#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome-Wide Associations between Genetic and Epigenetic Variation Influence mRNA Expression and Insulin Secretion in Human Pancreatic Islets


Inter-individual variation in genetics and epigenetics affects biological processes and disease susceptibility. However, most studies have investigated genetic and epigenetic mechanisms independently and to uncover novel mechanisms affecting disease susceptibility there is a highlighted need to study interactions between these factors on a genome-wide scale. To identify novel loci affecting islet function and potentially diabetes, we performed the first genome-wide methylation quantitative trait locus (mQTL) analysis in human pancreatic islets including DNA methylation of 468,787 CpG sites located throughout the genome. Our results showed that DNA methylation of 11,735 CpGs in 4,504 unique genes is regulated by genetic factors located in cis (67,438 SNP-CpG pairs). Furthermore, significant mQTLs cover previously reported diabetes loci including KCNJ11, INS, HLA, PDX1 and GRB10. We also found mQTLs associated with gene expression and insulin secretion in human islets. By performing causality inference tests (CIT), we identified CpGs where DNA methylation potentially mediates the genetic impact on gene expression and insulin secretion. Our functional follow-up experiments further demonstrated that identified mQTLs/genes (GPX7, GSTT1 and SNX19) directly affect pancreatic β-cell function. Together, our study provides a detailed map of genome-wide associations between genetic and epigenetic variation, which affect gene expression and insulin secretion in human pancreatic islets.


Vyšlo v časopise: Genome-Wide Associations between Genetic and Epigenetic Variation Influence mRNA Expression and Insulin Secretion in Human Pancreatic Islets. PLoS Genet 10(11): e32767. doi:10.1371/journal.pgen.1004735
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004735

Souhrn

Inter-individual variation in genetics and epigenetics affects biological processes and disease susceptibility. However, most studies have investigated genetic and epigenetic mechanisms independently and to uncover novel mechanisms affecting disease susceptibility there is a highlighted need to study interactions between these factors on a genome-wide scale. To identify novel loci affecting islet function and potentially diabetes, we performed the first genome-wide methylation quantitative trait locus (mQTL) analysis in human pancreatic islets including DNA methylation of 468,787 CpG sites located throughout the genome. Our results showed that DNA methylation of 11,735 CpGs in 4,504 unique genes is regulated by genetic factors located in cis (67,438 SNP-CpG pairs). Furthermore, significant mQTLs cover previously reported diabetes loci including KCNJ11, INS, HLA, PDX1 and GRB10. We also found mQTLs associated with gene expression and insulin secretion in human islets. By performing causality inference tests (CIT), we identified CpGs where DNA methylation potentially mediates the genetic impact on gene expression and insulin secretion. Our functional follow-up experiments further demonstrated that identified mQTLs/genes (GPX7, GSTT1 and SNX19) directly affect pancreatic β-cell function. Together, our study provides a detailed map of genome-wide associations between genetic and epigenetic variation, which affect gene expression and insulin secretion in human pancreatic islets.


Zdroje

1. BirdA (2002) DNA methylation patterns and epigenetic memory. Genes Dev 16: 6–21.

2. ListerR, PelizzolaM, DowenRH, HawkinsRD, HonG, et al. (2009) Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462: 315–322.

3. RobertsonKD, WolffeAP (2000) DNA methylation in health and disease. Nat Rev Genet 1: 11–19.

4. ChongS, WhitelawE (2004) Epigenetic germline inheritance. Curr Opin Genet Dev 14: 692–696.

5. AnwayMD, CuppAS, UzumcuM, SkinnerMK (2005) Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308: 1466–1469.

6. KaminskyZA, TangT, WangSC, PtakC, OhGH, et al. (2009) DNA methylation profiles in monozygotic and dizygotic twins. Nat Genet 41: 240–245.

7. OllikainenM, SmithKR, JooEJ, NgHK, AndronikosR, et al. (2010) DNA methylation analysis of multiple tissues from newborn twins reveals both genetic and intrauterine components to variation in the human neonatal epigenome. Hum Mol Genet 19: 4176–4188.

