大鼠颈脊髓不同传导束损伤后差异性基因表达的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:比较大鼠红核脊髓束和皮质脊髓背侧束损伤后24h的差异性基因表达,并对其行基因本体论和信号通路的分析,从基因层面探索脊髓损伤后的修复是否遵循进化论的原则,为将来以进化论为指导治疗脊髓损伤奠定基础。
     方法:使用SAM软件对前期实验中得到的基因表达谱芯片结果进行分析,筛选和比较两种传导束损伤后基因的差异性表达;并通过MAS、GO、KEGG和GenMapp对生物过程和信号通路进行分析和比较。
     选取8个感兴趣的基因:Fos, Ntrk1, Gadd45α, Myc, Sox7, Tnfrsf12a,Slc5a3和Rnf6,设计引物行RT-PCR验证。实验选取雌性SD大鼠12只,体重200-230g,平均220g,随机分为4组:双侧皮质脊髓背侧束+薄束楔束损伤组3只,双侧薄束楔束损伤组3只,双侧红核脊髓束损伤组3只,假手术组3只。建立动物模型后24h后处死动物,以损伤处为中心取5mm脊髓组织,将每组中的标本混合提取总RNA行RT-PCR检验,并将结果与基因芯片结果进行比较。
     结果:两种传导束损伤后24h基因表达变化存在差异,红核脊髓束损伤后表达发生变化的基因有153个,上调的有136个,下调的有17个。皮质脊髓背侧束损伤后表达发生变化的基因有26个,上调的有22个,下调的有4个。红核脊髓束损伤后发生显著性变化(P<0.05)的生物过程条目为164条,25条涉及到3个及以上的基因变化;发生显著性变化(P<0.05)的信号通路为25条,17条涉及到3个及以上的基因变化。皮质脊髓背侧束损伤后发生显著性变化(P<0.05)的生物过程条目为7条,发生显著性变化(P<0.05)的信号通路为1条,涉及到的基因均少于3个。这些基因主要都涉及到炎症、免疫、抗原处理及呈递等急性期反应,少数与神经发育和轴突再生相关。
     RT-PCR验证结果显示两种传导束损伤后Myc、Fos、Tnfrsfl 2a和Gadd45a的变化和基因芯片结果完全一致,Sox7和Rnf6在红核脊髓束损伤后表达无显著性变化,Ntrkl和Slc5a3基因在红核脊髓束损伤后表达和基因芯片结果相反。
     结论:红核脊髓束损伤后24h基因表达变化多于皮质脊髓背侧束损伤,该变化与损伤面积大小无关,是否与进化程度有关还需进一步验证。红核脊髓束损伤后利于神经再生的基因如Fos、TrkA表达上调高于皮质脊髓背侧束损伤组,利于轴突再生的信号通路如MAPK也被激活,在基因水平上支持红核脊髓束的修复能力强于皮质脊髓背侧束,但还需要蛋白质层面和组织学层面的进一步验证。红核脊髓束损伤后其他显著性上调的基因如Tnfrsfl2a和Gadd45a与神经再生有无直接关系值得进一步的研究。
Objective:To compare the different gene expressional changes and difference in gene ontology and pathway between RST and dCST after transection at 24h; try to explain different regeneration abilities of different evolved spinal cord tracts using evolutionism at the gene level; and provide a robust framework in the treatment of SCI in the light of evolutionism.
     Methods:Gene expressional profiles obtained in former experiments were analyzed, and the differentially expressed genes were screened and compared by SAM. The biological processes and pathways between RST and dCST were analyzed by MAS, GO, Kegg, and GenMAPP.
     To corroborate and validate microarray results, Fos, Ntrkl, Gadd45a, Myc, Sox7, Tnfrsfl2a, Slc5a3 and Rnf6 were selected for RT-PCR test.12 female Sprague-Dawley rats, which weighed 200-230g (average 220g), were divided randomly into 4 groups:bilateral dCST+FCFG transection group (n=3), bilateral FCFG transection group (n=3), bilateral RST transection group (n=3) and sham operation group (n=3). Rats were sacrificed24h after injury,5mm length spinal cord specimens centered injury site were procured. Total RNA were extracted and purified. RT-PCR was performed and the results were compared with results of microarray analysis.
