#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome-wide histone modification profiling of inner cell mass and trophectoderm of bovine blastocysts by RAT-ChIP


Autoři: Tõnis Org aff001;  Kati Hensen aff001;  Rita Kreevan aff001;  Elina Mark aff002;  Olav Sarv aff003;  Reidar Andreson aff004;  Ülle Jaakma aff002;  Andres Salumets aff003;  Ants Kurg aff001
Působiště autorů: Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia aff001;  Chair of Animal Breeding and Biotechnology, Estonian University of Life Sciences, Tartu, Estonia aff002;  Competence Centre on Health Technologies, Tartu, Estonia aff003;  Department of Bioinformatics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia aff004;  Institute of Genomics, University of Tartu, Tartu, Estonia aff005;  Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia aff006;  Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia aff007;  Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland aff008
Vyšlo v časopise: PLoS ONE 14(11)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0225801

Souhrn

Chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq) has revolutionized our understanding of chromatin-related biological processes. The method, however, requires thousands of cells and has therefore limited applications in situations where cell numbers are limited. Here we describe a novel method called Restriction Assisted Tagmentation Chromatin Immunoprecipitation (RAT-ChIP) that enables global histone modification profiling from as few as 100 cells. The method is simple, cost-effective and takes a single day to complete. We demonstrate the sensitivity of the method by deriving the first genome-wide maps of histone H3K4me3 and H3K27me3 modifications of inner cell mass and trophectoderm of bovine blastocyst stage embryos.

Klíčová slova:

Blastocysts – Embryos – Gene expression – Gene regulation – Histone modification – Histones – Chromatin – Immunoprecipitation


Zdroje

1. Park PJ. ChIP-seq: advantages and challenges of a maturing technology. Nat. Rev. Genet. 2009;10:669–80. doi: 10.1038/nrg2641 19736561

2. Schmidt D, Wilson MD, Spyrou C, Brown GD, Hadfield J, Odom DT. ChIP-seq: Using high-throughput sequencing to discover protein?DNA interactions. Methods. 2009;48:240–8. doi: 10.1016/j.ymeth.2009.03.001 19275939

3. Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60. doi: 10.1038/nature06008 17603471

4. Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature. 2009;459:108–12. doi: 10.1038/nature07829 19295514

5. Adli M, Zhu J, Bernstein BE. Genome-wide chromatin maps derived from limited numbers of hematopoietic progenitors. Nat. Methods. 2010;7:615–8. doi: 10.1038/nmeth.1478 20622861

6. Shankaranarayanan P, Mendoza-Parra M-A, Walia M, Wang L, Li N, Trindade LM, et al. Single-tube linear DNA amplification (LinDA) for robust ChIP-seq. Nat. Methods. 2011;8:565–7. doi: 10.1038/nmeth.1626 21642965

7. Zwart W, Koornstra R, Wesseling J, Rutgers E, Linn S, Carroll JS. A carrier-assisted ChIP-seq method for estrogen receptor-chromatin interactions from breast cancer core needle biopsy samples. BMC Genomics. 2013;14:232. doi: 10.1186/1471-2164-14-232 23565824

8. Ng J-H, Kumar V, Muratani M, Kraus P, Yeo J-C, Yaw L-P, et al. In Vivo Epigenomic Profiling of Germ Cells Reveals Germ Cell Molecular Signatures. Dev. Cell. 2013;24:324–33. doi: 10.1016/j.devcel.2012.12.011 23352811

9. Lara-Astiaso D, Weiner A, Lorenzo-Vivas E, Zaretsky I, Jaitin DA, David E, et al. Chromatin state dynamics during blood formation. Science (80-). 2014;345:943–9.

