Skip to content

Latest commit

 

History

History
382 lines (257 loc) · 51.4 KB

method_snippets.md

File metadata and controls

382 lines (257 loc) · 51.4 KB

Method snippets

Microarrays

Expression arrays

Illumina arrays

Arrays were processed using the 'oligo' [Carvalho B. S., and Irizarry, R. A. (2010). A Framework for Oligonucleotide Microarray Preprocessing. Bioinformatics, 26(19):2363-7.] BioConductor package, quality-controlled with arrayQualityMetrics [Kauffmann, A., Gentleman, R.,, Huber, W. (2009) arrayQualityMetrics--a bioconductor package for quality assessment of microarray data. Bioinformatics, 25(3):415-6.] and RMA [Rafael. A. Irizarry, Benjamin M. Bolstad, Francois Collin, Leslie M. Cope, Bridget Hobbs and Terence P. Speed (2003), Summaries of Affymetrix GeneChip probe level data Nucleic Acids Research 31(4):e15] normalized. Differentially expressed genes (as defined by a minimun 2 fold change in expression and a Benjamini-Hochberg FDR adjusted pvalue of less than 0.1) were identified using limma [Smyth, GK (2005). Limma: linear models for microarray data. In: 'Bioinformatics and Computational Biology Solutions using R and Bioconductor'. R. Gentleman, V. Carey, S. Dudoit, R. Irizarry, W. Huber (eds), Springer, New York, pages 397-420.].”

Affymetrix arrays

Arrays were processed using the 'oligo' [Carvalho B. S., and Irizarry, R. A. (2010). A Framework for Oligonucleotide Microarray Preprocessing. Bioinformatics, 26(19):2363-7.] BioConductor package, quality-controlled with arrayQualityMetrics [Kauffmann, A., Gentleman, R.,, Huber, W. (2009) arrayQualityMetrics--a bioconductor package for quality assessment of microarray data. Bioinformatics, 25(3):415-6.] and RMA [Rafael. A. Irizarry, Benjamin M. Bolstad, Francois Collin, Leslie M. Cope, Bridget Hobbs and Terence P. Speed (2003), Summaries of Affymetrix GeneChip probe level data Nucleic Acids Research 31(4):e15] normalized. Differentially expressed genes (as defined by a minimun 2 fold change in expression and a Benjamini-Hochberg FDR adjusted pvalue of less than 0.1) were identified using limma [Smyth, GK (2005). Limma: linear models for microarray data. In: 'Bioinformatics and Computational Biology Solutions using R and Bioconductor'. R. Gentleman, V. Carey, S. Dudoit, R. Irizarry, W. Huber (eds), Springer, New York, pages 397-420.].”

Methylation arrays

Illumina 850K DNA methylation arrays

Methylation arrays will be processed and analyzed using a combination of Bioconductor packages and custom scripts following the general steps outlined in this Bioconductor workflow (https://www.bioconductor.org/packages/devel/workflows/vignettes/methylationArrayAnalysis/inst/doc/methylationArrayAnalysis.html). Raw IDAT file values will be processed using minfi [Aryee, 2014]. For quality control, arrays with low mean detection p-values across probes or outlier status for internal control probes will be discarded. Probes with low mean detection p-values across samples, and cross-reactivity and/or potential SNPs [Pidsley, 2016] will be also be discarded. Arrays will be background corrected using the noom algorithm [Triche, 2013] and inter-array technical variation adjusted for via Functional normalization [Fortin, 2014]. In parallel, the methylumi library [Davis, 2020], will be used to process arrays and internal array SNP probes used to detect potential sample swaps. Beta methylation values from the minfi processed data will be logit transformed to M-values [Du, 2010] and differential methylation at the single probe and regional levels determined using limma [Ritchie, 2015] and DMRcate [Peters, 2015] respectively.

  1. Aryee MJ, Jaffe AE, Corrada-Bravo H, Ladd-Acosta C, Feinberg AP, Hansen KD, Irizarry RA (2014). “Minfi: A flexible and comprehensive Bioconductor package for the analysis of Infinium DNA Methylation microarrays.” Bioinformatics, 30(10), 1363–1369. doi: 10.1093/bioinformatics/btu049 .
  2. Davis S, Du P, Bilke S, Triche, Jr. T, Bootwalla M (2020). methylumi: Handle Illumina methylation data. R package version 2.36.0.
  3. Du, P., Zhang, X., Huang, C.-C., Jafari, N., Kibbe, W. A., Hou, L., & Lin, S. M. (2010). Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics, 11, 587.
  4. Fortin, J.-P., Labbe, A., Lemire, M., Zanke, B. W., Hudson, T. J., Fertig, E. J., Greenwood, C. M., & Hansen, K. D. (2014). Functional normalization of 450k methylation array data improves replication in large cancer studies. Genome Biology, 15(12), 503.
  5. Mansell, G., Gorrie-Stone, T. J., Bao, Y., Kumari, M., Schalkwyk, L. S., Mill, J., & Hannon, E. (2019). Guidance for DNA methylation studies: statistical insights from the Illumina EPIC array. BMC Genomics, 20(1), 366.
  6. Peters, T. J., Buckley, M. J., Statham, A. L., Pidsley, R., Samaras, K., V Lord, R., Clark, S. J., & Molloy, P. L. (2015). De novo identification of differentially methylated regions in the human genome. Epigenetics & Chromatin, 8, 6.
  7. Pidsley, R., Zotenko, E., Peters, T. J., Lawrence, M. G., Risbridger, G. P., Molloy, P., Van Djik, S., Muhlhausler, B., Stirzaker, C., & Clark, S. J. (2016). Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome Biology, 17(1), 208.
  8. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK (2015). “limma powers differential expression analyses for RNA-sequencing and microarray studies.” Nucleic Acids Research, 43(7), e47. doi: 10.1093/nar/gkv007 .
  9. Triche TJ, Weisenberger DJ, Van Den Berg D, Laird PW, Siegmund KD (2013). “Low-level processing of Illumina Infinium DNA Methylation BeadArrays.” Nucleic Acids Research, 41(7), e90. doi: 10.1093/nar/gkt090 .