8. ZhangD, ChengL, BadnerJA, ChenC, ChenQ, et al. (2010) Genetic control of individual differences in gene-specific methylation in human brain. Am J Hum Genet 86: 411–419.

9. GibbsJR, van der BrugMP, HernandezDG, TraynorBJ, NallsMA, et al. (2010) Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain. PLoS Genet 6: e1000952.

10. NumataS, YeT, HydeTM, Guitart-NavarroX, TaoR, et al. (2012) DNA methylation signatures in development and aging of the human prefrontal cortex. Am J Hum Genet 90: 260–272.

11. DrongAW, NicholsonG, HedmanAK, MeduriE, GrundbergE, et al. (2013) The presence of methylation quantitative trait loci indicates a direct genetic influence on the level of DNA methylation in adipose tissue. PLoS One 8: e55923.

12. GrundbergE, MeduriE, SandlingJK, HedmanAK, KeildsonS, et al. (2013) Global analysis of DNA methylation variation in adipose tissue from twins reveals links to disease-associated variants in distal regulatory elements. Am J Hum Genet 93: 876–890.

13. Gutierrez-ArcelusM, LappalainenT, MontgomerySB, BuilA, OngenH, et al. (2013) Passive and active DNA methylation and the interplay with genetic variation in gene regulation. eLife 2: e00523.

14. WagnerJR, BuscheS, GeB, KwanT, PastinenT, et al. (2014) The relationship between DNA methylation, genetic and expression inter-individual variation in untransformed human fibroblasts. Genome Biol 15: R37.

15. BarrettJC, ClaytonDG, ConcannonP, AkolkarB, CooperJD, et al. (2009) Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat Genet 41: 703–707.

16. BradfieldJP, QuHQ, WangK, ZhangH, SleimanPM, et al. (2011) A genome-wide meta-analysis of six type 1 diabetes cohorts identifies multiple associated loci. PLoS Genet 7: e1002293.

17. BurrenOS, AdlemEC, AchuthanP, ChristensenM, CoulsonRM, et al. (2011) T1DBase: update 2011, organization and presentation of large-scale data sets for type 1 diabetes research. Nucleic Acids Res 39: D997–1001.

18. FungEY, SmythDJ, HowsonJM, CooperJD, WalkerNM, et al. (2009) Analysis of 17 autoimmune disease-associated variants in type 1 diabetes identifies 6q23/TNFAIP3 as a susceptibility locus. Genes Immun 10: 188–191.

19. GrantSF, QuHQ, BradfieldJP, MarchandL, KimCE, et al. (2009) Follow-up analysis of genome-wide association data identifies novel loci for type 1 diabetes. Diabetes 58: 290–295.

20. GroopL, PociotF (2013) Genetics of diabetes - Are we missing the genes or the disease? Mol Cell Endocrinol 382: 726–739.

21. HakonarsonH, GrantSF, BradfieldJP, MarchandL, KimCE, et al. (2007) A genome-wide association study identifies KIAA0350 as a type 1 diabetes gene. Nature 448: 591–594.

22. LoweCE, CooperJD, BruskoT, WalkerNM, SmythDJ, et al. (2007) Large-scale genetic fine mapping and genotype-phenotype associations implicate polymorphism in the IL2RA region in type 1 diabetes. Nat Genet 39: 1074–1082.

23. PociotF, AkolkarB, ConcannonP, ErlichHA, JulierC, et al. (2010) Genetics of type 1 diabetes: what's next? Diabetes 59: 1561–1571.

24. QuHQ, GrantSF, BradfieldJP, KimC, FrackeltonE, et al. (2009) Association of RASGRP1 with type 1 diabetes is revealed by combined follow-up of two genome-wide studies. J Med Genet 46: 553–554.

25. SaxenaR, VoightBF, LyssenkoV, BurttNP, de BakkerPI, et al. (2007) Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316: 1331–1336.

26. ScottLJ, MohlkeKL, BonnycastleLL, WillerCJ, LiY, et al. (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316: 1341–1345.

27. SladekR, RocheleauG, RungJ, DinaC, ShenL, et al. (2007) A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445: 881–885.

