猪低氧适应相关DNA甲基化位点筛选及表观修饰酶表达分析
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
因为高原地区的低气压与低氧分压严重地影响着人类与动物的生存与发展,所以低氧反应和低氧适应一直都是国内外生物学和医学研究的重要热点之一。为了应对低氧,机体除了激活和稳定转录调控因子外,还会启动一些基因的DNA甲基化等表观调控机制。藏猪生活在青藏高原,能够适应恶劣的高寒气候和低劣的饲养管理条件,是研究低氧适应的宝贵的物种资源。而低氧适应是一个复杂的性状,至今藏猪适应低氧的分子遗传学机制以及表观调控机制尚未完全清楚。本研究选取生活在高原环境的藏猪(TH)和移居高原环境的大约克猪(YH),以及移居在低地饲养的藏猪(TL)和低地大约克猪(YL)4组猪群体作为研究对象,利用甲基化DNA免疫共沉淀(MeDIP),并结合二代测序技术来筛选影响低氧适应过程中DNA甲基化差异的关键基因,联合转录组测序结果分析,挑选基因进行DNA甲基化和表达量验证,旨在阐明藏猪低氧适应的表观遗传学机制,并筛选重要的低氧适应候选基因。本研究对5个重要的表观修饰酶进行了mRNA水平的半定量和定量分析,以揭示不同猪种的表观修饰酶的mRNA表达差异。
     MeDIP-Seq共得到486M reads,每个个体得到约60000个DNA甲基化peak。用MEDIPS软件包计算不同组别两两之间的差异甲基化区域(DMR),并利用UCSC数据库进行DMR的注释。对MeDIP-Seq得到的DMR进行聚类分析,结果显示环境对个体甲基化水平影响远小于品种的影响。对4种组间比较得到的差异甲基化基因,进行GO及KEGG Pathway富集分析后,发现低氧时,两品种差异甲基化基因与低氧适应相关通路(PPAR)及脂肪合成代谢相关;常氧变为低氧时,藏猪差异甲基化基因与低氧适应有关(VEGF、PPAR等通路)。
     从既差异甲基化又差异表达的基因中筛选到5个可能与低氧适应相关的基因:EPHX2、RXRG、 BCKDHB、UBD和GOT2。对于这5个基因通过克隆测序和荧光定量PCR方法进行验证基因的差异甲基化程度和差异表达水平。结果表明,RXRG基因的差异甲基化水平和差异表达水平与高通量测序结果一致,即它们同时在4组中差异甲基化和差异表达。而BCKDHB和EPHX2,虽然差异甲基化,但mRNA表达水平并不存在显著性差异,GOT2和UBD基因不存在甲基化差异位点,表达上也无显著性差异。但所有基因的DNA甲基化和mRNA表达在不同组间的趋势与高通量测序相同。
     5个表观修饰酶基因的半定量结果表明,它们在高原藏猪(TH)的表达具有组织特异性。Dnmt1和Uhrfl表达量最高的组织是心脏、肺脏、垂体,而其他三个基因表达量最高的组织均为肺脏、肾脏、垂体。肺脏中所有表观修饰酶在TH组的表达量均高于大约克猪;肾脏中,常氧变为低氧时,藏猪表观修饰酶表达量下调,而大约克猪表达上调。
     本研究利用高通量测序法对4组猪的心脏组织进行DNA甲基化测序,筛选并验证得到了RXRG基因在不同组间差异DNA甲基化和差异表达。对Dnmtl,Dnmt3a,Kdm3a,Hdac1, Uhrf15个表观修饰酶进行了表达分析,发现肺脏和肾脏可能是低氧适应中表观修饰相关功能器官,且肺脏中低氧时,可能藏猪大部分基因相对于大约克猪下调,肾脏中,常氧变为低氧时,低氧反应/适应可能导致大约克猪很多基因表达受到抑制。本研究对于完善藏猪低氧适应的表观遗传学调控提供了依据,也为进一步研究低氧适应相关基因提供了方向和目标。
Low pressure and low partial pressure of oxygen of plateau affects survival and development of human and animals, so hypoxia response and hypoxia adaptation are always focus of biological and medicine. To deal with hypoxia, the organism will activate and stable HIFs, also needs some epigenetic regulation, DNA methylation is one of the most important epigenetic machnisms. Tibet pigs live at Qinghai-Tibet Plateau, their adaptation to hypoxia makes them precious species resource and object for hypoxia adaptation machnism. We selected four groups of pig as the study object, which were Tibet pig in highland (TH), Yorkshire in highland (YH), Tibet pig in lowland (TL), Yorkshire in lowland (YL). Using methylated DNA immunoprecipitation and subsequent high throughput sequencing (MeDIP-Seq), and combining RNA-Seq results, we filtered key genes related to hypoxia adaptation that were differentially methylated and differentially expressed simultaneously. We conducted clone sequencing and real time PCR to validate these high throughput results. mRNA semi-quantitative and quantitative assay were conducted for five epigenetic modifying enzymes to reveal their expression differences.486M reads was obtained from MeDIP-Seq, and there were about60K peaks for each individual. Then we used MEDIPS package to obtain differentially methylated regions which were annotated in UCSC. Cluster analysis of differentially methylated genes (DMGs) from MeDIP-Seq assay showed that breed affect DNA methylation level much more than altitude. GO and KEGG pathway analysis results showed that, in condition of hypoxia, DMGs between two breeds were related to PPAR pathway and Fatty acid elongation GO term. From normoxia to hypoxia, Tibet pigs changed DNA methylation of hypoxia adaptation related pathway(VEGF and PPAR).