     Results:Differences of gene expressional profiles exist between RST and dCST after injury. The expression of 153 genes changed after transection of RST, 136 were up-regulated and 17 were down-regulated. The expression of 26 genes changed after transection of dCST,22 were up-regulated and 4 were down-regulated. The number of biological process terms significantly changed (P < 0.05) after RST transection was 164,25 terms involved with no less than 3 gene counts; the number of pathways significantly changed (P< 0.05) was 25,17 terms involved with no less than 3 gene counts. The number of biological process terms significantly changed (P< 0.05) after dCST transection was 7, the number of pathways significantly changed (P< 0.05) was 1, all involved with less than 3 gene counts. Most of the functions were related to acute phase response such as immune response, antigen processing and presentation, inflammatory response. Few genes were related to neural regeneration.
     The expressional changes of Myc, Fos, Tnfrsfl2a, and Gadd45a tested by RT-PCR were in accordance with the results of microarray analysis. The expressional changes of Sox7 and Rnf6 showed no significant changes after RST transection. The expressional changes of Ntrkl and Slc5a3 after RST transection were contrast to the results of microarray analysis.
     Conclusion:The different expressional changes of genes have nothing to do with injured area. Further experiments should be performed to testify if it is related to different evolution. Genes related to neural regeneration such as Fos and Ntrkl were more up-regulated after RST transection, pathways related to axon regeneration such as MAPK pathway was activated after RST transection, which suggest RST has stronger regeneration ability at the gene level. However, this should be testified at the protein and histology level. Other genes significantly up-regulated after RST injury, such as Tnfrsfl2a and Gadd45a may have relationship with axon regeneration, and deserves further research.
引文
[1]Wyndaele, M. and J.J. Wyndaele, Incidence, prevalence and epidemiology of spinal cord injury:what learns a worldwide literature survey? Spinal Cord, 2006.44(9):p.523-9.
    [2]Li, Y., P.M. Field, and G. Raisman, Repair of adult rat corticospinal tract by transplants of olfactory ensheathing cells. Science,1997.277(5334):p. 2000-2.
    [3]Keyvan-Fouladi, N., G. Raisman, and Y. Li, Functional repair of the corticospinal tract by delayed transplantation of olfactory ensheathing cells in adult rats. J Neurosci,2003.23(28):p.9428-34.
    [4]Garcia-Alias G, Lopez-Vales R, Fores J, et al., Acute transplantation of olfactory ensheathing cells or Schwann cells promotes recovery after spinal cord injury in the rat. J Neurosci Res,2004.75(5):p.632-41.
    [5]Ramer LM, Au E, Richter MW, et al., Peripheral olfactory ensheathing cells reduce scar and cavity formation and promote regeneration after spinal cord injury. J Comp Neurol,2004.473(1):p.1-15.
    [6]Mackay-Sim A, Feron F, Cochrane J, et al., Autologous olfactory ensheathing cell transplantation in human paraplegia:a 3-year clinical trial. Brain,2008. 131(Pt9):p.2376-86.
    [7]Lima C, Pratas-Vital J, Escada P, et al., Olfactory mucosa autografts in human spinal cord injury:a pilot clinical study. J Spinal Cord Med,2006.29(3):p. 191-203; discussion 204-6.
    [8]孙天胜,任继鑫,吴军等,嗅鞘细胞移植促进脊髓损伤修复的长期实验和临床观察.中国临床康复,2005.9(2):p.122-124.
    [9]Northcutt, R.G., Evolution of the vertebrate central nervous system:patterns and processes.1984, Soc Integ Comp Biol. p.701.
    [10]孙天胜,以进化论的观点评价细胞移植治疗脊髓损伤。中国脊柱脊髓杂志,2007.17(9):p.651-653.
    [11]Dobzhansky, T., Nothing in biology makes sense except in the light of evolution.1973, National Association of Biology Teachers, p.125-129.
    [12]Piper, M. and C. Holt, RNA translation in axons. Annu Rev Cell Dev Biol, 2004.20:p.505-23.
    [13]Donnelly, D.J. and P.G. Popovich, Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol,2008.209(2):p.378-88.