28. SteinthorsdottirV, ThorleifssonG, ReynisdottirI, BenediktssonR, JonsdottirT, et al. (2007) A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet 39: 770–775.

29. ZegginiE, WeedonMN, LindgrenCM, FraylingTM, ElliottKS, et al. (2007) Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science 316: 1336–1341.

30. ProkopenkoI, PoonW, MagiR, PrasadBR, SalehiSA, et al. (2014) A central role for GRB10 in regulation of islet function in man. PLoS Genet 10: e1004235.

31. VoightBF, ScottLJ, SteinthorsdottirV, MorrisAP, DinaC, et al. (2010) Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat Genet 42: 579–589.

32. MillsteinJ, ZhangB, ZhuJ, SchadtEE (2009) Disentangling molecular relationships with a causal inference test. BMC Genet 10: 23.

33. ChenYA, LemireM, ChoufaniS, ButcherDT, GrafodatskayaD, et al. (2013) Discovery of cross-reactive probes and polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray. Epigenetics 8: 203–209.

34. DoiA, ParkIH, WenB, MurakamiP, AryeeMJ, et al. (2009) Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet 41: 1350–1353.

35. IrizarryRA, Ladd-AcostaC, WenB, WuZ, MontanoC, et al. (2009) The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet 41: 178–186.

36. LindgrenCM, MahtaniMM, WidenE, McCarthyMI, DalyMJ, et al. (2002) Genomewide search for type 2 diabetes mellitus susceptibility loci in Finnish families: the Botnia study. American journal of human genetics 70: 509–516.

37. PociotF, McDermottMF (2002) Genetics of type 1 diabetes mellitus. Genes Immun 3: 235–249.

38. ShiinaT, InokoH, KulskiJK (2004) An update of the HLA genomic region, locus information and disease associations: 2004. Tissue Antigens 64: 631–649.

39. BibikovaM, BarnesB, TsanC, HoV, KlotzleB, et al. (2011) High density DNA methylation array with single CpG site resolution. Genomics 98: 288–295.

40. YangBT, DayehTA, VolkovPA, KirkpatrickCL, MalmgrenS, et al. (2012) Increased DNA methylation and decreased expression of PDX-1 in pancreatic islets from patients with type 2 diabetes. Mol Endocrinol 26: 1203–1212.

41. SandoviciI, SmithNH, NitertMD, Ackers-JohnsonM, Uribe-LewisS, et al. (2011) Maternal diet and aging alter the epigenetic control of a promoter-enhancer interaction at the Hnf4a gene in rat pancreatic islets. Proc Natl Acad Sci U S A 108: 5449–5454.

42. ParkerSC, StitzelML, TaylorDL, OrozcoJM, ErdosMR, et al. (2013) Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proc Natl Acad Sci U S A 110: 17921–17926.

43. PasqualiL, GaultonKJ, Rodriguez-SeguiSA, MularoniL, Miguel-EscaladaI, et al. (2014) Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat Genet 46: 136–143.

44. BirdA (2007) Perceptions of epigenetics. Nature 447: 396–398.

45. GiladY, RifkinSA, PritchardJK (2008) Revealing the architecture of gene regulation: the promise of eQTL studies. Trends Genet 24: 408–415.

46. MantelN (1967) The detection of disease clustering and a generalized regression approach. Cancer research 27: 209–220.

47. BekrisLM, ShephardC, PetersonM, HoehnaJ, Van YserlooB, et al. (2005) Glutathione-s-transferase M1 and T1 polymorphisms and associations with type 1 diabetes age-at-onset. Autoimmunity 38: 567–575.

48. CircuML, AwTY (2008) Glutathione and apoptosis. Free Radic Res 42: 689–706.

49. RobertsonRP, HarmonJS (2007) Pancreatic islet beta-cell and oxidative stress: the importance of glutathione peroxidase. FEBS Lett 581: 3743–3748.

50. HarashimaSI, HarashimaC, NishimuraT, HuY, NotkinsAL (2007) Overexpression of the autoantigen IA-2 puts beta cells into a pre-apoptotic state: autoantigen-induced, but non-autoimmune-mediated, tissue destruction. Clin Exp Immunol 150: 49–60.