     We selected five genes that may play important roles in hypoxia adaptation from DMRs that were both differentially methylated and expressed:EPHX2, RXRG, BCKDHB, UBD and GOT2. Clone sequencing and real time quantitative PCR were implemented for these five genes, and validation results showed that RXRG was both differentially methylated and expressed between different groups; although BCKDHB and EPHX2was differentially methylated, there were no significantly difference between four groups; and there were neither significant difference of methylation nor significant difference of expression between different groups. The methylation difference and expression difference of these five genes had the same trend with that resulted from MeDIP-Seq and RNA-Seq.
     These five epigenetic modifying genes presented tissue specific expression patterns. Dnmtl and Uhrfl expressed highly in heart, lung and pituitary gland, and the other three genes expressed highly in lung, kidney and pituitary gland. In lung, all these genes expressed higher in TH than that in Yorkshire. In kidney, from nomoxia to hypoxia, genes expressed lower in Tibet pigs, but higher in Yorkshire.
     This research obtained DNA methylation profile of heart using four groups of pigs, then selected and validated five genes whih may play roles in hypoxia adaptation, validation turned out that RXRG was both differentially methylated and expressed between different groups. We also conducted semi-quantitative expression analysis and quantitative expression analysis of Dnmtl, Dnmt3a, Kdm3a, Hdacl and Uhrfl, results suggested that lung and kidney may play important roles of epigenetic regulation to adapt to hypoxia. In lung, in condition of hypoxia, most genes in Tibet pigs maybe were down regulated compared to Yorkshire. In kidney, from nomoxia to hypoxia, hypoxic response or adaptation may induce many genes' decreased expression. All these results will be useful to reveal the epigentic regulation of hypoxia adaptation and useful to provide suggestions to do research towards hypoxia adaptation related genes.
引文
1. Aerts S, Thijs G, Dabrowski M, et al. (2004) Comprehensive analysis of the base composition around the transcription start site in Metazoa. BMC genomics 5:34.
    2. Aggarwal S, Negi S, Jha P, et al. (2010) EGLN1 involvement in high-altitude adaptation revealed through genetic analysis of extreme constitution types defined in Ayurveda. Proceedings of the National Academy of Sciences 107:18961-18966.
    3. Aksyonov SA, Bittner M, Bloom LB, et al. (2006) Multiplexed DNA sequencing-by-synthesis. Anal Biochem 348:127-138.
    4. Alkorta-Aranburu G, Beall CM, Witonsky DB, et al. (2012) The genetic architecture of adaptations to high altitude in Ethiopia. PLoS genetics 8:e1003110.
    5. Avivi A, Resnick M, Nevo E, et al. (1999) Adaptive hypoxic tolerance in the subterranean mole rat< i> Spalax ehrenbergi:the role of vascular endothelial growth factor. FEBS letters 452:133-140.
    6. Azad P, Zhou D, Zarndt R, et al. (2012) Identification of genes underlying hypoxia tolerance in Drosophila by a P-element screen. G3:Genes|Genomes|Genetics 2:1169-1178.
    7. Ball MP, Li JB, Gao Y, et al. (2009) Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nature biotechnology 27:361-368.
