#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Lipid-Induced Epigenomic Changes in Human Macrophages Identify a Coronary Artery Disease-Associated Variant that Regulates Expression through Altered C/EBP-Beta Binding


Coronary artery disease is a complex disease where over 40 genomic loci contributing to genetic risk have been identified. However, identifying the precise variants, genomic elements and genes that mediate this risk at each locus has proved challenging. We hypothesized that some genetic risk variants may influence a key step in development of coronary artery disease, which occurs when macrophages encounter environmentally-derived lipid. These cells take up lipid and accumulate in atherosclerotic plaques in the walls of blood vessels where they contribute to the inflammatory atherosclerotic disease process. Therefore, we studied the effects of this lipid exposure on the genomic activity of these cells. Environmental lipid exposure triggered changes in transcriptional regulation and gene expression. Variants at coronary artery disease risk loci were enriched for genomic regions altered by lipid exposure. We studied one such risk variant rs72664324 in detail and found that it altered binding of the C/EBP-beta transcription factor and altered expression of the PPAP2B gene. PPAP2B encodes an enzyme that degrades pro-inflammatory substances. Our study demonstrates a hitherto unknown genetic mechanism underlying atherosclerotic heart disease and demonstrates the value of studying changes in transcriptional regulation in key disease processes involving environmental influences.


Vyšlo v časopise: Lipid-Induced Epigenomic Changes in Human Macrophages Identify a Coronary Artery Disease-Associated Variant that Regulates Expression through Altered C/EBP-Beta Binding. PLoS Genet 11(4): e32767. doi:10.1371/journal.pgen.1005061
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005061

Souhrn

Coronary artery disease is a complex disease where over 40 genomic loci contributing to genetic risk have been identified. However, identifying the precise variants, genomic elements and genes that mediate this risk at each locus has proved challenging. We hypothesized that some genetic risk variants may influence a key step in development of coronary artery disease, which occurs when macrophages encounter environmentally-derived lipid. These cells take up lipid and accumulate in atherosclerotic plaques in the walls of blood vessels where they contribute to the inflammatory atherosclerotic disease process. Therefore, we studied the effects of this lipid exposure on the genomic activity of these cells. Environmental lipid exposure triggered changes in transcriptional regulation and gene expression. Variants at coronary artery disease risk loci were enriched for genomic regions altered by lipid exposure. We studied one such risk variant rs72664324 in detail and found that it altered binding of the C/EBP-beta transcription factor and altered expression of the PPAP2B gene. PPAP2B encodes an enzyme that degrades pro-inflammatory substances. Our study demonstrates a hitherto unknown genetic mechanism underlying atherosclerotic heart disease and demonstrates the value of studying changes in transcriptional regulation in key disease processes involving environmental influences.


Zdroje

1. World Health Organization. World health statistics. Geneva, Switzerland: World Health Organization.

2. Glass CK, Witztum JL (2001) Atherosclerosis. the road ahead. Cell 104: 503–516. 11239408

3. Hansson GK, Hermansson A (2011) The immune system in atherosclerosis. Nature immunology 12: 204–212. doi: 10.1038/ni.2001 21321594

4. Stamler J, Wentworth D, Neaton JD (1986) Is relationship between serum cholesterol and risk of premature death from coronary heart disease continuous and graded? Findings in 356,222 primary screenees of the Multiple Risk Factor Intervention Trial (MRFIT). JAMA 256: 2823–2828. 3773199

5. Pekkanen J, Linn S, Heiss G, Suchindran CM, Leon A, et al. (1990) Ten-year mortality from cardiovascular disease in relation to cholesterol level among men with and without preexisting cardiovascular disease. N Engl J Med 322: 1700–1707. 2342536

6. Scandinavian Simvastatin Survival Study G (1994) Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). The Lancet 344: 1383–1389. 7968073

7. Sayols-Baixeras S, Lluis-Ganella C, Lucas G, Elosua R (2014) Pathogenesis of coronary artery disease: focus on genetic risk factors and identification of genetic variants. Appl Clin Genet 7: 15–32. doi: 10.2147/TACG.S35301 24520200

8. Consortium CAD, Deloukas P, Kanoni S, Willenborg C, Farrall M, et al. (2013) Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet 45: 25–33. doi: 10.1038/ng.2480 23202125