51. NguyenVD, SaaranenMJ, KaralaAR, LappiAK, WangL, et al. (2011) Two endoplasmic reticulum PDI peroxidases increase the efficiency of the use of peroxide during disulfide bond formation. J Mol Biol 406: 503–515.

52. PengD, BelkhiriA, HuT, ChaturvediR, AsimM, et al. (2012) Glutathione peroxidase 7 protects against oxidative DNA damage in oesophageal cells. Gut 61: 1250–1260.

53. UtomoA, JiangX, FurutaS, YunJ, LevinDS, et al. (2004) Identification of a novel putative non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase (NPGPx) essential for alleviating oxidative stress generated from polyunsaturated fatty acids in breast cancer cells. J Biol Chem 279: 43522–43529.

54. LuJ, LiQ, XieH, ChenZJ, BorovitskayaAE, et al. (1996) Identification of a second transmembrane protein tyrosine phosphatase, IA-2beta, as an autoantigen in insulin-dependent diabetes mellitus: precursor of the 37-kDa tryptic fragment. Proc Natl Acad Sci U S A 93: 2307–2311.

55. NotkinsAL, LernmarkA (2001) Autoimmune type 1 diabetes: resolved and unresolved issues. J Clin Invest 108: 1247–1252.

56. McCarthyMI (2010) Genomics, type 2 diabetes, and obesity. N Engl J Med 363: 2339–2350.

57. HindorffLA, SethupathyP, JunkinsHA, RamosEM, MehtaJP, et al. (2009) Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A 106: 9362–9367.

58. JohnsonAD, HandsakerRE, PulitSL, NizzariMM, O'DonnellCJ, et al. (2008) SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap. Bioinformatics 24: 2938–2939.

59. MorrisAP, VoightBF, TeslovichTM, FerreiraT, SegreAV, et al. (2012) Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet 44: 981–990.

60. DupuisJ, LangenbergC, ProkopenkoI, SaxenaR, SoranzoN, et al. (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42: 105–116.

61. SaxenaR, HivertMF, LangenbergC, TanakaT, PankowJS, et al. (2010) Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat Genet 42: 142–148.

62. SoranzoN, SannaS, WheelerE, GiegerC, RadkeD, et al. (2010) Common variants at 10 genomic loci influence hemoglobin A(1)(C) levels via glycemic and nonglycemic pathways. Diabetes 59: 3229–3239.

63. StrawbridgeRJ, DupuisJ, ProkopenkoI, BarkerA, AhlqvistE, et al. (2011) Genome-wide association identifies nine common variants associated with fasting proinsulin levels and provides new insights into the pathophysiology of type 2 diabetes. Diabetes 60: 2624–2634.

64. ManningAK, HivertMF, ScottRA, GrimsbyJL, Bouatia-NajiN, et al. (2012) A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat Genet 44: 659–669.

65. DayehTA, OlssonAH, VolkovP, AlmgrenP, RonnT, et al. (2013) Identification of CpG-SNPs associated with type 2 diabetes and differential DNA methylation in human pancreatic islets. Diabetologia 56: 1036–1046.

66. JonesPA (2012) Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13: 484–492.

67. LingC, GroopL (2009) Epigenetics: a molecular link between environmental factors and type 2 diabetes. Diabetes 58: 2718–2725.

68. YangBT, DayehTA, KirkpatrickCL, TaneeraJ, KumarR, et al. (2011) Insulin promoter DNA methylation correlates negatively with insulin gene expression and positively with HbA(1c) levels in human pancreatic islets. Diabetologia 54: 360–367.

69. LingC, Del GuerraS, LupiR, RonnT, GranhallC, et al. (2008) Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. Diabetologia 51: 615–622.

70. LingC, PoulsenP, SimonssonS, RonnT, HolmkvistJ, et al. (2007) Genetic and epigenetic factors are associated with expression of respiratory chain component NDUFB6 in human skeletal muscle. J Clin Invest 117: 3427–3435.