    8. Bartke T, Vermeulen M, Xhemalce B, et al. (2010) Nucleosome-interacting proteins regulated by DNA and histone methylation. Cell 143:470-484.
    9. Bentley DR, Balasubramanian S, Swerdlow HP, et al. (2008) Accurate whole human genome sequencing using reversible terminator chemistry. Nature 456:53-59.
    10. Bertout JA, Patel SA and Simon MC. (2008) The impact of 02 availability on human cancer. Nature Reviews Cancer 8:967-975.
    11. Bigham AW, Mao X, Mei R, et al. (2009) Identifying positive selection candidate loci for high-altitude adaptation in Andean populations. Human genomics 4:79.
    12. Bird AP and Wolffe AP. (1999) Methylation-induced repression-belts, braces, and chromatin. Cell 99:451-454.
    13. Borland MG, Khozoie C, Albrecht PP, et al. (2011) Stable over-expression of PPARbeta/delta and PPARgamma to examine receptor signaling in human HaCaT keratinocytes. Cell Signal 23:2039-2050.
    14. Brahimi-Horn MC and Pouyssegur J. (2007) Harnessing the hypoxia-inducible factor in cancer and ischemic disease. Biochem Pharmacol 73:450-457.
    15. Brinkman AB, Simmer F, Ma K, et al. (2010) Whole-genome DNA methylation profiling using MethylCap-seq. Methods 52:232-236.
    16. Brunner AL, Johnson DS, Kim SW, et al. (2009) Distinct DNA methylation patterns characterize differentiated human embryonic stem cells and developing human fetal liver. Genome research 19:1044-1056.
    17. Cedar H and Bergman Y. (2009) Linking DNA methylation and histone modification:patterns and paradigms. Nat Rev Genet 10:295-304.
    18. Chavez L, Jozefczuk J, Grimm C, et al. (2010) Computational analysis of genome-wide DNA methylation during the differentiation of human embryonic stem cells along the endodermal lineage. Genome research 20:1441-1450.
    19. Chen T, Tsujimoto N and Li E. (2004) The PWWP domain of Dnmt3a and Dnmt3b is required for directing DNA methylation to the major satellite repeats at pericentric heterochromatin. Mol CellBiol 24:9048-9058.
    20. Chen Z-x and Riggs AD. (2011) DNA methylation and demethylation in mammals. Journal of Biological Chemistry 286:18347-18353.
    21. Cokus SJ, Feng S, Zhang X, et al. (2008) Shotgun bisulphite sequencing of the Arabidopsis genome reveals DNA methylation patterning. Nature 452:215-219.
    22. Dasgupta C, Chen M, Zhang H, et al. (2012) Chronic hypoxia during gestation causes epigenetic repression of the estrogen receptor-a gene in ovine uterine arteries via heightened promoter methylation. Hypertension 60:697-704.
    23. Dawson MA and Kouzarides T. (2012) Cancer epigenetics:from mechanism to therapy. Cell 150:12-27.
    24. Deaton AM and Bird A. (2011) CpG islands and the regulation of transcription. Genes Dev 25: 1010-1022.
    25. Dekanty A, Romero NM, Bertolin AP, et al. (2010) Drosophila Genome-Wide RNAi Screen Identifies Multiple Regulators of HIF-Dependent Transcription in Hypoxia. PLoS genetics 6: e1000994.
    26. Down TA, Rakyan VK, Turner DJ, et al. (2008) A Bayesian deconvolution strategy for immunoprecipitation-based DNA methylome analysis. Nature biotechnology 26:779-785.
    27. Erzurum SC, Ghosh S, Janocha AJ, et al. (2007) Higher blood flow and circulating NO products offset high-altitude hypoxia among Tibetans. Proc Natl Acad Sci USA 104: 17593-17598.
    28. Fatemi M, Hermann A, Pradhan S, et al. (2001) The activity of the murine DNA methyltransferase Dnmtl is controlled by interaction of the catalytic domain with the N-terminal part of the enzyme leading to an allosteric activation of the enzyme after binding to methylated DNA. Journal of molecular biology 309:1189-1199.
    29. Feinberg AP. (2007) Phenotypic plasticity and the epigenetics of human disease. Nature 447: 433-440.
    30. Feinberg AP and Irizarry RA. (2010) Stochastic epigenetic variation as a driving force of development, evolutionary adaptation, and disease. Proceedings of the National Academy of Sciences 107:1757-1764.