Variant calling

Overall, the parameters of our workflows are based on GATK best practices (https://gatk.broadinstitute.org/hc/en-us/sections/360007226651-Best-Practices-Workflows), contributions from bcbio community (https://github.com/bcbio/bcbio-nextgen) and our own validations (https://github.com/bcbio/bcbio_validations/).

Read alignment

We align reads with bwa mem [1], using samtools [2], and sambamba [3], to sort bam files and mark duplicate reads.

[1]: Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. 2013 arXiv:1303.3997. https://github.com/lh3/bwa.

[2]: Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, and 1000 Genome Project Data Processing Subgroup, The Sequence alignment/map (SAM) format and SAMtools, Bioinformatics (2009) 25(16) 2078-9 [19505943]. https://github.com/samtools/.

[3] A. Tarasov, A. J. Vilella, E. Cuppen, I. J. Nijman, and P. Prins. Sambamba: fast processing of NGS alignment formats. Bioinformatics, 2015. https://github.com/biod/sambamba.

Quality control

We run many tools to gather QC metrics:

We aggregate all metrics in a single QC report with multiqc [4-8], (https://multiqc.info/).

[4]: DKFZ bias filter (https://github.com/DKFZ-ODCF/DKFZBiasFilter).

[5]: fastqc (https://www.bioinformatics.babraham.ac.uk/projects/fastqc).

6: qualimap (http://qualimap.bioinfo.cipf.es).

[7]: bcftools (http://www.htslib.org/doc/bcftools.html).

[8]: Ewels P, Magnusson M, Lundin S, Käller M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics. 2016 Oct 1;32(19):3047-8. doi: 10.1093/bioinformatics/btw354. Epub 2016 Jun 16. PMID: 27312411; PMCID: PMC5039924.

Coverage and callable regions

We calculate coverage using mosdepth [9] and calculate callable regions based on real coverage and bed files provided.

[9]: Pedersen BS, Quinlan AR. Mosdepth: quick coverage calculation for genomes and exomes. Bioinformatics. 2018;34(5):867‐868. doi:10.1093/bioinformatics/btx699. https://github.com/brentp/mosdepth.

SNP and indels in germline (WES, WGS, gene panels)

We support variant calling in germline with:

We support:

  • single sample variant calling
  • batch variant calling
  • population variant calling (joint genotyping).

Structural and copy number variants in germline (WGS data)

We call structural variants with

We annotate structural variant calls with coverage information using duphold (https://github.com/brentp/duphold)

[10]: Chen, X. et al. (2016) Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics, 32, 1220-1222. doi:10.1093/bioinformatics/btv710.

[11]: Layer RM, Chiang C, Quinlan AR, Hall IM. LUMPY: a probabilistic framework for structural variant discovery. Genome Biol. 2014;15(6):R84. Published 2014 Jun 26. doi:10.1186/gb-2014-15-6-r84.

[12]: Rausch T, Zichner T, Schlattl A, Stütz AM, Benes V, Korbel JO. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics. 2012;28(18):i333‐i339. doi:10.1093/bioinformatics/bts378.

[13]: Kronenberg ZN, Osborne EJ, Cone KR, et al. Wham: Identifying Structural Variants of Biological Consequence. PLoS Comput Biol. 2015;11(12):e1004572. Published 2015 Dec 1. doi:10.1371/journal.pcbi.1004572.

Somatic small variants

We call somatic variants in tumor only or tumor/normal mode with:

We support ensemble approach to combine somatic calls from several callers (https://github.com/bcbio/bcbio.variation.recall)

Somatic copy number variants

We use

Variant annotation

We annotate variants with

VEP

McLaren W, Gil L, Hunt SE, et al. The Ensembl Variant Effect Predictor. Genome Biol. 2016;17(1):122. Published 2016 Jun 6. doi:10.1186/s13059-016-0974-4, https://useast.ensembl.org/info/docs/tools/vep/index.html.

snpEff

A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3.", Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L, Land SJ, Lu X, Ruden DM. Fly (Austin). 2012 Apr-Jun;6(2):80-92. PMID: 22728672, https://pcingola.github.io/SnpEff/se_introduction/

vcfanno

Pedersen BS, Layer RM, Quinlan AR. Vcfanno: fast, flexible annotation of genetic variants. Genome Biol. 2016;17(1):118. Published 2016 Jun 1. doi:10.1186/s13059-016-0973-5, https://github.com/brentp/vcfanno.

We use many annotation sources:

gnomad

Karczewski et al.The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020 May;581(7809):434-443. doi: 10.1038/s41586-020-2308-7. Epub 2020 May 27. PMID: 32461654; PMCID: PMC7334197, https://gnomad.broadinstitute.org/

We create gemini database [16] as output (https://gemini.readthedocs.io/en/latest/). We support any internal vcf or bed based annotation (internal frequency database) via vcfanno.

[16]: Paila U, Chapman BA, Kirchner R, Quinlan AR. GEMINI: integrative exploration of genetic variation and genome annotations. PLoS Comput Biol. 2013;9(7):e1003153. doi:10.1371/journal.pcbi.1003153.

RNA-Seq

HiSeq Illumina sequencing will be performed on our behalf by ###. All samples will be indexed so that pools can be run across ### lanes of an 8-laned Illumina flow cell, providing an estimated 20-30 million single-end reads per sample.

All samples will be processed using an RNA-seq pipeline implemented in the bcbio-nextgen project (https://bcbio-nextgen.readthedocs.org/en/latest/). Raw reads will be examined for quality issues using FastQC (http://www.bioinformatics.babraham.ac.uk/projects/fastqc/) to ensure library generation and sequencing are suitable for further analysis.

If necessary, adapter sequences, other contaminant sequences such as polyA tails and low quality sequences with PHRED quality scores less than five will be trimmed from reads using atropos [https://github.com/jdidion/atropos; 10.5281/zenodo.596588]. Trimmed reads will be aligned to UCSC build ### of the ## # genome (###), augmented with transcript information from Ensembl release ### using STAR [Dobin, Alexander, Carrie A Davis, Felix Schlesinger, Jorg Drenkow, Chris Zaleski, Sonali Jha, Philippe Batut, Mark Chaisson, and Thomas R Gingeras. 2013. “STAR: Ultrafast Universal RNA-Seq Aligner..” Bioinformatics (Oxford, England) 29 (1). Oxford University Press: 15–21. doi:10.1093/bioinformatics/bts635.].