71. NitertMD, DayehT, VolkovP, ElgzyriT, HallE, et al. (2012) Impact of an exercise intervention on DNA methylation in skeletal muscle from first-degree relatives of patients with type 2 diabetes. Diabetes 61: 3322–3332.

72. RonnT, VolkovP, DavegardhC, DayehT, HallE, et al. (2013) A Six Months Exercise Intervention Influences the Genome-wide DNA Methylation Pattern in Human Adipose Tissue. PLoS Genet 9: e1003572.

73. VolkmarM, DedeurwaerderS, CunhaDA, NdlovuMN, DefranceM, et al. (2012) DNA methylation profiling identifies epigenetic dysregulation in pancreatic islets from type 2 diabetic patients. EMBO J 31: 1405–1426.

74. ToddJA, WalkerNM, CooperJD, SmythDJ, DownesK, et al. (2007) Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 39: 857–864.

75. MooreF, ColliML, CnopM, EsteveMI, CardozoAK, et al. (2009) PTPN2, a candidate gene for type 1 diabetes, modulates interferon-gamma-induced pancreatic beta-cell apoptosis. Diabetes 58: 1283–1291.

76. Wellcome Trust Case Control Consortium (2007) Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447: 661–678.

77. SmythDJ, PlagnolV, WalkerNM, CooperJD, DownesK, et al. (2008) Shared and distinct genetic variants in type 1 diabetes and celiac disease. N Engl J Med 359: 2767–2777.

78. de BakkerPI, McVeanG, SabetiPC, MirettiMM, GreenT, et al. (2006) A high-resolution HLA and SNP haplotype map for disease association studies in the extended human MHC. Nat Genet 38: 1166–1172.

79. ZegginiE, ScottLJ, SaxenaR, VoightBF, MarchiniJL, et al. (2008) Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet 40: 638–645.

80. JacobsenSC, BronsC, Bork-JensenJ, Ribel-MadsenR, YangB, et al. (2012) Effects of short-term high-fat overfeeding on genome-wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia 55: 3341–3349.

81. GillbergL, JacobsenSC, RonnT, BronsC, VaagA (2014) PPARGC1A DNA methylation in subcutaneous adipose tissue in low birth weight subjects - impact of 5days of high-fat overfeeding. Metabolism 63: 263–271.

82. BronsC, JacobsenS, NilssonE, RonnT, JensenCB, et al. (2010) Deoxyribonucleic acid methylation and gene expression of PPARGC1A in human muscle is influenced by high-fat overfeeding in a birth-weight-dependent manner. J Clin Endocrinol Metab 95: 3048–3056.

83. FlorezJC (2008) Newly identified loci highlight beta cell dysfunction as a key cause of type 2 diabetes: where are the insulin resistance genes? Diabetologia 51: 1100–1110.

84. RosengrenAH, BraunM, MahdiT, AnderssonSA, TraversME, et al. (2012) Reduced insulin exocytosis in human pancreatic beta-cells with gene variants linked to type 2 diabetes. Diabetes 61: 1726–1733.

85. RuchatSM, ElksCE, LoosRJ, VohlMC, WeisnagelSJ, et al. (2009) Association between insulin secretion, insulin sensitivity and type 2 diabetes susceptibility variants identified in genome-wide association studies. Acta Diabetol 46: 217–226.

86. CooperJD, SmythDJ, SmilesAM, PlagnolV, WalkerNM, et al. (2008) Meta-analysis of genome-wide association study data identifies additional type 1 diabetes risk loci. Nat Genet 40: 1399–1401.

87. PerryJR, VoightBF, YengoL, AminN, DupuisJ, et al. (2012) Stratifying type 2 diabetes cases by BMI identifies genetic risk variants in LAMA1 and enrichment for risk variants in lean compared to obese cases. PLoS Genet 8: e1002741.

88. SantinI, EizirikDL (2013) Candidate genes for type 1 diabetes modulate pancreatic islet inflammation and beta-cell apoptosis. Diabetes Obes Metab 15 Suppl 3: 71–81.