9. Musunuru K, Strong A, Frank-Kamenetsky M, Lee NE, Ahfeldt T, et al. (2010) From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature 466: 714–719. doi: 10.1038/nature09266 20686566

10. Harismendy O, Notani D, Song X, Rahim NG, Tanasa B, et al. (2011) 9p21 DNA variants associated with coronary artery disease impair interferon-gamma signalling response. Nature 470: 264–268. doi: 10.1038/nature09753 21307941

11. Miller CL, Haas U, Diaz R, Leeper NJ, Kundu RK, et al. (2014) Coronary heart disease-associated variation in TCF21 disrupts a miR-224 binding site and miRNA-mediated regulation. PLoS Genet 10: e1004263. doi: 10.1371/journal.pgen.1004263 24676100

12. Moore KJ, Sheedy FJ, Fisher EA (2013) Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol 13: 709–721. doi: 10.1038/nri3520 23995626

13. Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, et al. (1995) Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci U S A 92: 8264–8268. 7667279

14. Ball RY, Stowers EC, Burton JH, Cary NR, Skepper JN, et al. (1995) Evidence that the death of macrophage foam cells contributes to the lipid core of atheroma. Atherosclerosis 114: 45–54. 7605375

15. Silvestre-Roig C, de Winther MP, Weber C, Daemen MJ, Lutgens E, et al. (2014) Atherosclerotic plaque destabilization: mechanisms, models, and therapeutic strategies. Circ Res 114: 214–226. doi: 10.1161/CIRCRESAHA.114.302355 24385514

16. Hopkins PN (2013) Molecular biology of atherosclerosis. Physiol Rev 93: 1317–1542. doi: 10.1152/physrev.00004.2012 23899566

17. Kobayashi E, Suzuki T, Yamamoto M (2013) Roles nrf2 plays in myeloid cells and related disorders. Oxid Med Cell Longev 2013: 529219. doi: 10.1155/2013/529219 23819012

18. Consortium EP, Bernstein BE, Birney E, Dunham I, Green ED, et al. (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489: 57–74. doi: 10.1038/nature11247 22955616

19. Song L, Zhang Z, Grasfeder LL, Boyle AP, Giresi PG, et al. (2011) Open chromatin defined by DNaseI and FAIRE identifies regulatory elements that shape cell-type identity. Genome Res 21: 1757–1767. doi: 10.1101/gr.121541.111 21750106

20. John S, Sabo PJ, Thurman RE, Sung MH, Biddie SC, et al. (2011) Chromatin accessibility pre-determines glucocorticoid receptor binding patterns. Nat Genet 43: 264–268. doi: 10.1038/ng.759 21258342

21. Biddie SC, John S, Sabo PJ, Thurman RE, Johnson TA, et al. (2011) Transcription factor AP1 potentiates chromatin accessibility and glucocorticoid receptor binding. Mol Cell 43: 145–155. doi: 10.1016/j.molcel.2011.06.016 21726817

22. Simon JM, Giresi PG, Davis IJ, Lieb JD (2012) Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat Protoc 7: 256–267. doi: 10.1038/nprot.2011.444 22262007

23. Gaulton KJ, Nammo T, Pasquali L, Simon JM, Giresi PG, et al. (2010) A map of open chromatin in human pancreatic islets. Nat Genet 42: 255–259. doi: 10.1038/ng.530 20118932

24. Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, et al. (2010) Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A 107: 21931–21936. doi: 10.1073/pnas.1016071107 21106759

25. Pasquali L, Gaulton KJ, Rodriguez-Segui SA, Mularoni L, Miguel-Escalada I, et al. (2014) Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat Genet.