Alignments will be checked for evenness of coverage, rRNA content, genomic context of alignments (for example, alignments in known transcripts and introns), complexity and other quality checks using a combination of FastQC, Qualimap [García-Alcalde, F., Okonechnikov, K., Carbonell, J., Cruz, L. M., Götz, S., Tarazona, S., et al. (2012). Qualimap: evaluating next-generation sequencing alignment data. Bioinformatics (Oxford, England), 28(20), 2678–2679. http://doi.org/10.1093/bioinformatics/bts503], MultiQC (https://github.com/ewels/MultiQC) and custom tools.

Counts of reads aligning to known genes are generated by featureCounts [Liao, Yang, Gordon K Smyth, and Wei Shi. 2014. “featureCounts: an Efficient General Purpose Program for Assigning Sequence Reads to Genomic Features..” Bioinformatics (Oxford, England) 30 (7). Oxford University Press: 923–30. doi:10.1093/bioinformatics/btt656.]. In parallel, Transcripts Per Million (TPM) measurements per isoform will be generated by quasialignment using Salmon [Patro, R., Duggal, G., Love, M. et al. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods 14, 417–419 (2017). https://doi.org/10.1038/nmeth.4197].

If necessary for a project, novel transcripts will be identified via reference-guided assembly with Stringtie [Pertea, Mihaela, Geo M. Pertea, Corina M. Antonescu, Tsung-Cheng Chang, Joshua T. Mendell, and Steven L. Salzberg. 2015. “StringTie Enables Improved Reconstruction of a Transcriptome from RNA-Seq Reads.” Nature Biotechnology, February. Nature Publishing Group. http://www.nature.com/doifinder/10.1038/nbt.3122]., with novel transcripts filtered for coding potential agreement [Wang, Liguo, Hyun Jung Park, Surendra Dasari, Shengqin Wang, Jean-Pierre Kocher, and Wei Li. 2013. “CPAT: Coding-Potential Assessment Tool Using an Alignment-Free Logistic Regression Model..” Nucleic Acids Research 41 (6). Oxford University Press: e74–e74. doi: 10.1093/nar/gkt006.] with known genes to reduce false positive assemblies.

Differential expression at the gene level will be called with DESeq2 [Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 550 (2014). https://doi.org/10.1186/s13059-014-0550-8], preferring to use counts per gene estimated from the Salmon quasialignments by tximport [Soneson, C., Love, M. I., & Robinson, M. D. (2016). Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Research, 4. http://doi.org/10.12688/f1000research.7563.1]. Quantitating at the isoform level has been shown to produce more accurate results at the gene level.

Isoform differential expression will be called using the TPM quasialignments from Salmon converted to HDF5 format for use with Sleuth (https://github.com/pachterlab/sleuth). If necessary for a project, splicing event level calls will be generated using a combination of DEXSeq [Anders, S., Reyes, A., & Huber, W. (2012). Detecting differential usage of exons from RNA-seq data. Genome Research, 22(10), 2008–2017. http://doi.org/10.1101/gr.133744.111] and MISO [Analysis and design of RNA sequencing experiments for identifying isoform regulation. (2010)].

Lists of differentially expressed genes will be examined for gene ontology (GO) and KEGG term enrichment with clusterProfiler [Yu G, Wang L, Han Y and He Q (2012). “clusterProfiler: an R package for comparing biological themes among gene clusters.” OMICS: A Journal of Integrative Biology, 16(5), pp. 284-287.]. Functional redundancy in enriched GO terms will be reduced with GOSemSim [Yu G, Li F, Qin Y, Bo X, Wu Y and Wang S (2010). “GOSemSim: an R package for measuring semantic similarity among GO terms and gene products.” Bioinformatics, 26(7), pp. 976-978.] In addition, a cut-off-free gene set enrichment analysis (GSEA) will be performed using clusterProfiler and weighted fold change calculations from DESeq2.

mRNA

RNA quality will be performed using the Agilent Bioanalyzer (Agilent, Santa Clara, CA). Transcriptional profiling using the ### array will be performed at ###. All data analysis will be performed using the BioConductor framework [1]. Array quality will be assessed using the arrayQualityMetrics package [2], batch adjusted [3], normalized and summarized using Robust Multiarray Averaging (RMA) [4] and tested for genes with significant differential expression using limma [5].

[1]: Reimers, Mark, and Vincent J Carey. “Bioconductor: an Open Source Framework for Bioinformatics and Computational Biology.” Methods in Enzymology 411: 119–134. doi:10.1016/S0076-6879(06)11008-3. [2]: Kauffmann, Audrey, Robert Gentleman, and Wolfgang Huber. “arrayQualityMetrics--a Bioconductor Package for Quality Assessment of Microarray Data.” Bioinformatics (Oxford, England) 25, no. 3: 415–416. doi:10.1093/bioinformatics/btn647. [3]: Leek, Jeffrey T, Robert B Scharpf, Héctor Corrada Bravo, David Simcha, Benjamin Langmead, W Evan Johnson, Donald Geman, Keith Baggerly, and Rafael A Irizarry. “Tackling the Widespread and Critical Impact of Batch Effects in High-Throughput Data.” Nature Reviews Genetics 11, no. 10: 733–739. doi:10.1038/nrg2825. [4]: Wilson, Claire L, and Crispin J Miller. “Simpleaffy: a BioConductor Package for Affymetrix Quality Control and Data Analysis..” Bioinformatics (Oxford, England) 21, no. 18: 3683–3685. doi:10.1093/bioinformatics/bti605. [5]: Smyth, G. K. (2005). Limma: linear models for microarray data. In: Bioinformatics and Computational Biology Solutions using R and Bioconductor, R. Gentleman, V. Carey, S. Dudoit, R. Irizarry, W. Huber (eds.), Springer, New York, pages 397-420.

small RNA-seq

All samples will be processed using an small RNA-seq pipeline implemented in the bcbio-nextgen project (https://bcbio-nextgen.readthedocs.org/en/latest/). Raw reads will be examined for quality issues using FastQC (http://www.bioinformatics.babraham.ac.uk/projects/fastqc/) to ensure library generation and sequencing are suitable for further analysis.