89. KanetoH, MiyatsukaT, KawamoriD, YamamotoK, KatoK, et al. (2008) PDX-1 and MafA play a crucial role in pancreatic beta-cell differentiation and maintenance of mature beta-cell function. Endocr J 55: 235–252.

90. DuttaS, GannonM, PeersB, WrightC, Bonner-WeirS, et al. (2001) PDX:PBX complexes are required for normal proliferation of pancreatic cells during development. Proc Natl Acad Sci U S A 98: 1065–1070.

91. DayehT, VolkovP, SaloS, HallE, NilssonE, et al. (2014) Genome-wide DNA methylation analysis of human pancreatic islets from type 2 diabetic and non-diabetic donors identifies candidate genes that influence insulin secretion. PLoS Genet 10: e1004160.

92. CaromileLA, OganesianA, CoatsSA, SeifertRA, Bowen-PopeDF (2010) The neurosecretory vesicle protein phogrin functions as a phosphatidylinositol phosphatase to regulate insulin secretion. J Biol Chem 285: 10487–10496.

93. DoiA, ShonoT, NishiM, FurutaH, SasakiH, et al. (2006) IA-2beta, but not IA-2, is induced by ghrelin and inhibits glucose-stimulated insulin secretion. Proc Natl Acad Sci U S A 103: 885–890.

94. CaiT, HiraiH, ZhangG, ZhangM, TakahashiN, et al. (2011) Deletion of Ia-2 and/or Ia-2beta in mice decreases insulin secretion by reducing the number of dense core vesicles. Diabetologia 54: 2347–2357.

95. ToriiS, SaitoN, KawanoA, HouN, UekiK, et al. (2009) Gene silencing of phogrin unveils its essential role in glucose-responsive pancreatic beta-cell growth. Diabetes 58: 682–692.

96. LiuY, AryeeMJ, PadyukovL, FallinMD, HesselbergE, et al. (2013) Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat Biotechnol 31: 142–147.

97. StorlingJ, OvergaardAJ, BrorssonCA, PivaF, Bang-BerthelsenCH, et al. (2013) Do post-translational beta cell protein modifications trigger type 1 diabetes? Diabetologia 56: 2347–2354.

98. KellyC, McClenaghanNH, FlattPR (2011) Role of islet structure and cellular interactions in the control of insulin secretion. Islets 3: 41–47.

99. SabraG, DubielEA, KuehnC, KhalfaouiT, BeaulieuJF, et al. (2013) INS-1 cell glucose-stimulated insulin secretion is reduced by the downregulation of the 67 kDa laminin receptor. Journal of tissue engineering and regenerative medicine 10.1002/term.1689.

100. SmithGC, KonychevaG, DziadekMA, RavelichSR, PatelS, et al. (2011) Pre- and postnatal methyl deficiency in the rat differentially alters glucose homeostasis. Journal of nutrigenetics and nutrigenomics 4: 175–191.

101. CnopM, WelshN, JonasJC, JornsA, LenzenS, et al. (2005) Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 54 Suppl 2: S97–107.

102. LernmarkA, LarssonHE (2013) Immune therapy in type 1 diabetes mellitus. Nat Rev Endocrinol 9: 92–103.

103. YangSN, BerggrenPO (2005) Beta-cell CaV channel regulation in physiology and pathophysiology. Am J Physiol Endocrinol Metab 288: E16–28.

104. CalderonB, CarreroJA, UnanueER (2014) The central role of antigen presentation in islets of Langerhans in autoimmune diabetes. Current opinion in immunology 26: 32–40.

105. GuoY, ZhuLR, LuG, WangH, HongJY (2012) Selective expression of CYP2A13 in human pancreatic alpha-islet cells. Drug metabolism and disposition: the biological fate of chemicals 40: 1878–1882.

106. In't VeldPA, PipeleersDG (1988) In situ analysis of pancreatic islets in rats developing diabetes. Appearance of nonendocrine cells with surface MHC class II antigens and cytoplasmic insulin immunoreactivity. J Clin Invest 82: 1123–1128.

107. StandopJ, SchneiderM, UlrichA, BuchlerMW, PourPM (2003) Differences in immunohistochemical expression of xenobiotic-metabolizing enzymes between normal pancreas, chronic pancreatitis and pancreatic cancer. Toxicologic pathology 31: 506–513.