26. Ward LD, Kellis M (2012) Interpreting noncoding genetic variation in complex traits and human disease. Nat Biotechnol 30: 1095–1106. doi: 10.1038/nbt.2422 23138309

27. Trynka G, Sandor C, Han B, Xu H, Stranger BE, et al. (2013) Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat Genet 45: 124–130. doi: 10.1038/ng.2504 23263488

28. Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, et al. (2012) Systematic Localization of Common Disease-Associated Variation in Regulatory DNA. Science 337: 1190–1195. doi: 10.1126/science.1222794 22955828

29. Buechler C, Ritter M, Duong CQ, Orso E, Kapinsky M, et al. (2001) Adipophilin is a sensitive marker for lipid loading in human blood monocytes. Biochim Biophys Acta 1532: 97–104. 11420178

30. Nord AS, Blow MJ, Attanasio C, Akiyama JA, Holt A, et al. (2013) Rapid and pervasive changes in genome-wide enhancer usage during mammalian development. Cell 155: 1521–1531. doi: 10.1016/j.cell.2013.11.033 24360275

31. Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-Andre V, et al. (2013) Super-enhancers in the control of cell identity and disease. Cell 155: 934–947. doi: 10.1016/j.cell.2013.09.053 24119843

32. Nagy ZS, Czimmerer Z, Nagy L (2013) Nuclear receptor mediated mechanisms of macrophage cholesterol metabolism. Mol Cell Endocrinol 368: 85–98. doi: 10.1016/j.mce.2012.04.003 22546548

33. Huber R, Pietsch D, Panterodt T, Brand K (2012) Regulation of C/EBPbeta and resulting functions in cells of the monocytic lineage. Cell Signal 24: 1287–1296. doi: 10.1016/j.cellsig.2012.02.007 22374303

34. Genomes Project C, Abecasis GR, Auton A, Brooks LD, DePristo MA, et al. (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491: 56–65. doi: 10.1038/nature11632 23128226

35. Collaboration IRGCERF, Sarwar N, Butterworth AS, Freitag DF, Gregson J, et al. (2012) Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet 379: 1205–1213. doi: 10.1016/S0140-6736(11)61931-4 22421339

36. Ferreira RC, Freitag DF, Cutler AJ, Howson JM, Rainbow DB, et al. (2013) Functional IL6R 358Ala allele impairs classical IL-6 receptor signaling and influences risk of diverse inflammatory diseases. PLoS Genet 9: e1003444. doi: 10.1371/journal.pgen.1003444 23593036

37. Sciorra VA, Morris AJ (1999) Sequential actions of phospholipase D and phosphatidic acid phosphohydrolase 2b generate diglyceride in mammalian cells. Mol Biol Cell 10: 3863–3876. 10564277

38. Long JS, Pyne NJ, Pyne S (2008) Lipid phosphate phosphatases form homo- and hetero-oligomers: catalytic competency, subcellular distribution and function. The Biochemical journal 411: 371–377. doi: 10.1042/BJ20071607 18215144

39. Panchatcharam M, Miriyala S, Salous A, Wheeler J, Dong A, et al. (2013) Lipid phosphate phosphatase 3 negatively regulates smooth muscle cell phenotypic modulation to limit intimal hyperplasia. Arterioscler Thromb Vasc Biol 33: 52–59. doi: 10.1161/ATVBAHA.112.300527 23104851

40. Getz GS, Reardon CA (2012) Animal models of atherosclerosis. Arterioscler Thromb Vasc Biol 32: 1104–1115. doi: 10.1161/ATVBAHA.111.237693 22383700

41. Steinberg D, Parthasarathy S, Carew TE, Khoo JC, Witztum JL (1989) Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity. N Engl J Med 320: 915–924. 2648148

42. Moore KJ, Tabas I (2011) Macrophages in the pathogenesis of atherosclerosis. Cell 145: 341–355. doi: 10.1016/j.cell.2011.04.005 21529710

43. Magnani L, Ballantyne EB, Zhang X, Lupien M (2011) PBX1 genomic pioneer function drives ERalpha signaling underlying progression in breast cancer. PLoS Genet 7: e1002368. doi: 10.1371/journal.pgen.1002368 22125492

44. Nammo T, Rodriguez-Segui SA, Ferrer J (2011) Mapping open chromatin with formaldehyde-assisted isolation of regulatory elements. Methods in molecular biology 791: 287–296. doi: 10.1007/978-1-61779-316-5_21 21913087

45. Paul DS, Albers CA, Rendon A, Voss K, Stephens J, et al. (2013) Maps of open chromatin highlight cell type-restricted patterns of regulatory sequence variation at hematological trait loci. Genome Res 23: 1130–1141. doi: 10.1101/gr.155127.113 23570689