Adapter sequences, other contaminant sequences such as polyA tails and low quality sequences with PHRED quality scores less than five will be trimmed from reads using atropos (Didion, John P et al. “Atropos: specific, sensitive, and speedy trimming of sequencing reads.” PeerJ vol. 5 e3720. 30 Aug. 2017, doi:10.7717/peerj.3720).

Trimmed reads will be aligned to miRBase v22 (Ana Kozomara, Maria Birgaoanu, Sam Griffiths-Jones, miRBase: from microRNA sequences to function, Nucleic Acids Research, Volume 47, Issue D1, 08 January 2019, Pages D155–D162, https://doi.org/10.1093/nar/gky1141) to the specific species with seqbuster (Pantano L, Estivill X, Martí E. SeqBuster, a bioinformatic tool for the processing and analysis of small RNAs datasets, reveals ubiquitous miRNA modifications in human embryonic cells. Nucleic Acids Res. 2010;38(5):e34. doi:10.1093/nar/gkp1127). As well, they will be aligned to ## # genome (version ##) using STAR (Dobin A., Davis C. A., Schlesinger F., Drenkow J., Zaleski C., Jha S., et al. (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29 15–21. 10.1093/bioinformatics/bts635). Alignments will be checked for evenness of coverage, rRNA content, genomic context, complexity and other quality checks using a combination of FastQC, MultiQC (Ewels P., Magnusson M., Lundin S., Kaller M. (2016). MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 32 3047–3048) and custom code inside bcbio-nextgen pipeline.The aligned sequences will be used by seqcluster (Pantano L., Estivill X., Marti E. (2011). A non-biased framework for the annotation and classification of the non-miRNA small RNA transcriptome. Bioinformatics 27 3202–3203) to characterize the whole small RNA transcriptome and classify reads into rRNA, miRNA, repeats, genes, tRNAs and others from USCC annotation (Mangan M. E., Williams J. M., Kuhn R.M., Lathe W.C., 3rd (2014). The UCSC genome browser: what every molecular biologist should know. Curr. Protoc. Mol. Biol. 107 19.9.1–19.9.36. 10.1002/0471142727.mb1909s107). Finally, aligned reads will be analyzed with miRDeep2 (Friedlander M. R., Mackowiak S. D., Li N., Chen W., Rajewsky N. (2012). miRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades. Nucleic Acids Res. 40 37–52. 10.1093/nar/gkr688), an algorithm that assesses the fit of sequenced RNAs to a biological model of miRNA biogeneesis and correct folding.

Data will be loaded into R with bcbioSmallRna R package (https://github.com/lpantano/bcbioSmallRna) and isomiRs BioC package (Ramos et al. 2017; L. Pantano, Escaramis, and Argyropoulos 2016) to generate the miRNA/isomiR count matrix and perform quality control, partial least square discriminant analysis (PLS-DA) and differential expression analysis. Differential expression of miRNAs will be called with DESeq2 [Love, Michael I, Wolfgang Huber, and Simon Anders. 2014. “Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2..” Genome Biology 15 (12): 550. doi:10.1186/PREACCEPT-8897612761307401] using negative binomial generalized linear model to set up the relevant comparisons. Pathway analysis will be performed using DIANA miRPath v3 which can use predicted miRNA targets or experimentally validated miRNA interactions to identify pathways that may be affected (Vlachos, Ioannis S., Konstantinos Zagganas, Maria D. Paraskevopoulou, Georgios Georgakilas, Dimitra Karagkouni, Thanasis Vergoulis, Theodore Dalamagas, and Artemis G. Hatzigeorgiou. "DIANA-miRPath v3. 0: deciphering microRNA function with experimental support." Nucleic acids research (2015): gkv403).

miRNA/mRNA integration

To comprehensively integrate the interaction of differentially expressed genes and miRNAs we will use miRTrail [ Laczny, Cedric, Petra Leidinger, Jan Haas, Nicole Ludwig, Christina Backes, Andreas Gerasch, Michael Kaufmann, et al. “miRTrail--a Comprehensive Webserver for Analyzing Gene and miRNA Patterns to Enhance the Understanding of Regulatory Mechanisms in Diseases..” BMC Bioinformatics 13 (2012): 36. doi:10.1186/1471-2105-13-36.] which integrates information on 20.000 genes and almost 1.000 miRNAs. A secondary screen will be performed using MMIA [Nam, Seungyoon, Meng Li, Kwangmin Choi, Curtis Balch, Sun Kim, and Kenneth P Nephew. “MicroRNA and mRNA Integrated Analysis (MMIA): a Web Tool for Examining Biological Functions of microRNA Expression.” Nucleic Acids Research 37, no. Web Server issue (July 1, 2009): W356–62. doi:10.1093/nar/gkp294.], an algorithm incorporating three miRNA prediction algorithms (TargetScan, PITA and PicTar) into the mRNA expression analysis.

ChIP-Seq

ChIP-seq data quality will be evaluated using FASTQC 6, and if required filtering and trimming of reads will be performed with Atropos [7]. High quality reads will be mapped to the current mouse genome build using Bowtie2 [8]. Multi-mapping reads will be excluded. We will use MACS2 [9] to call peaks on unique reads only and assess peak quality using ChIPQC [10]. Depending on signal strength and the width of the peaks, EPIC2 [11] may also be used to verify broad peak calls. Likely peak artifacts will be filtered out using the ENCODE blacklist [12]. Data will be visualized using IGV [13]. DeepTools [14] will be used to assess coverage and the reproducibility of peaks across replicates. Consensus peak sets will be generated by expanding each summit 250 bases and choosing among overlapping peaks the peak with the highest score [15]. ChIPseeker [16] will be used to annotate ChIP-seq peaks to their closest transcriptional start site and perform functional enrichment analysis. Binding sites will be evaluated using the MEME [17] suite of tools for motif discovery and motif enrichment.

[7]: Didion JP, Martin M, Collins FS. Atropos: specific, sensitive, and speedy trimming of sequencing reads. PeerJ. 2017 Aug 30;5:e3720. doi: 10.7717/peerj.3720. PMID: 28875074; PMCID: PMC5581536.

[8]: Langmead B, Salzberg S. Fast gapped-read alignment with Bowtie 2. Nature Methods. 2012, 9:357-359.

