scala media through the fenestra. One day after transplantation, the transplant cells were examined with fluorescent microscope.
     After transplantation, NSCs labeled with GFP were observed in chicken's cochlea. In the cochlea tissues of guinea pigs, NSCs were detected in scala tympani and scala media, respectively. The methods for transplantation of stem cells into the cochlea of chickens and mature guinea pigs were established, which provides a basis for application of stem cells in studies of sensorineural hearing loss.
引文
1. Malgrange B, Belachew S, Thiry M, Nguyen L, Rogister B, Alvarez ML, Rigo JM, Van De Water TR, Moonen G, Lefebvre PP. Proliferative generation of mammalian auditory hair cells in culture. Mech. Dev. 2002;112(1-2):79-88.
    2. Li H, Liu H, Heller S. Pluripotent stem cells from the adult mouse inner ear. Nat Med. 2003;9(10):1293-1299.
    3. Lou X, Zhang Y, Yuan C. Multipotent stem cells from the young rat inner ear. Neurosci Lett. 2007;416(1):28-33.
    4.郑鸿燕,邰浩清,周春祥,吴颢昕,詹臻,王明艳.耳蜗前体细胞体外培养和诱导分化的实验研究.复旦学报(医学版). 2006;33(5):601-605.
    5. Oshima K, Grimm CM, Corrales CE, Senn P, Martinez Monedero R, Géléoc GS, Edge A, Holt JR, Heller S. Differential distribution of stem cells in the auditory and vestibular organs of the inner ear. J Assoc Res Otolaryngol. 2007;8(1):18-31.
    6. Savary E, Hugnot JP, Chassigneux Y, Travo C, Duperray C, Van De Water T, Zine A. Distinct population of hair cell progenitors can be isolated from the postnatal mouse cochlea using side population analysis. Stem Cells. 2007;25(2):332-339.
    7. Savary E, Sabourin JC, Santo J, Hugnot JP, Chabbert C, Van De Water T, Uziel A, Zine A. Cochlear stem/progenitor cells from a postnatal cochlea respond to Jagged1 and demonstrate that notch signaling promotes sphere formation and sensory potential. Mech Dev. 2008;125(8):674-686.
    8. Raft S, Koundakjian EJ, Quinones H, Jayasena CS, Goodrich LV, Johnson JE,Segil N, Groves AK. Cross-regulation of Ngn1 and Math1 coordinates the production of neurons and sensory hair cells during inner ear development. Development. 2007;134(24):4405-4415.
    9. Lowenheim H, Furness DN, Kil J, Zinn C, Gültig K, Fero ML, Frost D, Gummer AW, Roberts JM, Rubel EW, Hackney CM, Zenner HP. Gene disruption of p27(Kip1) allows cell proliferation in the postnatal and adult organ of corti. Proc Natl Acad Sci U S A. 1999;96(7):4084-4088.
    10. Kanzaki S, Beyer LA, Swiderski DL, Izumikawa M, St?ver T, Kawamoto K, Raphael Y. p27(Kip1) deficiency causes organ of Corti pathology and hearing loss. Hear Res. 2006;214(1-2):28-36.
    11. Chen P, Zindy F, Abdala C, Liu F, Li X, Roussel MF, Segil N. Progressive hearing loss in mice lacking the cyclin-dependent kinase inhibitor Ink4d. Nat Cell Biol. 2003;5(5):422-426.
    12. Sage C, Huang M, Karimi K, Gutierrez G, Vollrath MA, Zhang DS, García-A?overos J, Hinds PW, Corwin JT, Corey DP, Chen ZY. Proliferation of functional hair cells in vivo in the absence of the retinoblastoma protein. Science. 2005;307(5712):1114-1118.
    13. Liu Z, Zuo J. Cell cycle regulation in hair cell development and regeneration in the mouse cochlea. Cell Cycle. 2008;7(14):2129-2133.
    14. Dong Y, Nakagawa T, Endo T, Kim TS, Iguchi F, Yamamoto N, Naito Y, Ito J. Role of the F-box protein Skp2 in cell proliferation in the developing auditory system in mice. Neuroreport. 2003;14(5):759-761.
    15. Minoda R, Izumikawa M, Kawamoto K, Zhang H, Raphael Y. Manipulating cell cycle regulation in the mature cochlea. Hear Res. 2007;232(1-2):44-51.