46. Waki H, Nakamura M, Yamauchi T, Wakabayashi K, Yu J, et al. (2011) Global mapping of cell type-specific open chromatin by FAIRE-seq reveals the regulatory role of the NFI family in adipocyte differentiation. PLoS Genet 7: e1002311. doi: 10.1371/journal.pgen.1002311 22028663

47. Zhang B, Day DS, Ho JW, Song L, Cao J, et al. (2013) A dynamic H3K27ac signature identifies VEGFA-stimulated endothelial enhancers and requires EP300 activity. Genome Res 23: 917–927. doi: 10.1101/gr.149674.112 23547170

48. Shlyueva D, Stampfel G, Stark A (2014) Transcriptional enhancers: from properties to genome-wide predictions. Nat Rev Genet 15: 272–286. doi: 10.1038/nrg3682 24614317

49. Siersbaek R, Nielsen R, John S, Sung MH, Baek S, et al. (2011) Extensive chromatin remodelling and establishment of transcription factor 'hotspots' during early adipogenesis. EMBO J 30: 1459–1472. doi: 10.1038/emboj.2011.65 21427703

50. Rhie SK, Hazelett DJ, Coetzee SG, Yan C, Noushmehr H, et al. (2014) Nucleosome positioning and histone modifications define relationships between regulatory elements and nearby gene expression in breast epithelial cells. BMC Genomics 15: 331. doi: 10.1186/1471-2164-15-331 24885402

51. Nagy L, Szanto A, Szatmari I, Szeles L (2012) Nuclear hormone receptors enable macrophages and dendritic cells to sense their lipid environment and shape their immune response. Physiol Rev 92: 739–789. doi: 10.1152/physrev.00004.2011 22535896

52. Gupte AA, Lyon CJ, Hsueh WA (2013) Nuclear factor (erythroid-derived 2)-like-2 factor (Nrf2), a key regulator of the antioxidant response to protect against atherosclerosis and nonalcoholic steatohepatitis. Curr Diab Rep 13: 362–371. doi: 10.1007/s11892-013-0372-1 23475581

53. Rahman SM, Janssen RC, Choudhury M, Baquero KC, Aikens RM, et al. (2012) CCAAT/enhancer-binding protein beta (C/EBPbeta) expression regulates dietary-induced inflammation in macrophages and adipose tissue in mice. J Biol Chem 287: 34349–34360. doi: 10.1074/jbc.M112.410613 22902781

54. Trynka G, Sandor C, Han B, Xu H, Stranger BE, et al. (2012) Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat Genet.

55. Kai M, Wada I, Imai S, Sakane F, Kanoh H (1997) Cloning and characterization of two human isozymes of Mg2+-independent phosphatidic acid phosphatase. The Journal of biological chemistry 272: 24572–24578. 9305923

56. Ren H, Panchatcharam M, Mueller P, Escalante-Alcalde D, Morris AJ, et al. (2013) Lipid phosphate phosphatase (LPP3) and vascular development. Biochim Biophys Acta 1831: 126–132. doi: 10.1016/j.bbalip.2012.07.012 22835522

57. Escalante-Alcalde D, Sanchez-Sanchez R, Stewart CL (2007) Generation of a conditional Ppap2b/Lpp3 null allele. Genesis 45: 465–469. 17610274

58. Knowlden S, Georas SN (2014) The autotaxin-LPA axis emerges as a novel regulator of lymphocyte homing and inflammation. J Immunol 192: 851–857. doi: 10.4049/jimmunol.1302831 24443508

59. Yung YC, Stoddard NC, Chun J (2014) LPA Receptor Signaling: Pharmacology, Physiology, and Pathophysiology. J Lipid Res.

60. Bot M, Bot I, Lopez-Vales R, van de Lest CH, Saulnier-Blache JS, et al. (2010) Atherosclerotic lesion progression changes lysophosphatidic acid homeostasis to favor its accumulation. Am J Pathol 176: 3073–3084. doi: 10.2353/ajpath.2010.090009 20431029

61. Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, et al. (1999) Lysophosphatidic acid mediates the rapid activation of platelets and endothelial cells by mildly oxidized low density lipoprotein and accumulates in human atherosclerotic lesions. Proc Natl Acad Sci U S A 96: 6931–6936. 10359816