    [14]Saville LR, Pospisil CH, Mawhinney LA, et al., A monoclonal antibody to CD11d reduces the inflammatory infiltrate into the injured spinal cord:a potential neuroprotective treatment. J Neuroimmunol,2004.156(1-2):p. 42-57.
    [15]Stirling DP, Khodarahmi K, Liu J, et al., Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J Neurosci,2004.24(9):p. 2182-90.
    [16]Nguyen, H.X., T.J. O'Barr, and A.J. Anderson, Polymorphonuclear leukocytes promote neurotoxicity through release of matrix metalloproteinases, reactive oxygen species, and TNF-alpha. J Neurochem,2007.102(3):p.900-12.
    [17]Dinkel, K., F.S. Dhabhar, and R.M. Sapolsky, Neurotoxic effects of polymorphonuclear granulocytes on hippocampal primary cultures. Proc Natl Acad Sci U S A,2004.101(1):p.331-6.
    [18]Howe CL, Ure D, Adelson JD, et al., CD8+ T cells directed against a viral peptide contribute to loss of motor function by disrupting axonal transport in a viral model of fulminant demyelination. J Neuroimmunol,2007.188(1-2):p. 13-21.
    [19]Crutcher KA, Gendelman HE, Kipnis J, et al., Debate:"is increasing neuroinflammation beneficial for neural repair?" J Neuroimmune Pharmacol, 2006.1(3):p.195-211.
    [20]Yin Y, Henzl MT, Lorber B, et al., Oncomodulin is a macrophage-derived signal for axon regeneration in retinal ganglion cells. Nat Neurosci,2006.9(6): p.843-52.
    [21]Kaczorowski DJ, Mollen KP, Edmonds R, et al., Early events in the recognition of danger signals after tissue injury. J Leukoc Biol,2008.83(3):p. 546-52.
    [22]Medzhitov, R., P. Preston-Hurlburt, and C.A. Janeway, Jr., A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature,1997.388(6640):p.394-7.
    [23]Pearson G, Robinson F, Beers Gibson T, et al., Mitogen-activated protein (MAP) kinase pathways:regulation and physiological functions. Endocr Rev, 2001.22(2):p.153-83.
    [24]Davis, R.J., The mitogen-activated protein kinase signal transduction pathway. J Biol Chem,1993.268(20):p.14553-6.
    [25]Hammarlund M, Nix P, Hauth L, et al., Axon regeneration requires a conserved MAP kinase pathway. Science,2009.323(5915):p.802-6.
    [26]Yan D, Wu Z, Chisholm AD, et al., The DLK-1 kinase promotes mRNA stability and local translation in C. elegans synapses and axon regeneration. Cell,2009.138(5):p.1005-18.
    [27]Pan YA, Misgeld T, Lichtman JW, et al., Effects of neurotoxic and neuroprotective agents on peripheral nerve regeneration assayed by time-lapse imaging in vivo. J Neurosci,2003.23(36):p.11479-88.
    [28]Woolf CJ, Reynolds ML, Molander C, et al., The growth-associated protein GAP-43 appears in dorsal root ganglion cells and in the dorsal horn of the rat spinal cord following peripheral nerve injury. Neuroscience,1990.34(2):p. 465-78.
    [29]Willis D, Li KW, Zheng JQ, et al., Differential transport and local translation of cytoskeletal, injury-response, and neurodegeneration protein mRNAs in axons. J Neurosci,2005.25(4):p.778-91.
    [30]Morgan, J.I. and T. Curran, Stimulus-transcription coupling in neurons:role of cellular immediate-early genes. Trends Neurosci,1989.12(11):p.459-62.
    [31]Yu AC, Lee YL, Fu WY, et al., Gene expression in astrocytes during and after ischemia. Prog Brain Res,1995.105:p.245-53.
    [32]Chao, M.V., Neurotrophins and their receptors:a convergence point for many signalling pathways. Nat Rev Neurosci,2003.4(4):p.299-309.
    [33]Lee R, Kermani P, Teng KK, et al., Regulation of cell survival by secreted proneurotrophins. Science,2001.294(5548):p.1945-8.
    [34]Huang, E.J. and L.F. Reichardt, Trk receptors:roles in neuronal signal transduction. Annu Rev Biochem,2003.72:p.609-42.