10. Brind’Amour J, Liu S, Hudson M, Chen C, Karimi MM, Lorincz MC. An ultra-low-input native ChIP-seq protocol for genome-wide profiling of rare cell populations. Nat. Commun. 2015;6:6033. doi: 10.1038/ncomms7033 25607992

11. Schmidl C, Rendeiro AF, Sheffield NC, Bock C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat. Methods. 2015;12:963–5. doi: 10.1038/nmeth.3542 26280331

12. Zheng X, Yue S, Chen H, Weber B, Jia J, Zheng Y. Low-Cell-Number Epigenome Profiling Aids the Study of Lens Aging and Hematopoiesis. Cell Rep. 2015;13:1505–18. doi: 10.1016/j.celrep.2015.10.004 26549448

13. Cao Z, Chen C, He B, Tan K, Lu C. A microfluidic device for epigenomic profiling using 100 cells. Nat. Methods. 2015;12:959–62. doi: 10.1038/nmeth.3488 26214128

14. Rotem A, Ram O, Shoresh N, Sperling RA, Goren A, Weitz DA, et al. Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol. 2015;33:1165–72. doi: 10.1038/nbt.3383 26458175

15. van Galen P, Viny AD, Ram O, Ryan RJH, Cotton MJ, Donohue L, et al. A Multiplexed System for Quantitative Comparisons of Chromatin Landscapes. Mol. Cell. 2016;61:170–80. doi: 10.1016/j.molcel.2015.11.003 26687680

16. Zhang B, Zheng H, Huang B, Li W, Xiang Y, Peng X, et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature. 2016;537:553–7. doi: 10.1038/nature19361 27626382

17. Dahl JA, Jung I, Aanes H, Greggains GD, Manaf A, Lerdrup M, et al. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature. 2016;537:548–52. doi: 10.1038/nature19360 27626377

18. Skene PJ, Henikoff JG, Henikoff S. Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nat. Protoc. Nature Publishing Group; 2018;13:1006–19. doi: 10.1038/nprot.2018.015 29651053

19. Valensisi C, Liao JL, Andrus C, Battle SL, Hawkins RD. cChIP-seq: a robust small-scale method for investigation of histone modifications. BMC Genomics. BioMed Central; 2015;16:1083. doi: 10.1186/s12864-015-2285-7 26692029

20. Liu X, Wang C, Liu W, Li J, Li C, Kou X, et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature. 2016;537:558–62. doi: 10.1038/nature19362 27626379

21. Destouni A, Zamani Esteki M, Catteeuw M, Tšuiko O, Dimitriadou E, Smits K, et al. Zygotes segregate entire parental genomes in distinct blastomere lineages causing cleavage-stage chimerism and mixoploidy. Genome Res. 2016;26:567–78. doi: 10.1101/gr.200527.115 27197242

22. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods. 2013;10:1213–8. doi: 10.1038/nmeth.2688 24097267

23. Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein CB, et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature. 2011;473:43–9. doi: 10.1038/nature09906 21441907

24. Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, et al. The human genome browser at UCSC. Genome Res. 2002;12:996–1006. doi: 10.1101/gr.229102 12045153

25. García-Alcalde F, Okonechnikov K, Carbonell J, Cruz LM, Götz S, Tarazona S, et al. Qualimap: evaluating next-generation sequencing alignment data. Bioinformatics. 2012;28:2678–9. doi: 10.1093/bioinformatics/bts503 22914218

26. Ozawa M, Sakatani M, Yao J, Shanker S, Yu F, Yamashita R, et al. Global gene expression of the inner cell mass and trophectoderm of the bovine blastocyst. BMC Dev. Biol. 2012;12:33. doi: 10.1186/1471-213X-12-33 23126590

27. Brinkhof B, van Tol HT, Groot Koerkamp MJ, Riemers FM, IJzer SG, Mashayekhi K, et al. A mRNA landscape of bovine embryos after standard and MAPK-inhibited culture conditions: a comparative analysis. BMC Genomics. 2015;16:277. doi: 10.1186/s12864-015-1448-x 25888366

28. Nagatomo H, Akizawa H, Sada A, Kishi Y, Yamanaka K, Takuma T, et al. Comparing spatial expression dynamics of bovine blastocyst under three different procedures: in-vivo, in-vitro derived, and somatic cell nuclear transfer embryos. Jpn. J. Vet. Res. 2015;63:159–71. 26753242