    31. Feng S, Cokus SJ, Zhang X, et al. (2010) Conservation and divergence of methylation patterning in plants and animals. Proc Natl Acad Sci USA 107:8689-8694.
    32. Frommer M, McDonald LE, Millar DS, et al. (1992) A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proceedings of the National Academy of Sciences 89:1827-1831.
    33. Fuks F, Burgers WA, Brehm A, et al. (2000) DNA methyltransferase Dnmtl associates with histone deacetylase activity. Nat Genet 24:88-91.
    34. Giatromanolaki A, Bai M, Margaritis D, et al. (2010) Hypoxia and activated VEGF/receptor pathway in multiple myeloma. Anticancer research 30:2831-2836.
    35. Giles FJ. (2001) The vascular endothelial growth factor (VEGF) signaling pathway:a therapeutic target in patients with hematologic malignancies. The Oncologist 6:32-39.
    36. Hartley Ⅰ, Elkhoury FF, Shin JH, et al. (2013) Long-Lasting Changes in DNA Methylation Following Short-Term Hypoxic Exposure in Primary Hippocampal Neuronal Cultures. PloS one 8:e77859.
    37. Hayatsu H, Wataya Y and Kai K. (1970a) Addition of sodium bisulfite to uracil and to cytosine. Journal of the American Chemical Society 92:724-726.
    38. Hayatsu H, Wataya Y, Kai K, et al. (1970b) Reaction of sodium bisulfite with uracil, cytosine, and their derivatives. Biochemistry 9:2858-2865.
    39. Hochachka P, Rupert J and Monge C. (1999) Adaptation and conservation of physiological systems in the evolution of human hypoxia tolerance. Comparative Biochemistry and Physiology Part A:Molecular & Integrative Physiology 124:1-17.
    40. Holden J, Stone C, Clark C, et al. (1995) Enhanced cardiac metabolism of plasma glucose in high-altitude natives:adaptation against chronic hypoxia. Journal of Applied Physiology 79: 222-228.
    41. Hudlicka O, Brown MD, Walter H, et al. (1995) Factors involved in capillary growth in the heart. Molecular and cellular biochemistry 147:57-68.
    42. Huss JM, Levy FH and Kelly DP. (2001) Hypoxia Inhibits the Peroxisome Proliferator-activated Receptor a/Retinoid X Receptor Gene Regulatory Pathway in Cardiac Myocytes A MECHANISM FOR O2-DEPENDENT MODULATION OF MITOCHONDRIAL FATTY ACID OXIDATION. Journal of biological chemistry 276: 27605-27612.
    43. Islam KN and Mendelson CR. (2006) Permissive effects of oxygen on cyclic AMP and interleukin-1 stimulation of surfactant protein A gene expression are mediated by epigenetic mechanisms. Mol Cell Biol 26:2901-2912.
    44. Kai K, Tsuruo T and Hayatsu H. (1974) The effect of bisulfite modification on the template activity of DNA for DNA polymerase I1. Nucleic acids research 1:889-900.
    45. Kasper LH, Boussouar F, Boyd K, et al. (2005) Two transactivation mechanisms cooperate for the bulk of HIF-1-responsive gene expression. The EMBO journal 24:3846-3858.
    46. Ko YG, Nishino K, Hattori N, et al. (2005) Stage-by-stage change in DNA methylation status of Dnmtl locus during mouse early development. JBiol Chem 280:9627-9634.
    47. Koslowski M, Luxemburger U, Tureci O, et al. (2011) Tumor-associated CpG demethylation augments hypoxia-induced effects by positive autoregulation of HIF-lalpha. Oncogene 30: 876-882.
    48. Krieg AJ, Rankin EB, Chan D, et al. (2010) Regulation of the histone demethylase JMJD1A by hypoxia-inducible factor 1α enhances hypoxic gene expression and tumor growth. Mol Cell Biol 30:344-353.
    49. Lagger G, O'Carroll D, Rembold M, et al. (2002) Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. The EMBO journal 21:2672-2681.
    50. Law A, Lai KP, Ip CK, et al. (2008) Epigenetic and HIF-1 regulation of stanniocalcin-2 expression in human cancer cells. Experimental cell research 314:1823-1830.