[9]: Zhang, Yong, Tao Liu, Clifford A Meyer, Jérôme Eeckhoute, David S Johnson, Bradley E Bernstein, Chad Nussbaum, et al. “Model-Based Analysis of ChIP-Seq (MACS).” Genome Biology 9, no. 9: R137. doi:10.1186/gb-2008-9-9-r137.

[10]: Carroll TS, Liang Z, Salama R, Stark R, de Santiago I. Impact of artifact removal on ChIP quality metrics in ChIP-seq and ChIP-exo data. Front Genet. 2014;5(APR):1–11.

[11]: Stovner EB, Sætrom P. epic2 efficiently finds diffuse domains in ChIP-seq data. Bioinformatics. 2019 Nov 1;35(21):4392-4393. doi: 10.1093/bioinformatics/btz232. PubMed PMID: 30923821.

[12]: Amemiya, H.M., Kundaje, A. & Boyle, A.P. The ENCODE Blacklist: Identification of Problematic Regions of the Genome. Sci Rep 9, 9354 (2019). https://doi.org/10.1038/s41598-019-45839-z.

[13]: Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, Mesirov JP. Integrative genomics viewer. Nat Biotechnol. 2011 Jan;29(1):24-6. doi: 10.1038/nbt.1754. PMID: 21221095; PMCID: PMC3346182.

[14]: Ramírez, Fidel, Devon P. Ryan, Björn Grüning, Vivek Bhardwaj, Fabian Kilpert, Andreas S. Richter, Steffen Heyne, Friederike Dündar, and Thomas Manke. deepTools2: A next Generation Web Server for Deep-Sequencing Data Analysis. Nucleic Acids Research (2016). doi:10.1093/nar/gkw257.

[15]: Neph S, Kuehn MS, Reynolds AP, et al. BEDOPS: high-performance genomic feature operations. Bioinformatics. 2012;28(14):1919‐1920. doi:10.1093/bioinformatics/bts277.

[16]: Yu G, Wang L, He Q (2015). “ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization.” Bioinformatics, 31(14), 2382-2383. doi: 10.1093/bioinformatics/btv145.

[17]: Bailey, Timothy L, Mikael Bodén, Fabian A Buske, Martin Frith, Charles E Grant, Luca Clementi, Jingyuan Ren, Wilfred W Li, and William S Noble. “MEME SUITE: Tools for Motif Discovery and Searching..” Nucleic Acids Research 37, no. Web Server issue: W202–8. doi:10.1093/nar/gkp335.

TF

Likely transcription factors (TF) associated with differentially expressed genes will be identified using oPOSSUM-3[1], a web-accessible software system for identification of over-represented transcription factor binding sites (TFBS) and TFBS families in DNA sequences of co-expressed genes.

[1]: Kwon, Andrew T, David J Arenillas, Rebecca Worsley Hunt, and Wyeth W Wasserman. “oPOSSUM-3: Advanced Analysis of Regulatory Motif Over-Representation Across Genes or ChIP-Seq Datasets..” G3 (Bethesda, Md.) 2, no. 9 (September 2012): 987–1002. doi:10.1534/g3.112.003202.

Integrative analysis of RNA-seq and ChIP-seq data

RNA-seq and ChIP-seq data will be integrated using BETA [Wang S, Sun H, Ma J, Zang C, Wang C, Wang J, Tang Q, Meyer CA, Zhang Y, Liu XS. Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat Protoc. 2013 Dec;8(12):2502-15. doi: 10.1038/nprot.2013.150. Epub 2013 Nov 21. PMID: 24263090; PMCID: PMC4135175] to identify which of the differentially expressed genes are associated with epigenetic changes. BETA uses a computational model to quantify the regulatory potential of a gene based on the number and distance of regulatory sites around the gene, and categorizes the effects as activating, repressive or both.

RRBS

HiSeq Illumina sequencing will be performed on our behalf by ###. All samples will be indexed so that ### RRBS pools can be run on a single lane of an 8-laned Illumina flow cell, providing an estimated ### million reads per sample.

Illumina sequence quality will be surveyed using FastQC [Andrews, S.: FastQC http://www.bioinformatics.babraham.ac.uk/projects/fastqc/] to ensure library generation and sequencing are suitable for further analysis. Particular attention will be paid to per base sequence quality, overrepresented sequences (an indicator of adapter contamination) and the per base cytosine content (an indicator of conversion efficiency). Using a bioinformatics pipeline constructed in bpipe [Sadedin, S. P., Pope, B., & Oshlack, A. (2012). Bpipe: a tool for running and managing bioinformatics pipelines. Bioinformatics (Oxford, England), 28(11), 1525–1526. doi:10.1093/bioinformatics/bts167], sequences will then be trimmed, aligned and their cytosine methylation status determined. Adapters will be removed and low quality bases (<25–30 Phred quality scores) adaptively trimmed from reads with Trim Galore [Krueger F: Trim Galore!. http://www.bioinformatics.babraham.ac.uk/projects/trim_galore/]. In addition, bases containing a cytosine artificially introduced in the end-repair step during the MspI-RRBS library preparation will be removed. Sequence quality will be re-assessed with FastQC and trimming parameters adjusted if necessary. Trimmed reads will be aligned to the appropriate reference genome with Bismark [Krueger, F., & Andrews, S. R. (2011). Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics (Oxford, England), 27(11), 1571–1572. doi:10.1093/bioinformatics/btr167], a ‘three-letter’ bisulfite aligner based on the gapped aligner Bowtie2 [Langmead, B., & Salzberg, S. L. (2012). Fast gapped-read alignment with Bowtie 2. Nature Methods, 9(4), 357–359. doi:10.1038/nmeth.1923]. Aligned reads will be automatically trimmed with BSeQC to remove nucleotides subject to further technical biases that can result in inaccurate methylation estimation, such as higher bisulfite conversion failure at the 5’ end of reads [Lin, X., Lin, X., Sun, D., Sun, D., Rodriguez, B., Rodriguez, B., et al. (2013). BSeQC: quality control of bisulfite sequencing experiments. Bioinformatics, 29(24), 3227–3229. doi:10.1093/bioinformatics/btt548]. Cytosine methylation will be determined with Bis-SNP [Liu, Y., Siegmund, K. D., Laird, P. W., & Berman, B. P. (2012). Bis-SNP: Combined DNA methylation and SNP calling for Bisulfite-seq data. Genome Biology, 13(7), R61. doi:10.1186/gb–2012–13–7-r61 ], a GATK [McKenna, A., Hanna, M., Banks, E., Sivachenko, A., Cibulskis, K., Kernytsky, A., et al. (2010). The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Research, 20(9), 1297–1303. doi:10.1101/gr.107524.110] based framework capable of simultaneous genotyping and DNA methylation estimation; methylation at non-CpG cytosines will be used to assess conversion rates. DNA methylation will be validated by bisulfite PCR or pyrosequencing on a subset of CpGs.