29. Zhao X-M, Cui L-S, Hao H-S, Wang H-Y, Zhao S-J, Du W-H, et al. Transcriptome analyses of inner cell mass and trophectoderm cells isolated by magnetic-activated cell sorting from bovine blastocysts using single cell RNA-seq. Reprod. Domest. Anim. 2016;51:726–35. doi: 10.1111/rda.12737 27440443

30. Hosseini SM, Dufort I, Caballero J, Moulavi F, Ghanaei HR, Sirard MA. Transcriptome profiling of bovine inner cell mass and trophectoderm derived from in vivo generated blastocysts. BMC Dev. Biol. 2015;15:49. doi: 10.1186/s12861-015-0096-3 26681441

31. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat. Methods. 2012;9:357–9. doi: 10.1038/nmeth.1923 22388286

32. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–9. doi: 10.1093/bioinformatics/btp352 19505943

33. Ramírez F, Ryan DP, Grüning B, Bhardwaj V, Kilpert F, Richter AS, et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016;44:W160–5. doi: 10.1093/nar/gkw257 27079975

34. Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis CA, Doyle F, et al. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247 22955616

35. Xu S, Grullon S, Ge K, Peng W. Spatial clustering for identification of ChIP-enriched regions (SICER) to map regions of histone methylation patterns in embryonic stem cells. Methods Mol. Biol. 2014;1150:97–111. doi: 10.1007/978-1-4939-0512-6_5 24743992

36. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. Narnia; 2010;26:841–2. doi: 10.1093/bioinformatics/btq033 20110278

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

38. Blankenberg D, Von Kuster G, Coraor N, Ananda G, Lazarus R, Mangan M, et al. Galaxy: a web-based genome analysis tool for experimentalists. Curr. Protoc. Mol. Biol. 2010;Chapter 19:Unit 19.10.1–21.

39. Pohl A, Beato M. bwtool: a tool for bigWig files. Bioinformatics. Oxford University Press; 2014;30:1618–9. doi: 10.1093/bioinformatics/btu056 24489365

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

41. Heberle H, Meirelles GV, da Silva FR, Telles GP, Minghim R. InteractiVenn: a web-based tool for the analysis of sets through Venn diagrams. BMC Bioinformatics. BioMed Central; 2015;16:169. doi: 10.1186/s12859-015-0611-3 25994840

42. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods. 2013;10:1213–8. doi: 10.1038/nmeth.2688 24097267

43. Schmidl C, Rendeiro AF, Sheffield NC, Bock C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat. Methods. 2015;

44. Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT, Haugen E, et al. The accessible chromatin landscape of the human genome. Nature. 2012;489:75–82. doi: 10.1038/nature11232 22955617

45. Mercer TR, Edwards SL, Clark MB, Neph SJ, Wang H, Stergachis AB, et al. DNase I-hypersensitive exons colocalize with promoters and distal regulatory elements. Nat. Genet. 2013;45:852–9. doi: 10.1038/ng.2677 23793028

46. Kaya-Okur HS, Wu SJ, Codomo CA, Pledger ES, Bryson TD, Henikoff JG, et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. Nature Publishing Group; 2019;10:1930. doi: 10.1038/s41467-019-09982-5 31036827

47. Frank CL, Manandhar D, Gordân R, Crawford GE. HDAC inhibitors cause site-specific chromatin remodeling at PU.1-bound enhancers in K562 cells. Epigenetics Chromatin. 2016;9:15. doi: 10.1186/s13072-016-0065-5 27087856

48. Bogliotti YS, Wu J, Vilarino M, Okamura D, Soto DA, Zhong C, et al. Efficient derivation of stable primed pluripotent embryonic stem cells from bovine blastocysts. Proc. Natl. Acad. Sci. 2018;115:2090–5. doi: 10.1073/pnas.1716161115 29440377

49. Strumpf D, Mao C-A, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, et al. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132:2093–102. doi: 10.1242/dev.01801 15788452

50. Rayon T, Menchero S, Nieto A, Xenopoulos P, Crespo M, Cockburn K, et al. Notch and hippo converge on Cdx2 to specify the trophectoderm lineage in the mouse blastocyst. Dev. Cell. NIH Public Access; 2014;30:410–22.