    16.汪学勇,胡吟燕,翟所强. p27Kip在不同时期大鼠耳蜗大上皮嵴的表达.中华耳科学杂志. 2005;3(1):50-53.
    17. Hawkins RD, Bashiardes S, Helms CA, Hu L, Saccone NL, Warchol ME, Lovett M. Gene expression differences in quiescent versus regenerating hair cells of avian sensory epithelia: implications for human hearing and balance disorders. Hum Mol Genet. 2003;12(11):1261-1272.
    18.高雪,薛嬴,孙景豫,邱建华.小鼠CSRP2基因的克隆、融合表达及蛋白的分离纯化.生物技术通讯. 2007;18(6):918-920.
    19. Xue Gao, Zengyu Cao, Jun Chen, Shunli Liu, Dingjun Zha, Feng Wang, Yang Chen, Li Qiao, Lianjun Lu, Jianhua Qiu. Expression and subcellular localization of cysteine- and glycine-rich protein-2 in rat undifferentiated olfactory stem cells. Neural Regen Res. 2008;3(5):558-560.
    20. Ge RS, Hardy MP. Decreased cyclin A2 and increased cyclin G1 levels coincide with loss of proliferative capacity in rat Leydig cells during pubertal development. Endocrinology. 1997;138(9):3719-3726.
    21. Ravnik SE, Wolgemuth DJ. The developmentally restricted pattern of expression in the male germ line of a murine cyclin A, cyclin A2, suggests roles in both mitotic and meiotic cell cycles. Dev Biol .1996;173:69-78.
    22. Yoshizumi M, Lee WS, Hsieh CM, Tsai JC, Li J, Perrella MA, Patterson C, Endege WO, Schlegel R, Lee ME. Disappearance of cyclin A correlates with permanent withdrawal of cardiomyocytes from the cell cycle in human and rat hearts. J Clin Invest. 1995;95:2275-2280.
    23. Girard F, Strausfeld U, Fernandez A, Lamb NJ. Cyclin A is required for the onset of DNA replication in mammalian fibroblasts. Cell. 1991;67:1169-1179.
    24. Pagano M, Pepperkok R, Verde F, Ansorge W, Drawtta G. Cyclin A is required at two points in the human cell cycle. EMBO J 1992;11(3):961-971.
    25. Woo YJ, Panlilio CM, Cheng RK, Liao GP, Atluri P, Hsu VM, Cohen JE,Chaudhry HW. Therapeutic delivery of cyclin A2 induces myocardial regeneration and enhances cardiac function in ischemic heart failure. Circulation. 2006;114(1 Suppl):I206- I213.
    26.刘少锋,周梁,李华伟. Notch信号途径与哺乳动物的内耳感觉上皮发育.国外医学耳鼻咽喉科学分册. 2003;27(4):220-222.
    27.刘少锋,周梁,李华伟.脊椎动物内耳发育的基因调控.国外医学耳鼻咽喉科学分册. 2003;27(6):352-353.
    28.张媛,翟所强.哺乳动物内耳毛细胞再生研究现状.国际耳鼻咽喉头颈外科杂志. 2006;30(4):226-228.
    29.倪坤,黄红彦. bHLH转录因子在耳蜗发育中的作用.国际耳鼻咽喉头颈外科杂志. 2006;30(6):343-346.
    30.韩朝,迟放鲁. bHLH转录因子家族成员Math1及其在内耳的研究进展.复旦学报(医学版). 2007;4(4):623-626.
    31.张贤芬,杨仕明,韩东一. Math1基因及其研究进展.听力学及言语疾病杂志. 2007;15(6):497-501.
    32.唐珂,景乃禾.神经干细胞分化与bHLH转录调控因子.中国神经科学杂志. 2000;16(4): 360-364.
    33.贺强,王立峰. bHLH蛋白家族的功能.国外医学生理、病理科学与临床分册. 2004;24(6):545-547.
    34. Bermingham NA, Hassan BA, Price SD, Vollrath MA, Ben-Arie N, Eatock RA, Bellen HJ, Lysakowski A, Zoghbi HY. Math1: an essential gene for the generation of inner ear hair cells. Science. 1999;284(5421):1837-1841.
    35. Shou J, Zheng JL, Gao WQ. Robust generation of new hair cells in the mature mammalian inner ear by adenoviral expression of Hath1. Mol Cell Neurosci. 2003;23(2):169-179.