inDrop RNA-seq

Sequencing via the inDrop method (XXX link for inDrop) will be performed by the Single Cell Core at HMS. Prior to library preparation, cells will be rigorously quality controlled for viability. A total of XXX cells per sample will be captured, encapsulated in hydrogel and libraries will be prepared using a modified version of the protocol outlined previously [Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA, Kirschner MW (2015). Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells.]. Each cell will be marked by a unique cellular barcode and all transcript fragments for each cell will have a universal molecular identifier (UMI) attached. Sequencing of the inDrop libraries will be performed at (XXX sequencing core), with a target of (XXX) reads per sample.

Post sequencing, reads will be assigned to each cell via identifying the cellular barcodes, and the UMI will be extracted from each read [Power Analysis of Single Cell RNA‐Sequencing Experiments View ORCID ProfileValentine Svensson, Kedar N Natarajan, Lam-Ha Ly, Ricardo J Miragaia, Charlotte Labalette, Iain C Macaulay, Ana Cvejic, Sarah A Teichmann doi: http://dx.doi.org/10.1101/073692]. The distribution of reads per cell will be used to identify a cutoff for total reads sequenced that is the hallmark of high quality RNA from a cell. Reads from cells passing the quality filter will be aligned to (XXX genome/build) using Rapmap [RapMap: a rapid, sensitive and accurate tool for mapping RNA-seq reads to transcriptomes. (2016). RapMap: a rapid, sensitive and accurate tool for mapping RNA-seq reads to transcriptomes., 32(12), i192–i200. http://doi.org/10.1093/bioinformatics/btw277] and counts of reads per transcript per unique UMI will be generated for each cell. Reads will also be aligned to (XXX genome/build) using kallisto in single-cell mode to generate transcript compatibility counts (TCC) [Fast and accurate single-cell RNA-seq analysis by clustering of transcript-compatibility counts. (2016). Fast and accurate single-cell RNA-seq analysis by clustering of transcript-compatibility counts., 17(1), 112. http://doi.org/10.1186/s13059-016-0970-8]for each cell.

Heterogeneity analysis of the UMI disambiguated counts per gene per cell will be performed using the Seurat R package [Spatial reconstruction of single-cell gene expression data. (2015). Spatial reconstruction of single-cell gene expression data., 33(5), 495–502. http://doi.org/10.1038/nbt.3192]. This includes normalization and transformation of the raw gene counts per cell to account for differences in sequencing depth, identification of high variance genes, regression of sources of unwanted variation (e.g. cell cycle phrase), identification of primary sources of heterogeneity using PCA analysis, and clustering of cells based on significant PCs (metagenes).

Non-linear dimensional reduction using tSNE and UMAP [Etienne Becht, Charles-Antoine Dutertre, Immanuel W.H. Kwok, Lai Guan Ng, Florent Ginhoux, Evan W Newell (2018). Evaluation of UMAP as an alternative to t-SNE for single-cell data. bioRxiv 298430; doi: https://doi.org/10.1101/298430.] will be performed to visualize and explore data. Cluster quality control will be performed to assess possible cluster artifacts (variance correlated with UMI counts, mitochondrial ratio, cell cycle batch effects and any other principle component biases). After assigning clusters using sets of marker genes, differential expression analysis will be performed using zinbwave [Risso D, Perraudeau F, Gribkova S, Dudoit S, Vert J (2018). “A general and flexible method for signal extraction from single-cell RNA-seq data.” Nature Communications, 9, 284. https://doi.org/10.1038/s41467-017-02554-5] and edgeR [Robinson MD, McCarthy DJ, Smyth GK (2010). “edgeR: a Bioconductor package for differential expression analysis of digital gene expression data.” Bioinformatics, 26(1), 139-140].

Single cell RNA-seq-2

We used bcbio-nextgen https://bcbio-nextgen.readthedocs.io, https://github.com/bcbio/bcbio-nextgen, https://doi.org/10.5281/zenodo.3564938 to process data starting from Illumina fastq files to counts. Namely, we parsed barcode information, demultiplexed reads by samples, filtered erroneous barcodes, and calculated sample-cell-barcode counts with umis https://github.com/vals/umis. Before counting, we created a pseudoalignment with rapmap https://github.com/COMBINE-lab/RapMap. All steps are described in detail in the documentation https://bcbio-nextgen.readthedocs.io/en/latest/contents/single_cell.html#st…. We created a Seurat object using the count matrix and continued analysis (quality control, clustering, markers, differential expression) in Seurat https://satijalab.org/seurat/, https://www.nature.com/articles/nbt.4096 using R https://www.r-project.org/ and r-studio https://rstudio.com/. To create quality control plots we used ggplot2, tidyverse https://www.tidyverse.org/ and knitr https://yihui.org/knitr/. For the pseudobulk differential expression analysis, we used SingleCellExperiment object [https://bioconductor.org/packages/release/bioc/html/SingleCellExperiment.ht…](https://bioconductor.org/packages/release/bioc/html/SingleCellExperiment.ht…],

Lun A, Risso D (2020). SingleCellExperiment: S4 Classes for Single Cell Data. R package version 1.10.1.) ) and edgeR package https://bioconductor.org/packages/release/bioc/html/edgeR.html, Robinson MD, McCarthy DJ, Smyth GK (2010). “edgeR: a Bioconductor package for differential expression analysis of digital gene expression data.” Bioinformatics, 26(1), 139-140. doi: 10.1093/bioinformatics/btp616.)