    51. Lee J-S, Smith E and Shilatifard A. (2010) The language of histone crosstalk. Cell 142: 682-685.
    52. Li HG, Ren YM, Guo SC, et al. (2009a) The protein level of hypoxia-inducible factor-1α is increased in the plateau pika (Ochotona curzoniae) inhabiting high altitudes. Journal of Experimental Zoology Part A:Ecological Genetics and Physiology 311:134-141.
    53. Li J, Gao F, Li N, et al. (2009b) An improved method for genome wide DNA methylation profiling correlated to transcription and genomic instability in two breast cancer cell lines. BMC genomics 10:223.
    54. Li M, Tian S, Jin L, et al. (2013) Genomic analyses identify distinct patterns of selection in domesticated pigs and Tibetan wild boars. Nat Genet 45:1431-1438.
    55. Lister R, O'Malley RC, Tonti-Filippini J, et al. (2008) Highly Integrated Single-Base Resolution Maps of the Epigenome in< i> Arabidopsis. Cell 133:523-536.
    56. Lister R, Pelizzola M, Dowen RH, et al. (2009a) Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462:315-322.
    57. Lister R, Pelizzola M, Dowen RH, et al. (2009b) Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462:315-322.
    58. Liu Q, Liu L, Zhao Y, et al. (2011) Hypoxia induces genomic DNA demethylation through the activation of HIF-lalpha and transcriptional upregulation of MAT2A in hepatoma cells. Mol Cancer Ther 10:1113-1123.
    59. Liu X-Z, Li S-L, Jing H, et al. (2001) Avian haemoglobins and structural basis of high affinity for oxygen:structure of bar-headed goose aquomet haemoglobin. Acta Crystallographica Section D:Biological Crystallography 57:775-783.
    60. Liu X, Gao Q, Li P, et al. (2013) UHRF1 targets DNMT1 for DNA methylation through cooperative binding of hemi-methylated DNA and methylated H3K9. Nature communications 4:1563.
    61. Liu Z, Zhou S, Liao L, et al. (2010) Jmjdla demethylase-regulated histone modification is essential for cAMP-response element modulator-regulated gene expression and spermatogenesis. JBiol Chem 285:2758-2770.
    62. Lu X and Kang Y. (2010) Hypoxia and hypoxia-inducible factors:master regulators of metastasis. Clinical Cancer Research 16:5928-5935.
    63. Lutfullah G, Ali SA and Abbasi A. (2004) Molecular mechanism of high altitude respiration: primary structure of a minor hemoglobin component from Tufted duck (< i> Aythya fuligula, Anseriformes). Biochemical and biophysical research communications 326: 123-130.
    64. Majmundar AJ, Wong WJ and Simon MC. (2010) Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell 40:294-309.
    65. Makino Y, Cao R, Svensson K, et al. (2001) Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature 414:550-554.
    66. Margot JB, Cardoso MC and Leonhardt H. (2001) Mammalian DNA methyltransferases show different subnuclear distributions. Journal of cellular biochemistry 83:373-379.
    67. Meissner A, Gnirke A, Bell GW, et al. (2005) Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic acids research 33: 5868-5877.
    68. Meissner A, Mikkelsen TS, Gu H, et al. (2008) Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454:766-770.
    69. Melvin A and Rocha S. (2012) Chromatin as an oxygen sensor and active player in the hypoxia response. Cellular signalling 24:35-43.
    70. Montgomery RL, Davis CA, Potthoff MJ, et al. (2007) Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility. Genes Dev 21: 1790-1802.
    71. Moore LG. (2001) Human genetic adaptation to high altitude. High altitude medicine& biology 2:257-279.
    72. Nanduri J, Makarenko V, Reddy VD, et al. (2012) Epigenetic regulation of hypoxic sensing disrupts cardiorespiratory homeostasis. Proceedings of the National Academy of Sciences 109: 2515-2520.
    73. Narravula S and Colgan SP. (2001a) Hypoxia-inducible Factor 1-Mediated Inhibition of Peroxisome Proliferator-Activated Receptor Expression During Hypoxia. The Journal of Immunology 166:7543-7548.
    74. Narravula S and Colgan SP. (2001b) Hypoxia-inducible factor 1-mediated inhibition of peroxisome proliferator-activated receptor a expression during hypoxia. The Journal of Immunology 166:7543-7548.
    75. Ng H-H and Adrian B. (1999) DNA methylation and chromatin modification. Current opinion in genetics& development 9:158-163.