    [35]Dickson, B.J., Molecular mechanisms of axon guidance. Science,2002. 298(5600):p.1959-64.
    [36]DiAntonio, A. and L. Hicke, Ubiquitin-dependent regulation of the synapse. Annu Rev Neurosci,2004.27:p.223-46.
    [37]van Horck, F.P., C. Weinl, and C.E. Holt, Retinal axon guidance:novel mechanisms for steering. Curr Opin Neurobiol,2004.14(1):p.61-6.
    [38]Rosso S, Bollati F, Bisbal M., et al., LIMK1 regulates Golgi dynamics, traffic of Golgi-derived vesicles, and process extension in primary cultured neurons. Mol Biol Cell,2004.15(7):p.3433-49.
    [39]Tursun B, Schluter A, Peters MA, et al., The ubiquitin ligase Rnf6 regulates local LIM kinase 1 levels in axonal growth cones. Genes Dev,2005.19(19):p. 2307-19.
    [40]Gearhart, J., E.E. Pashos, and M.K. Prasad, Pluripotency redux--advances in stem-cell research. N Engl J Med,2007.357(15):p.1469-72.
    [41]Dominguez-Sola D, Ying CY, Grandori C, et al., Non-transcriptional control of DNA replication by c-Myc. Nature,2007.448(7152):p.445-51.
    [42]Jackson, J.G. and O.M. Pereira-Smith, p53 is preferentially recruited to the promoters of growth arrest genes p21 and GADD45 during replicative senescence of normal human fibroblasts. Cancer Res,2006.66(17):p. 8356-60.
    [43]Liebermann, D.A. and B. Hoffman, Myeloid differentiation (MyD)/growth arrest DNA damage (GADD) genes in tumor suppression, immunity and inflammation. Leukemia,2002.16(4):p.527-41.
    [44]Bodo J, Jakubikova J, Chalupa I, et al., Apoptotic effect of ethyl-4-isothiocyanatobutanoate is associated with DNA damage, proteasomal activity and induction of p53 and p21cip1/waf1. Apoptosis,2006.11(8):p. 1299-310.
    [45]Zhan, Q., Gadd45a, a p53-and BRCA1-regulated stress protein, in cellular response to DNA damage. Mutat Res,2005.569(1-2):p.133-43.
    [46]常重杰,杜启燕, 邵红伟,Sox基因家族研究的新进展。遗传,2002.24(4): p.470-476.
    [47]Ashwal S, Holshouser B, Tong K, et al., Proton spectroscopy detected myoinositol in children with traumatic brain injury. Pediatr Res,2004.56(4):p. 630-8.
    [1]Fitch, M.T. and J. Silver, CNS injury, glial scars, and inflammation:Inhibitory extracellular matrices and regeneration failure. Exp Neurol,2008.209(2):p. 294-301.
    [2]Yiu, G. and Z. He, Glial inhibition of CNS axon regeneration. Nat Rev Neurosci,2006.7(8):p.617-27.
    [3]Lee JK, Chan AF, Luu SM, et al., Reassessment of corticospinal tract regeneration in Nogo-deficient mice. J Neurosci,2009.29(27):p.8649-54.
    [4]Ghosh-Roy A, Wu Z, Goncharov A, et al., Calcium and cyclic AMP promote axonal regeneration in Caenorhabditis elegans and require DLK-1 kinase. J Neurosci,2010.30(9):p.3175-83.
    [5]Moore SW, Correia JP, Lai Wing Sun K, et al., Rho inhibition recruits DCC to the neuronal plasma membrane and enhances axon chemoattraction to netrin 1. Development,2008.135(17):p.2855-64.
    [6]Pak, C.W., K.C. Flynn, and J.R. Bamburg, Actin-binding proteins take the reins in growth cones. Nat Rev Neurosci,2008.9(2):p.136-47.
    [7]Langhorst, M.F., A. Reuter, and C.A. Stuermer, Scaffolding microdomains and beyond:the function of reggie/flotillin proteins. Cell Mol Life Sci,2005. 62(19-20):p.2228-40.
    [8]Munderloh C, Solis GP, Bodrikov V, et al., Reggies/flotillins regulate retinal axon regeneration in the zebrafish optic nerve and differentiation of hippocampal and N2a neurons. J Neurosci,2009.29(20):p.6607-15.