    36. Woods C, Montcouquiol M, Kelley MW. Math1 regulates development ofthe sensory epithelium in the mammalian cochlea. Nat Neurosci. 2004;7(12):1310-1318.
    37. Zhai S, Shi L, Wang BE, Zheng G, Song W, Hu Y, Gao WQ. Isolation and culture of hair cell progenitors from postnatal rat cochleae. J Neurobiol. 2005;65(3):282-293.
    38.张媛,孙建和,郭维,胡吟燕,翟所强. Hath1基因转染后大鼠耳蜗毛细胞前体细胞的扫描电镜观察.中国耳鼻咽喉头颈外科. 2006;13(5):309-312.
    39. Zhang Y, Zhai SQ, Shou J, Song W, Sun JH, Guo W, Zheng GL, Hu YY, Gao WQ. Isolation, growth and differentiation of hair cell progenitors from the newborn rat cochlear greater epithelial ridge. J Neurosci Methods. 2007;164(2):271-279.
    40. Zheng JL, Shou J, Guillemot F, Kageyama R, Gao WQ. Hes1 is a negative regulator of inner ear hair cell differentiation. Development. 2000;127(21):4551-4560.
    41. Zine A, Aubert A, Qiu J, Therianos S, Guillemot F, Kageyama R, de Ribaupierre F. Hes1 and Hes5 activities are required for the normal development of the hair cells in the mammalian inner ear. J Neurosci. 2001;21(13):4712-4720.
    42. Zine A, de Ribaupierre F. Notch/Notch ligands and Math1 expression patterns in the organ of Corti of wild-type and Hes1 and Hes5 mutant mice. Hear Res. 2002;170(1-2):22-31.
    43. Zine A. Molecular mechanisms that regulate auditory hair-cell differentiation in the mammalian cochlea. Mol Neurobiol. 2003;27(2):223-238.
    44. Bryant J, Goodyear RJ, Richardson GP. Sensory organ development in the inner ear: molecular and cellular mechanisms. Br Med Bull. 2002;63:39-57.
    45. Kelley MW. Regulation of cell fate in the sensory epithelia of the inner ear. Nat Rev Neurosci. 2006;7(11):837-849.
    46. Kelley MW. Cellular commitment and differentiation in the organ of Corti. Int J Dev Biol. 2007;51(6-7):571-583.
    47. Li S, Mark S, Radde-Gallwitz K, Schlisner R, Chin MT, Chen P. Hey2 functions in parallel with Hes1 and Hes5 for mammalian auditory sensory organ development. BMC Dev Biol. 2008;8:20.
    48. 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;102:4789-4799.
    49. Hu Z, Wei D, Johansson CB, Holmstrom N, Duan M, Frisen J, Ulfendahl M. Survival and neural differentiation of adult neural stem cells transplanted into the mature inner ear. Exp Cell Res. 2005;302:40-47.
    50. Altschuler RA, O'Shea KS, Miller JM. Stem cell transplantation for auditory nerve replacement. Hear Res. 2008;242(1-2):110-116.
    51. Coleman B, Hardman J, Coco A, Epp S, de Silva M, Crook J, Shepherd R. Fate of embryonic stem cells transplanted into the deafened mammalian cochlea. Cell Transplant. 2006;15:369-380.
    52. Parker MA, Corliss DA, Gray B, Anderson JK, Bobbin RP, Snyder EY, Cotanche DA. Neural stem cells injected into the sound-damaged cochlea migrate throughout the cochlea and express markers of hair cells, supporting cells, and spiral ganglion cells. Hear Res. 2007;232:29-43.
    53. Matsuoka AJ, Kondo T, Miyamoto RT, Hashino E. In vivo and in vitro characterization of bone marrow-derived stem cells in the cochlea. Laryngoscope. 2006;116:1363-1367.
    54. Matsuoka AJ, Kondo T, Miyamoto RT, Hashino E. Enhanced survival of bone-marrow-derived pluripotent stem cells in an animal model of auditory neuropathy. Laryngoscope. 2007;117(9):1629-1635.
    55.邱建华.现实与希望.中国耳鼻咽喉头颈外科. 2009;16(1):1-2.