62. Smyth SS, Mueller P, Yang F, Brandon JA, Morris AJ (2014) Arguing the case for the autotaxin-lysophosphatidic acid-lipid phosphate phosphatase 3-signaling nexus in the development and complications of atherosclerosis. Arterioscler Thromb Vasc Biol 34: 479–486. doi: 10.1161/ATVBAHA.113.302737 24482375

63. Stegemann C, Drozdov I, Shalhoub J, Humphries J, Ladroue C, et al. (2011) Comparative lipidomics profiling of human atherosclerotic plaques. Circ Cardiovasc Genet 4: 232–242. doi: 10.1161/CIRCGENETICS.110.959098 21511877

64. Schober A, Siess W (2012) Lysophosphatidic acid in atherosclerotic diseases. Br J Pharmacol 167: 465–482. doi: 10.1111/j.1476-5381.2012.02021.x 22568609

65. Mendelson K, Evans T, Hla T (2014) Sphingosine 1-phosphate signalling. Development 141: 5–9. doi: 10.1242/dev.094805 24346695

66. Skoura A, Michaud J, Im DS, Thangada S, Xiong Y, et al. (2011) Sphingosine-1-phosphate receptor-2 function in myeloid cells regulates vascular inflammation and atherosclerosis. Arterioscler Thromb Vasc Biol 31: 81–85. doi: 10.1161/ATVBAHA.110.213496 20947824

67. Schunkert H, Konig IR, Kathiresan S, Reilly MP, Assimes TL, et al. (2011) Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nature genetics 43: 333–338. doi: 10.1038/ng.784 21378990

68. Coronary Artery Disease Genetics C (2011) A genome-wide association study in Europeans and South Asians identifies five new loci for coronary artery disease. Nat Genet 43: 339–344. doi: 10.1038/ng.782 21378988

69. Andersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, et al. (2014) An atlas of active enhancers across human cell types and tissues. Nature 507: 455–461. doi: 10.1038/nature12787 24670763

70. Havel RJ, Eder HA, Bragdon JH (1955) The distribution and chemical composition of ultracentrifugally separated lipoproteins in human serum. The Journal of clinical investigation 34: 1345–1353. 13252080

71. Lunter G, Goodson M (2011) Stampy: a statistical algorithm for sensitive and fast mapping of Illumina sequence reads. Genome Res 21: 936–939. doi: 10.1101/gr.111120.110 20980556

72. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, et al. (2009) The Sequence Alignment/Map format and SAMtools. Bioinformatics 25: 2078–2079. doi: 10.1093/bioinformatics/btp352 19505943

73. Boyle AP, Guinney J, Crawford GE, Furey TS (2008) F-Seq: a feature density estimator for high-throughput sequence tags. Bioinformatics 24: 2537–2538. doi: 10.1093/bioinformatics/btn480 18784119

74. McCarthy MT, O'Callaghan CA (2014) PeaKDEck: a kernel density estimator-based peak calling program for DNaseI-seq data. Bioinformatics 30: 1302–1304. doi: 10.1093/bioinformatics/btt774 24407222

75. Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, et al. (2008) Model-based analysis of ChIP-Seq (MACS). Genome Biol 9: R137. doi: 10.1186/gb-2008-9-9-r137 18798982

76. Shen L, Shao NY, Liu X, Maze I, Feng J, et al. (2013) diffReps: detecting differential chromatin modification sites from ChIP-seq data with biological replicates. PLoS One 8: e65598. doi: 10.1371/journal.pone.0065598 23762400

77. Pham T-H, Benner C, Lichtinger M, Schwarzfischer L, Hu Y, et al. (2012) Dynamic epigenetic enhancer signatures reveal key transcription factors associated with monocytic differentiation states. Blood 119: e161–e171. doi: 10.1182/blood-2012-01-402453 22550342

78. Shin H, Liu T, Manrai AK, Liu XS (2009) CEAS: cis-regulatory element annotation system. Bioinformatics 25: 2605–2606. doi: 10.1093/bioinformatics/btp479 19689956

79. McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, et al. (2010) GREAT improves functional interpretation of cis-regulatory regions. Nat Biotechnol 28: 495–501. doi: 10.1038/nbt.1630 20436461