    76. Oda M, Glass JL, Thompson RF, et al. (2009) High-resolution genome-wide cytosine methylation profiling with simultaneous copy number analysis and optimization for limited cell numbers. Nucleic acids research 37:3829-3839.
    77. Ogoshi K, Hashimoto S-i, Nakatani Y, et al. (2011) Genome-wide profiling of DNA methylation in human cancer cells. Genomics 98:280-287.
    78. Ohler U, Wang K, Narayanan M, et al. (2009) Meta-analysis of Inter-species Liver Co-expression Networks Elucidates Traits Associated with Common Human Diseases. PLoS Computational Biology 5:e1000616.
    79. Okano M, Bell DW, Haber DA, et al. (1999) DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99:247-257.
    80. Perez-Perri JI, Acevedo JM and Wappner P. (2011) Epigenetics:new questions on the response to hypoxia. International journal of molecular sciences 12:4705-4721.
    81. Philip S, Castro LF, da Fonseca RR, et al. (2012) Adaptive evolution of the Retinoid X receptor in vertebrates. Genomics 99:81-89.
    82. Raspaglio G, Filippetti F, Prislei S, et al. (2008) Hypoxia induces class Ⅲ beta-tubulin gene expression by HIF-1α binding to its 3'flanking region. Gene 409:100-108.
    83. Rocha S. (2007) Gene regulation under low oxygen:holding your breath for transcription. Trends in biochemical sciences 32:389-397.
    84. Rollins RA, Haghighi F, Edwards JR, et al. (2006) Large-scale structure of genomic methylation patterns. Genome Res 16:157-163.
    85. Rountree MR, Bachman KE and Baylin SB. (2000) DNMT1 binds HDAC2 and a new co-repressor, DMAP1, to form a complex at replication foci. Nat Genet 25:269-277.
    86. Rupert J. (2010) Will blood tell? Three recent articles demonstrate genetic selection in Tibetans. High altitude medicine & biology 11:307-308.
    87. Rupert JL and Koehle MS. (2006) Evidence for a genetic basis for altitude-related illness. High altitude medicine & biology 7:150-167.
    88. Saxonov S, Berg P and Brutlag DL. (2006) A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci U S A 103:1412-1417.
    89. Schodel J, Oikonomopoulos S, Ragoussis J, et al. (2011) High-resolution genome-wide mapping of HIF-binding sites by ChIP-seq. Blood 117:e207-e217.
    90. Scott GR, Schulte PM, Egginton S, et al. (2011) Molecular evolution of cytochrome c oxidase underlies high-altitude adaptation in the bar-headed goose. Molecular biology and evolution 28:351-363.
    91. Semenza GL. (2011) Hypoxia-inducible factor 1:regulator of mitochondrial metabolism and mediator of ischemic preconditioning. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 1813:1263-1268.
    92. Semenza GL. (2012a) Hypoxia-inducible factors in physiology and medicine. Cell 148: 399-408.
    93. Semenza GL. (2012b) Hypoxia-inducible factors:mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci 33:207-214.
    94. Serre D, Lee BH and Ting AH. (2010) MBD-isolated Genome Sequencing provides a high-throughput and comprehensive survey of DNA methylation in the human genome. Nucleic acids research 38:391-399.
    95. Service R. (1998) Microchip arrays put DNA on the spot. Science (New York, NY) 282:396.
    96. Shahrzad S, Bertrand K, Minhas K, et al. (2007) Induction of DNA hypomethylation by tumor hypoxia. Epigenetics 2:119-125.
    97. Shell SS, Prestwich EG, Baek S-H, et al. (2013) DNA methylation impacts gene expression and ensures hypoxic survival of Mycobacterium tuberculosis. PLoSpathogens 9:el003419.
    98. Simonson TS, Yang Y, Huff CD, et al. (2010) Genetic evidence for high-altitude adaptation in Tibet. Science 329:72-75.
    99. Skowronski K, Dubey S, Rodenhiser D, et al. (2010) Ischemia dysregulates DNA methyltransferases andp161NK4amethylation in human colorectal cancer cells. Epigenetics 5: 547-556.
    100. Storz JF, Sabatino SJ, Hoffmann FG, et al. (2007) The molecular basis of high-altitude adaptation in deer mice. PLoS Genetics 3:e45.
    101. Suzuki M, Jing Q, Lia D, et al. (2010) Method Optimized design and data analysis of tag-based cytosine methylation assays.