    56. Corrales CE, Pan L, Li H, Liberman MC, Heller S, Edge AS. Engraftment and differentiation of embryonic stem cell-derived neural progenitor cells in the cochlear nerve trunk: growth of processes into the organ of Corti. J Neurobiol. 2006;66:1489-1500.
    57. Sekiya T, Kojima K, Matsumoto M, Kim TS, Tamura T, Ito J. Cell transplantation to the auditory nerve and cochlear duct. Exp Neurol. 2006;198(1):12-24.
    58. Chen Y, Qiu J, Chen F, Liu S. Migration of neural precursor cells derived from olfactory bulb in cochlear nucleus exposed to an augmented acoustic environment. Hear Res. 2007;228(1-2):3-10.
    59.汪红群,王正敏,沈云珍,李雯.新生大鼠耳蜗增殖细胞的培养鉴定及超微结构观察.中华耳鼻咽喉头颈外科杂志. 2007;42(1):42-47.
    60. Simile MM, De Miglio MR, Muroni MR, Frau M, Asara G, Serra S, Muntoni MD, Seddaiu MA, Daino L, Feo F, Pascale RM. Down-regulation of c-myc and Cyclin D1 genes by antisense oligodeoxy nucleotides inhibits the expression of E2F1 and in vitro growth of HepG2 and Morris 5123 liver cancer cells. Carcinogenesis. 2004;25(3):333-341.
    61. Chaudhry HW, Dashoush NH, Tang H, Zhang L, Wang X, Wu EX, Wolgemuth DJ. Cyclin A2 mediates cardiomyocyte mitosis in the postmitotic myocardium. J Biol Chem. 2004;279(34):35858-35866.
    62. Han Y, Qiu J, Qiao L, Zhong C. Gentamicin-induced deafness may be reversed by restarting cell cycle. Med Hypotheses. 2009;72(3):368-369.
    63. Chen J, Wang F, Gao X, Zha DJ, Xue T, Cheng X, Zhong CP, Han Y, Qiu JH. Decreased level of cyclin A2 in rat cochlea development and cochlear stem cell differentiation. Neurosci Lett. 2009;453(3):166-169.
    64. van Ham M, Croes H, Schepens J, Fransen J, Wieringa B, Hendriks W. Cloning and characterization of mCRIP2, a mouse LIM-only protein that interacts with PDZ domain IV of PTP-BL. Genes Cells. 2003;8(7):631-644.
    65. Weiskirchen R, Pino JD, Macalma T, Bister K, Beckerle MC. The cysteine-rich protein family of highly related LIM domain proteins. J Biol Chem. 1995;270(48):28946-28954.
    66.陈阳,王枫,陈俊,高雪,米文娟,邱建华. C57BL/6-gfp小鼠嗅球神经干细胞的培养鉴定及耳蜗核移植.中国耳鼻咽喉头颈外科杂志. 2009;16(1):36-39.
    67.孙建军,汪吉宝,李桂榕.雏鸡内耳听器的比较解剖学研究.临床耳鼻咽喉科杂志. 1994;8:348-350.
    68.张鄂,龚树生.干细胞移植在治疗感音神经性聋方面的研究进展.国外医学耳鼻咽喉科学分册. 2005;29:372-375.
    69. Jeon SJ, Oshima K, Heller S, Edge AS. Bone marrow mesenchymal stem cells are progenitors in vitro for inner ear hair cells. Mol Cell Neurosci. 2007;34:59-68.
    70. Sekiya T, Kojima K, Matsumoto M, Holley MC, Ito J. Rebuilding lost hearing using cell transplantation. Neurosurgery. 2007;60(3):417-433
    71. Beisel K, Hansen L, Soukup G, Fritzsch B. Regenerating cochlear hair cells: quo vadis stem cell. Cell Tissue Res. 2008;333(3):373-379.
    72. White PM, Doetzlhofer A, Lee YS, Groves AK, Segil N. Mammalian cochlear supporting cells can divide and trans-differentiate into hair cells. Nature. 2006;441(7096):984-987.
    73.钟时勋,刘兆华.豚鼠耳蜗血管纹黑色素的分布及其合成.第三军医大学学报. 2000;22:645-648.
    74. Ishimoto S, Kawamoto K, Kanzaki S, Raphael Y. Gene transfer into supporting cells of the organ of Corti. Hear Res. 2002;173(1-2):187-197.

© 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