108. TsoTK, HuangHY, ChangCK, LiaoYJ, HuangWN (2004) Clinical evaluation of insulin resistance and beta-cell function by the homeostasis model assessment in patients with systemic lupus erythematosus. Clinical rheumatology 23: 416–420.

109. VerganiA, FotinoC, D'AddioF, TezzaS, PodettaM, et al. (2013) Effect of the purinergic inhibitor oxidized ATP in a model of islet allograft rejection. Diabetes 62: 1665–1675.

110. ZengYJ, ZengFQ, DaiL, YangC, LinBZ, et al. (2011) [Insulin sensitivity and beta cell function in female systemic lupus erythematosus patients]. Zhonghua nei ke za zhi 50: 18–22.

111. ZhangL, Moffatt-BruceSD, GaughanAA, WangJJ, RajabA, et al. (2009) An anti-CD103 immunotoxin promotes long-term survival of pancreatic islet allografts. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons 9: 2012–2023.

112. QuonG, LippertC, HeckermanD, ListgartenJ (2013) Patterns of methylation heritability in a genome-wide analysis of four brain regions. Nucleic Acids Res 41: 2095–2104.

113. BellJT, PaiAA, PickrellJK, GaffneyDJ, Pique-RegiR, et al. (2011) DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol 12: R10.

114. NilssonE, JanssonPA, PerfilyevA, VolkovP, PedersenM, et al. (2014) Altered DNA methylation and differential expression of genes influencing metabolism and inflammation in adipose tissue from subjects with type 2 diabetes. Diabetes 63: 2962–2976.

115. SmemoS, TenaJJ, KimKH, GamazonER, SakabeNJ, et al. (2014) Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507: 371–375.

116. JjingoD, ConleyAB, YiSV, LunyakVV, JordanIK (2012) On the presence and role of human gene-body DNA methylation. Oncotarget 3: 462–474.

117. ZhiD, AslibekyanS, IrvinMR, ClaasSA, BoreckiIB, et al. (2013) SNPs located at CpG sites modulate genome-epigenome interaction. Epigenetics 8: 802–806.

118. ReynardLN, BuiC, SyddallCM, LoughlinJ (2014) CpG methylation regulates allelic expression of GDF5 by modulating binding of SP1 and SP3 repressor proteins to the osteoarthritis susceptibility SNP rs143383. Hum Genet 133: 1059–1073.

119. TaqiMM, BazovI, WatanabeH, SheedyD, HarperC, et al. (2011) Prodynorphin CpG-SNPs associated with alcohol dependence: elevated methylation in the brain of human alcoholics. Addiction biology 16: 499–509.

120. ParkJH, LeeJ, KimCH, LeeS (2014) The polymorphism (−600 C>A) of CpG methylation site at the promoter region of CYP17A1 and its association of male infertility and testosterone levels. Gene 534: 107–112.

121. MaunakeaAK, NagarajanRP, BilenkyM, BallingerTJ, D'SouzaC, et al. (2010) Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature 466: 253–257.

122. DeatonAM, BirdA (2011) CpG islands and the regulation of transcription. Genes Dev 25: 1010–1022.

123. DeatonAM, WebbS, KerrAR, IllingworthRS, GuyJ, et al. (2011) Cell type-specific DNA methylation at intragenic CpG islands in the immune system. Genome Res 21: 1074–1086.

124. ShuklaS, KavakE, GregoryM, ImashimizuM, ShutinoskiB, et al. (2011) CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature 479: 74–79.

125. SigurdssonMI, SmithAV, BjornssonHT, JonssonJJ (2009) HapMap methylation-associated SNPs, markers of germline DNA methylation, positively correlate with regional levels of human meiotic recombination. Genome Res 19: 581–589.

126. RakyanVK, BeyanH, DownTA, HawaMI, MaslauS, et al. (2011) Identification of type 1 diabetes-associated DNA methylation variable positions that precede disease diagnosis. PLoS Genet 7: e1002300.