We used mm10 mouse genome reference (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/) and ensembl-94 annotation of genes (ftp://ftp.ensembl.org/pub/release-94/gtf/mus_musculus).

10X single cell analysis

We used Cell Ranger 3.0.2 (10X Genomics) to process the raw sequencing data. This pipeline converted Illumina basecall files to fastq format, aligned sequencing reads to the mm10 transcriptome using the STAR aligner (Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013 Jan;29(1):15-21. doi: 10.1093/bioinformatics/bts635. Epub 2012 Oct 25. PubMed PMID: 23104886; PubMed Central PMCID: PMC3530905.), and quantified the expression of transcripts in each cell using Chromium barcodes.

We carried out analyses of processed scRNASeq data in R version [#.#.#] using the Seurat suite version [#.#.#] (Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018 Jun;36(5):411-420. doi: 10.1038/nbt.4096. Epub 2018 Apr 2. PubMed PMID: 29608179.; Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, Trombetta JJ, Weitz DA, Sanes JR, Shalek AK, Regev A, McCarroll SA. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015 May 21;161(5):1202-1214. doi: 10.1016/j.cell.2015.05.002. PubMed PMID: 26000488; PubMed Central PMCID: PMC4481139) and tidyverse packages (Hadley Wickham (2017). tidyverse: Easily Install and Load the 'Tidyverse'. R package version 1.2.1. https://CRAN.R-project.org/package=tidyverse). We obtained data from [#] cells from [#] samples that passed quality control steps implemented in Cell Ranger.

As a further quality-control measure, we filtered out cells meeting any of the following criteria: <### or >#### unique genes expressed, <#### or >#### UMIs, or >#% of reads mapping to mitochondria. These steps removed an additional #### cells, resulting in a final dataset of ### cells. We quantified gene expression across ### genes, with ### cells expression > # genes.

To explore transcriptional heterogeneity and to undertake initial cell clustering, we reduced dimensionality using PCA. For the overall dataset, we selected ## PCs that explained more variability than expected by chance using a permutation-based test as implemented in Seurat (Macosko et al., 2015). We used PC loadings as input for a graph-based approach to cluster cells by cell type (Villani et al., 2017) and as input for t-distributed stochastic neighbor embedding (t-SNE; L van der Maaten, G Hinton (2008). “Visualizating data using t-SNE.” The Journal of Machine Learning Research 9 (2579-2605), 85) for reduction to two dimensions for visualization purposes. To maintain a standard procedure for clustering, we used a value of #.# for the resolution. This resulted in ## clusters.

Specific gene markers for each cluster were identified with the FindAllMarkers() function using the Wilcoxon rank sum test. Cluster quality was assessed for possible cluster artifacts (variance correlated with UMI counts, mitochondrial ratio, batch effects, and any other principle component biases). Clusters were assigned based on specific gene markers as visualized in Spring (Weinreb C, Wolock S, Klein AM. SPRING: a kinetic interface for visualizing high dimensional single-cell expression data. Bioinformatics. 2018 Apr 1;34(7):1246-1248. doi: 10.1093/bioinformatics/btx792. Epub 2017 Dec 7. PubMed PMID: 29228172; PubMed Central PMCID: PMC6030950.). Differential expression at the gene level between clusters was performed with DESeq2 1.20 (Love MI, Huber W, Anders S (2014). “Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.” Genome Biology, 15, 550. doi: 10.1186/s13059-014-0550-8.] (https://bioconductor.org/packages/DESeq2/).

DGE

Reads were processed to counts through the bcbio-nextgen single cell/DGE RNA-seq analysis pipeline (1). A brief description follows: The well barcode and UMIs were identified for all reads and all reads not within one edit distance of a known well barcode were discarded. Each surviving read was quasialigned to the transcriptome (GRCh38)using RapMap (2) Reads per well were counted using umis (3), discarding duplicated UMIs, weighting multimapped reads by the number of transcripts they aligned to and collapsing counts to genes by adding all counts for each transcript of a gene. The R package DESeq2 (4) will be used for differential expression analysis.

  1. https://bcbio-nextgen.readthedocs.io/en/latest/contents/3prime_dge.html.
  2. Srivastava, Avi, Hirak Sarkar, Nitish Gupta, and Rob Patro. 2016. “RapMap: A Rapid, Sensitive and Accurate Tool for Mapping RNA-Seq Reads to Transcriptomes.” Bioinformatics 32 (12): i192–200.
  3. Svensson, Valentine, Kedar Nath Natarajan, Lam-Ha Ly, Ricardo J. Miragaia, Charlotte Labalette, Iain C. Macaulay, Ana Cvejic, and Sarah A. Teichmann. 2017. “Power Analysis of Single-Cell RNA-Sequencing Experiments.” Nature Methods 14 (4): 381–87.
  4. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi:10.1186/s13059-014-0550-8.

ATAC-seq

(Newer)

Data were processed using the ATAC-seq pipeline implemented in bcbio-nextgen vx.x.x. ATAC-seq data was evaluated for quality using FASTQC (Andrews, 2010)[1a]. Reads were filtered and trimmed with Atropos (Didion et al., 2017)[2a]. High quality reads were mapped to the xxx genome (build xxx) using Bowtie2 (Langmead et al., 2009)[3a]. After filtering reads from mitochondrial DNA, properly paired reads with high mapping quality (MAPQ score >10, non-duplicates, qualified reads) were retained using Sambamba (Tarasov, et al., 2015)[4a] for further analysis. The 'alignmentSieve' function of Deeptools (Ramirez et al., 2014)[5a] and 'sort' and 'index' functions of Samtools (Li et al., 2009)[6a] were used to isolate fragments in nucleosome free regions (NFRs). Reads were shifted by 9 bp (+4 in positive and -5 in negative strand) to account for the dimeric binding of the Tn5 transposase that results in insertion of two adaptors separated by 9 bp. To call the peaks with unique reads, we used MACS2 (Zhang et al, 2008)[7a]. ATAC-seq data quality was assessed using ataqv (Orchard et al, 2020)[8a]. CPM-normalized bigwig files (bin size=20) were visualized using IGV (Robinson et al., 2011)[9a]. Sets of peaks were compared using BEDTools (Quinlan & Hall, 2010)[10a].