    [9]Duffy P, Schmandke A, Schmandke A, et al., Rho-associated kinase II (ROCKII) limits axonal growth after trauma within the adult mouse spinal cord. J Neurosci,2009.29(48):p.15266-76.
    [10]Veldman, M.B., M.A. Bemben, and D. Goldman, Tubala gene expression is regulated by KLF6/7 and is necessary for CNS development and regeneration in zebrafish. Mol Cell Neurosci,2010.43(4):p.370-83.
    [11]Nikulina E, Tidwell JL, Dai HN, et al., The phosphodiesterase inhibitor rolipram delivered after a spinal cord lesion promotes axonal regeneration and functional recovery. Proc Natl Acad Sci U S A,2004.101(23):p.8786-90.
    [12]Cai D, Qiu J, Cao Z, et al., Neuronal cyclic AMP controls the developmental loss in ability of axons to regenerate. J Neurosci,2001.21(13):p.4731-9.
    [13]Gao Y, Deng K, Hou J, et al., Activated CREB is sufficient to overcome inhibitors in myelin and promote spinal axon regeneration in vivo. Neuron, 2004.44(4):p.609-21.
    [14]Cai D, Deng K, Mellado W, et al., Arginase I and polyamines act downstream from cyclic AMP in overcoming inhibition of axonal growth MAG and myelin in vitro. Neuron,2002.35(4):p.711-9.
    [15]Deng K, He H, Qiu J, et al., Increased synthesis of spermidine as a result of upregulation of arginase I promotes axonal regeneration in culture and in vivo. J Neurosci,2009.29(30):p.9545-52.
    [16]Sun, F. and Z. He, Neuronal intrinsic barriers for axon regeneration in the adult CNS. Curr Opin Neurobiol.20(4):p.510-8.
    [17]Raivich, G. and M. Makwana, The making of successful axonal regeneration: genes, molecules and signal transduction pathways. Brain Res Rev,2007. 53(2):p.287-311.
    [18]Yan D, Wu Z, Chisholm AD, et al., The DLK-1 kinase promotes mRNA stability and local translation in C. elegans synapses and axon regeneration. Cell,2009.138(5):p.1005-18.
    [19]Massaro, C.M., J. Pielage, and G.W. Davis, Molecular mechanisms that enhance synapse stability despite persistent disruption of the spectrin/ankyrin/microtubule cytoskeleton. J Cell Biol,2009.187(1):p. 101-17.
    [20]Raivich G, Bohatschek M, Da Costa C, et al., The AP-1 transcription factor c-Jun is required for efficient axonal regeneration. Neuron,2004.43(1):p. 57-67.
    [21]Takahashi K, Tanabe K, Ohnuki M, et al., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell,2007.131(5):p.861-72.
    [22]Veldman MB, Bemben MA, Thompson RC, et al., Gene expression analysis of zebrafish retinal ganglion cells during optic nerve regeneration identifies KLF6a and KLF7a as important regulators of axon regeneration. Dev Biol, 2007.312(2):p.596-612.
    [23]Moore DL, Blackmore MG, Hu Y, et al., KLF family members regulate intrinsic axon regeneration ability. Science,2009.326(5950):p.298-301.
    [24]Park KK, Liu K, Hu Y, et al., Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science,2008.322(5903):p. 963-6.
    [25]Carracedo, A. and P.P. Pandolfi, The PTEN-PI3K pathway:of feedbacks and cross-talks. Oncogene,2008.27(41):p.5527-41.
    [26]Ma XM, Yoon SO, Richardson CJ, et al., SKAR links pre-mRNA splicing to mTOR/S6K1-mediated enhanced translation efficiency of spliced mRNAs. Cell,2008.133(2):p.303-13.
    [27]Zhou, F.Q. and W.D. Snider, Cell biology. GSK-3beta and microtubule assembly in axons. Science,2005.308(5719):p.211-4.
    [28]Park KK, Liu K, Hu Y, et al., PTEN/mTOR and axon regeneration. Exp Neurol.223(1):p.45-50.
    [29]Raisman, G., Myelin inhibitors:does NO mean GO? Nat Rev Neurosci,2004. 5(2):p.157-61.

© 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