80. Liu T, Ortiz JA, Taing L, Meyer CA, Lee B, et al. (2011) Cistrome: an integrative platform for transcriptional regulation studies. Genome Biol 12: R83. doi: 10.1186/gb-2011-12-8-r83 21859476

81. Ward LD, Kellis M (2012) HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res 40: D930–934. doi: 10.1093/nar/gkr917 22064851

82. Samani NJ, Erdmann J, Hall AS, Hengstenberg C, Mangino M, et al. (2007) Genomewide association analysis of coronary artery disease. N Engl J Med 357: 443–453. 17634449

83. Myocardial Infarction Genetics C, Kathiresan S, Voight BF, Purcell S, Musunuru K, et al. (2009) Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat Genet 41: 334–341. doi: 10.1038/ng.327 19198609

84. Erdmann J, Willenborg C, Nahrstaedt J, Preuss M, Konig IR, et al. (2011) Genome-wide association study identifies a new locus for coronary artery disease on chromosome 10p11.23. Eur Heart J 32: 158–168. doi: 10.1093/eurheartj/ehq405 21088011

85. Wild PS, Zeller T, Schillert A, Szymczak S, Sinning CR, et al. (2011) A genome-wide association study identifies LIPA as a susceptibility gene for coronary artery disease. Circ Cardiovasc Genet 4: 403–412. doi: 10.1161/CIRCGENETICS.110.958728 21606135

86. O'Donnell CJ, Kavousi M, Smith AV, Kardia SL, Feitosa MF, et al. (2011) Genome-Wide Association Study for Coronary Artery Calcification With Follow-Up in Myocardial Infarction. Circulation 124: 2855–2864. doi: 10.1161/CIRCULATIONAHA.110.974899 22144573

87. Du P, Kibbe WA, Lin SM (2008) lumi: a pipeline for processing Illumina microarray. Bioinformatics 24: 1547–1548. doi: 10.1093/bioinformatics/btn224 18467348

88. Lin D, Lavender H, Soilleux EJ, O'Callaghan CA (2012) NF-kappaB regulates MICA gene transcription in endothelial cell through a genetically inhibitable control site. J Biol Chem 287: 4299–4310. doi: 10.1074/jbc.M111.282152 22170063

89. Escalante-Alcalde D, Hernandez L, Le Stunff H, Maeda R, Lee HS, et al. (2003) The lipid phosphatase LPP3 regulates extra-embryonic vasculogenesis and axis patterning. Development 130: 4623–4637. 12925589

90. Lopez-Juarez A, Morales-Lazaro S, Sanchez-Sanchez R, Sunkara M, Lomeli H, et al. (2011) Expression of LPP3 in Bergmann glia is required for proper cerebellar sphingosine-1-phosphate metabolism/signaling and development. Glia 59: 577–589. doi: 10.1002/glia.21126 21319224

91. Breart B, Ramos-Perez WD, Mendoza A, Salous AK, Gobert M, et al. (2011) Lipid phosphate phosphatase 3 enables efficient thymic egress. The Journal of experimental medicine 208: 1267–1278. doi: 10.1084/jem.20102551 21576386

92. Salous AK, Panchatcharam M, Sunkara M, Mueller P, Dong A, et al. (2013) Mechanism of rapid elimination of lysophosphatidic acid and related lipids from the circulation of mice. J Lipid Res 54: 2775–2784. doi: 10.1194/jlr.M039685 23948545

93. Federico L, Ren H, Mueller PA, Wu T, Liu S, et al. (2012) Autotaxin and its product lysophosphatidic acid suppress brown adipose differentiation and promote diet-induced obesity in mice. Mol Endocrinol 26: 786–797. doi: 10.1210/me.2011-1229 22474126

94. Dillen L, Cools W, Vereyken L, Lorreyne W, Huybrechts T, et al. (2012) Comparison of triple quadrupole and high-resolution TOF-MS for quantification of peptides. Bioanalysis 4: 565–579. doi: 10.4155/bio.12.3 22409554

95. Humtsoe JO, Liu M, Malik AB, Wary KK (2010) Lipid phosphate phosphatase 3 stabilization of beta-catenin induces endothelial cell migration and formation of branching point structures. Molecular and cellular biology 30: 1593–1606. doi: 10.1128/MCB.00038-09 20123964

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 4
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#