127. ChenH, KazemierHG, de GrooteML, RuitersMH, XuGL, et al. (2014) Induced DNA demethylation by targeting Ten-Eleven Translocation 2 to the human ICAM-1 promoter. Nucleic Acids Res 42: 1563–1574.

128. ChristensenDP, DahllofM, LundhM, RasmussenDN, NielsenMD, et al. (2011) Histone deacetylase (HDAC) inhibition as a novel treatment for diabetes mellitus. Molecular medicine 17: 378–390.

129. ChristensenDP, GysemansC, LundhM, DahllofMS, NoesgaardD, et al. (2014) Lysine deacetylase inhibition prevents diabetes by chromatin-independent immunoregulation and beta-cell protection. Proc Natl Acad Sci U S A 111: 1055–1059.

130. KrusU, KingBC, NagarajV, GandasiNR, SjolanderJ, et al. (2014) The complement inhibitor CD59 regulates insulin secretion by modulating exocytotic events. Cell Metab 19: 883–890.

131. MahdiT, HanzelmannS, SalehiA, MuhammedSJ, ReinbotheTM, et al. (2012) Secreted frizzled-related protein 4 reduces insulin secretion and is overexpressed in type 2 diabetes. Cell Metab 16: 625–633.

132. TaneeraJ, LangS, SharmaA, FadistaJ, ZhouY, et al. (2012) A systems genetics approach identifies genes and pathways for type 2 diabetes in human islets. Cell Metab 16: 122–134.

133. OlssonAH, YangBT, HallE, TaneeraJ, SalehiA, et al. (2011) Decreased expression of genes involved in oxidative phosphorylation in human pancreatic islets from patients with type 2 diabetes. Eur J Endocrinol 165: 589–595.

134. StahleMU, BrandhorstD, KorsgrenO, KnutsonF (2011) Pathogen inactivation of human serum facilitates its clinical use for islet cell culture and subsequent transplantation. Cell Transplant 20: 775–781.

135. PurcellS, NealeB, Todd-BrownK, ThomasL, FerreiraMA, et al. (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81: 559–575.

136. TeschendorffAE, MenonU, Gentry-MaharajA, RamusSJ, GaytherSA, et al. (2009) An epigenetic signature in peripheral blood predicts active ovarian cancer. PLoS One 4: e8274.

137. DuP, KibbeWA, LinSM (2008) lumi: a pipeline for processing Illumina microarray. Bioinformatics 24: 1547–1548.

138. DuP, ZhangX, HuangCC, JafariN, KibbeWA, et al. (2010) Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics 11: 587.

139. BolstadBM, IrizarryRA, AstrandM, SpeedTP (2003) A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19: 185–193.

140. JohnsonWE, LiC, RabinovicA (2007) Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8: 118–127.

141. ShabalinAA (2012) Matrix eQTL: ultra fast eQTL analysis via large matrix operations. Bioinformatics 28: 1353–1358.

142. HohmeierHE, MulderH, ChenG, Henkel-RiegerR, PrentkiM, et al. (2000) Isolation of INS-1-derived cell lines with robust ATP-sensitive K+ channel-dependent and -independent glucose-stimulated insulin secretion. Diabetes 49: 424–430.

143. WeissT, GrellM, HessabiB, BourteeleS, MullerG, et al. (1997) Enhancement of TNF receptor p60-mediated cytotoxicity by TNF receptor p80: requirement of the TNF receptor-associated factor-2 binding site. J Immunol 158: 2398–2404.

144. MalmgrenS, SpegelP, DanielssonAP, NagornyCL, AnderssonLE, et al. (2013) Coordinate changes in histone modifications, mRNA levels, and metabolite profiles in clonal INS-1 832/13 beta-cells accompany functional adaptations to lipotoxicity. J Biol Chem 288: 11973–11987.

145. O'ConnellJ, GurdasaniD, DelaneauO, PirastuN, UliviS, et al. (2014) A general approach for haplotype phasing across the full spectrum of relatedness. PLoS Genet 10: e1004234.

146. HowieBN, DonnellyP, MarchiniJ (2009) A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet 5: e1000529.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 11
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#