51. Davies JOJ, Oudelaar AM, Higgs DR, Hughes JR. How best to identify chromosomal interactions: a comparison of approaches. Nat. Methods. 2017;14:125–34. doi: 10.1038/nmeth.4146 28139673

52. Arrigoni L, Al-Hasani H, Ramírez F, Panzeri I, Ryan DP, Santacruz D, et al. RELACS nuclei barcoding enables high-throughput ChIP-seq. Commun. Biol. Nature Publishing Group; 2018;1:214. doi: 10.1038/s42003-018-0219-z 30534606

53. Gustafsson C, De Paepe A, Schmidl C, Månsson R. High-throughput ChIPmentation: freely scalable, single day ChIPseq data generation from very low cell-numbers. BMC Genomics. 2019;20:59. doi: 10.1186/s12864-018-5299-0 30658577

54. Wallerman O, Nord H, Bysani M, Borghini L, Wadelius C. lobChIP: from cells to sequencing ready ChIP libraries in a single day. Epigenetics Chromatin. BioMed Central; 2015;8:25. doi: 10.1186/s13072-015-0017-5 26195988

55. Hainer SJ, Fazzio TG. High-Resolution Chromatin Profiling Using CUT&RUN. Curr. Protoc. Mol. Biol. 2019;e85. doi: 10.1002/cpmb.85 30688406

56. Hainer SJ, Bošković A, McCannell KN, Rando OJ, Fazzio TG. Profiling of Pluripotency Factors in Single Cells and Early Embryos. Cell. Cell Press; 2019;177:1319–1329.e11.

57. Ku WL, Nakamura K, Gao W, Cui K, Hu G, Tang Q, et al. Single-cell chromatin immunocleavage sequencing (scChIC-seq) to profile histone modification. Nat. Methods. 2019;16:323–5. doi: 10.1038/s41592-019-0361-7 30923384

58. Wang Q, Xiong H, Ai S, Yu X, Liu Y, Zhang J, et al. CoBATCH for High-Throughput Single-Cell Epigenomic Profiling. Mol. Cell. Cell Press; 2019;

59. Harada A, Maehara K, Handa T, Arimura Y, Nogami J, Hayashi-Takanaka Y, et al. A chromatin integration labelling method enables epigenomic profiling with lower input. Nat. Cell Biol. 2019;21:287–96. doi: 10.1038/s41556-018-0248-3 30532068

60. Hughes AL, Rando OJ. Mechanisms Underlying Nucleosome Positioning In Vivo. Annu. Rev. Biophys. 2014;43:41–63. doi: 10.1146/annurev-biophys-051013-023114 24702039

61. Kubik S, Bruzzone MJ, Jacquet P, Falcone J-L, Rougemont J, Shore D. Nucleosome Stability Distinguishes Two Different Promoter Types at All Protein-Coding Genes in Yeast. Mol. Cell. 2015;60:422–34. doi: 10.1016/j.molcel.2015.10.002 26545077

62. Voong LN, Xi L, Sebeson AC, Xiong B, Wang J-P, Wang X. Insights into Nucleosome Organization in Mouse Embryonic Stem Cells through Chemical Mapping. Cell. Cell Press; 2016;167:1555–1570.e15.

63. Ramani V, Qiu R, Shendure J. High Sensitivity Profiling of Chromatin Structure by MNase-SSP. Cell Rep. Cell Press; 2019;26:2465–2476.e4. doi: 10.1016/j.celrep.2019.02.007 30811994

64. Brahma S, Henikoff S. RSC-Associated Subnucleosomes Define MNase-Sensitive Promoters in Yeast. Mol. Cell. Elsevier; 2019;73:238–249.e3. doi: 10.1016/j.molcel.2018.10.046 30554944

65. Kidder BL, Hu G, Zhao K. ChIP-Seq: technical considerations for obtaining high-quality data. Nat. Immunol. 2011;12:918–22. doi: 10.1038/ni.2117 21934668

66. Flensburg C, Kinkel SA, Keniry A, Blewitt ME, Oshlack A. A comparison of control samples for ChIP-seq of histone modifications. Front. Genet. 2014;5.