Statistical analysis was performed in R v.x.x.x. Differential accessibility was assessed with Diffbind (Stark et al, 2011)[11a] using DESeq2 (Love et al., 2014)[12a] [[including batch in the model]]. Peaks were considered differentially enriched at FDR < 0.05. The genomic distribution of the peaks was annotated using ChIPseeker (Yu et al, 2015)[13a]. Functional enrichment analysis was performed using ClusterProfiler (Wu et al, 2021)[14a].

[1a]: Andrews S. (2010). "FastQC: a quality control tool for high throughput sequence data." Available online at: http://www.bioinformatics.babraham.ac.uk/projects/fastqc.

[2a]: Didion, John P et al. “Atropos: specific, sensitive, and speedy trimming of sequencing reads.” PeerJ vol. 5 e3720. 30 Aug. 2017, doi:10.7717/peerj.3720.

[3a]: Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25. doi: 10.1186/gb-2009-10-3-r25. Epub 2009 Mar 4. PubMed PMID: 19261174; PubMed Central PMCID: PMC2690996.

[4a]: Tarasov, Artem et al. “Sambamba: fast processing of NGS alignment formats.” Bioinformatics (Oxford, England) vol. 31,12 (2015): 2032-4. doi:10.1093/bioinformatics/btv098

[5a]: Ramírez, Fidel et al. “deepTools: a flexible platform for exploring deep-sequencing data.” Nucleic acids research vol. 42,Web Server issue (2014): W187-91. doi:10.1093/nar/gku365

[6a]: Li H., Handsaker B., Wysoker A., Fennell T., Ruan J., Homer N., Marth G., Abecasis G., Durbin R. and 1000 Genome Project Data Processing Subgroup (2009) The Sequence alignment/map (SAM) format and SAMtools. Bioinformatics, 25, 2078-9. [PMID: 19505943]

[7a]: Zhang, Yong, Tao Liu, Clifford A Meyer, Jérôme Eeckhoute, David S Johnson, Bradley E Bernstein, Chad Nussbaum, et al. “Model-Based Analysis of ChIP-Seq (MACS).” Genome Biology 9, no. 9: R137. doi:10.1186/gb-2008-9-9-r137

[8a]: Carroll TS, Liang Z, Salama R, Stark R, de Santiago I. Impact of artifact removal on ChIP quality metrics in ChIP-seq and ChIP-exo data. Front Genet. 2014 Apr 10;5:75. doi: 10.3389/fgene.2014.00075. eCollection 2014. PubMed PMID: 24782889; PubMed Central PMCID: PMC3989762.

[9a]: Robinson JT, Thorvaldsdóttir H, Winckler W, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24-26. doi:10.1038/nbt.1754

[10a]: Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010 Mar 15;26(6):841-2. doi: 10.1093/bioinformatics/btq033.

[11a]: Stark R, Brown G (2011). DiffBind: differential binding analysis of ChIP-Seq peak data. http://bioconductor.org/packages/release/bioc/vignettes/DiffBind/inst/doc/DiffBind.pdf

[12a]: Love MI, Huber W, Anders S (2014). “Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.” Genome Biology, 15, 550. doi: 10.1186/s13059-014-0550-8.

[13a]: Yu G, Wang LG, He QY. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics. 2015 Jul 15;31(14):2382-3. doi: 10.1093/bioinformatics/btv145.

[14a]: Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, Fu X, Liu S, Bo X, Yu G. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Camb). 2021 Jul 1;2(3):100141. doi: 10.1016/j.xinn.2021.100141.

(older)
Reads were aligned with bwa[1] version 0.7.17-r1188 against mm10. Alignments to mitochondria and non-assembled chromosomes, duplicate alignments and multimapping alignments were removed. Alignment files were split into nucleosome (NF) free, mono/di/tri nucleosome free fractions and peaks were called individually with MACS2[2] version 2.2.6 with the parameters `--nomodel -f BAMPE -g 2730871774 --bdg --nolambda`. Consensus peaks were called on NF regions by expanding each summit 250 bases and choosing among overlapping peaks the peak with the highest score (see https://bedops.readthedocs.io/en/latest/content/usage-examples/master-list.html). Counts per peak per sample were called using featureCounts with the consensus peak file using the NF fraction BAM file. Differential peaks were called with DESeq2[3], fitting a model of the form `~0 + sg` where sg is a combined sex-genotype factor. Differential expression of the various comparisons were made with contrasts of the sex-genotype factor, using a BH adjusted p-value cutoff of 0.1. Peaks were annotated for genomic context using the ChIPseeker[4] package.

[1]: Li H. (2013) Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv:1303.3997v2 [q-bio.GN].  whole BWA package)
[2]: Zhang et al. Model-based Analysis of ChIP-Seq (MACS). Genome Biol (2008) vol. 9 (9) pp. R137
[3]: Love MI, Huber W, Anders S (2014). “Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.” Genome Biology, 15, 550. doi: 10.1186/s13059-014-0550-8.
[4]: Yu G, Wang L, He Q (2015). “ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization.” Bioinformatics, 31(14), 2382-2383. doi: 10.1093/bioinformatics/btv145

Mass spectrometry based

Metabolomics

Our analysis strategy for the metabolomic data generally follows the methods implemented in MetaboAnalyst [1], but has the advantage of versatility for customized experimental design. Metabolites with constant values across samples and with more than 80% values missing will be removed before further analysis. Remaining missing values will be imputed by k-means imputation using the impute package in R [2]. A median normalization will be performed for each sample and the resulting data log-transformed. The data will then be examined via Principal Component Analysis (PCA) and hierarchically clustering to identify outliers and potential confounding factors. Finally, differentially abundant metabolites will be identified using Limma [3], which allows for complex customized comparisons.

  1. Chong, J., Soufan, O., Li, C., Caraus, I., Li, S., Bourque, G., Wishart, D.S. and Xia, J. (2018) MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucl. Acids Res. 46, W486-494.
  2. Trevor Hastie, Robert Tibshirani, Balasubramanian Narasimhan and Gilbert Chu (2019). impute: impute: Imputation for microarray data. R package version 1.60.0.
  3. Ritchie, M.E., Phipson, B., Wu, D., Hu, Y., Law, C.W., Shi, W., and Smyth, G.K. (2015). limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research 43(7), e47.