    102. Szanto A, Narkar V, Shen Q, et al. (2004) Retinoid X receptors:X-ploring their (patho)physiological functions. Cell Death and Differentiation 11:S126-S143.
    103.Takai D and Jones PA. (2002) Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc Natl Acad Sci USA 99:3740-3745.
    104. Tausendschon M, Dehne N and Briine B. (2011) Hypoxia causes epigenetic gene regulation in macrophages by attenuating Jumonji histone demethylase activity. Cytokine 53:256-262.
    105. Taylor CT and Colgan SP. (1999) Therapeutic targets for hypoxia-elicited pathways. Pharmaceutical research 16:1498-1505.
    106. Uniacke J, Holterman CE, Lachance G, et al. (2012) An oxygen-regulated switch in the protein synthesis machinery. Nature 486:126-129.
    107. Wang D, Li H, Li Y, et al. (2006) Hypoxia-inducible factor lα cDNA cloning and its mRNA and protein tissue specific expression in domestic yak (< i> Bos grunniens) from Qinghai-Tibetan plateau. Biochemical and biophysical research communications 348: 310-319.
    108. Wang F, Zhang R, Wu X, et al. (2010) Roles of coactivators in hypoxic induction of the erythropoietin gene. PloS one 5:e10002.
    109. Wang Q, Fujii H and Knipp GT. (2002) Expression of PPAR and RXR Isoforms in the Developing Rat and Human Term Placentas. Placenta 23:661-671.
    110. Watson JA, Watson CJ, McCann A, et al. (2010) Epigenetics, the epicenter of the hypoxic response. Epigenetics 5:293-296.
    111. Watson JA, Watson CJ, McCrohan A-M, et al. (2009) Generation of an epigenetic signature by chronic hypoxia in prostate cells. Human molecular genetics 18:3594-3604.
    112. Weber M, Davies JJ, Wittig D, et al. (2005) Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nat Genet 37:853-862.
    113. Weber RE, Jessen T-H, Malte H, et al. (1993) Mutant hemoglobins (alpha 119-Ala and beta 55-Ser):functions related to high-altitude respiration in geese. Journal of Applied Physiology 75:2646-2655.
    114. Wenger RH. (2000) Mammalian oxygen sensing, signalling and gene regulation. Journal of Experimental Biology 203:1253-1263.
    115.Xia X, Lemieux ME, Li W, et al. (2009) Integrative analysis of HIF binding and transactivation reveals its role in maintaining histone methylation homeostasis. Proceedings of the National Academy of Sciences 106:4260-4265.
    116. Xie S, Wang Z, Okano M, et al. (1999) Cloning, expression and chromosome locations of the human< i> DNMT3 gene family. Gene 236:87-95.
    117.Yoder JA, Soman NS, Verdine GL, et al. (1997) DNA (cytosine-5)-methyltransferases in mouse cells and tissues. Studies with a mechanism-based probe. Journal of molecular biology 270:385-395.
    118. Zhou X, Sun H, Chen H, et al. (2010) Hypoxia induces trimethylated H3 lysine 4 by inhibition of JARID1A demethylase. Cancer research 70:4214-4221.
    119.李庆芬.(1991)人与动物的呼吸系统对高海拔低氧的适应.生物学通报10:19-21.
    120.周兆年.(2012)低氧生理学研究的回顾.生理群学进展1:1-4.
    121.刘国富,温得启,胡晓梅.(1985)高原鼠兔和高原鼢鼠乳酸脱氢酶同工酶的初步研究.兽类学报3:223-228.
    122.孙希武,叶益新,邓希贤.(1994)低氧适应对缺氧性心功能损伤的保护作用及其机制探讨.中国应用生理学杂志3:205-208.
    123.强巴央宗,谢庄,商鹏等(2009)藏鸡血管内皮生长因子基因表达与低氧适应.畜牧兽医学报7:1101-1105.
    124.马兰,格日力.(2007)高原鼠兔低氧适应分子机制的研究进展.生理科学进展2:143-146.
    125.黄庆愿,高钰琪,史景泉等(2001)低氧习服大鼠骨骼肌毛细血管密度和血流供应的变化 特点.中国应用生理学杂志3:220-223.
    126.龙超良,周智,尹昭云等(1999)急、慢性缺氧对大鼠心功能的影响.航天医学与医学工程4:35-37.

© 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