67. Zhang Y-F, Su B. Peak identification for ChIP-seq data with no controls. Dong wu xue yan jiu = Zool. Res. 2012;33:E121–8. doi: 10.3724/SP.J.1141.2012.E120-06E121 23266983

68. Cheung M-S, Down TA, Latorre I, Ahringer J. Systematic bias in high-throughput sequencing data and its correction by BEADS. Nucleic Acids Res. Oxford University Press; 2011;39:e103–e103. doi: 10.1093/nar/gkr425 21646344

69. de Boer BA, van Duijvenboden K, van den Boogaard M, Christoffels VM, Barnett P, Ruijter JM. OccuPeak: ChIP-Seq Peak Calling Based on Internal Background Modelling. Langmann T, editor. PLoS One. Public Library of Science; 2014;9:e99844. doi: 10.1371/journal.pone.0099844 24936875

70. Szalkowski AM, Schmid CD. Rapid innovation in ChIP-seq peak-calling algorithms is outdistancing benchmarking efforts. Brief. Bioinform. Oxford University Press; 2011;12:626–33. doi: 10.1093/bib/bbq068 21059603

71. Thomas R, Thomas S, Holloway AK, Pollard KS. Features that define the best ChIP-seq peak calling algorithms. Brief. Bioinform. 2016;18:bbw035.

72. Chen K, Hu Z, Xia Z, Zhao D, Li W, Tyler JK. The Overlooked Fact: Fundamental Need for Spike-In Control for Virtually All Genome-Wide Analyses. Mol. Cell. Biol. American Society for Microbiology (ASM); 2016;36:662.

73. Egan B, Yuan C-C, Craske ML, Labhart P, Guler GD, Arnott D, et al. An Alternative Approach to ChIP-Seq Normalization Enables Detection of Genome-Wide Changes in Histone H3 Lysine 27 Trimethylation upon EZH2 Inhibition. PLoS One. Public Library of Science; 2016;11:e0166438. doi: 10.1371/journal.pone.0166438 27875550

74. Santos RR, Schoevers EJ, Roelen BAJ. Usefulness of bovine and porcine IVM/IVF models for reproductive toxicology. Reprod. Biol. Endocrinol. BioMed Central; 2014;12:117. doi: 10.1186/1477-7827-12-117 25427762

75. Benayoun BA, Pollina EA, Ucar D, Mahmoudi S, Karra K, Wong ED, et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell. NIH Public Access; 2014;158:673–88. doi: 10.1016/j.cell.2014.06.027 25083876

76. Topalovic V, Schwirtlich M, Stevanovic M, Mojsin M. Histone modifications on the promoters of human OCT4 and NANOG genes at the onset of neural differentiation of NT2/D1 cells. Biochem. 2017;82:715–22.

77. Zhao S, Xu J, Liu S, Cui K, Li Z, Liu N. Dppa3 in pluripotency maintenance of ES cells and early embryogenesis. J. Cell. Biochem. 2019;120:4794–9. doi: 10.1002/jcb.28063 30417435

78. Bakhtari A, Ross PJ. DPPA3 prevents cytosine hydroxymethylation of the maternal pronucleus and is required for normal development in bovine embryos. Epigenetics. 2014;9:1271–9. doi: 10.4161/epi.32087 25147917

79. Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, et al. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. Nature Research; 2017;550:67–73. doi: 10.1038/nature24033 28953884

80. Tang F, Barbacioru C, Bao S, Lee C, Nordman E, Wang X, et al. Tracing the derivation of embryonic stem cells from the inner cell mass by single-cell RNA-Seq analysis. Cell Stem Cell. Elsevier; 2010;6:468–78. doi: 10.1016/j.stem.2010.03.015 20452321

81. Kasinathan S, Orsi GA, Zentner GE, Ahmad K, Henikoff S. High-resolution mapping of transcription factor binding sites on native chromatin. Nat. Methods. 2014;11:203–9. doi: 10.1038/nmeth.2766 24336359


Článek vyšel v časopise

PLOS One


2019 Číslo 11
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#