Skip to main content

Advertisement

Log in

Chromatin’s thread to alternative splicing regulation

  • Review
  • Published:
Chromosoma Aims and scope Submit manuscript

Abstract

Intron removal (pre-mRNA splicing) is a necessary step for expression of most genes in higher eukaryotes. Alternative splice site selection is a prevalent mechanism that diversifies genome outputs and offers ample opportunities for gene regulation in these organisms. Pre-mRNA splicing occurs co-transcriptionally and is influenced by features in chromatin structure, including nucleosome density and epigenetic modifications. We review here the molecular mechanisms by which the reciprocal interplay between chromatin and RNA processing can contribute to alternative splicing regulation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  • Alexander RD, Innocente SA, Barrass JD, Beggs JD (2010) Splicing-dependent RNA polymerase pausing in yeast. Mol Cell 40:582–593

    Article  PubMed  CAS  Google Scholar 

  • Allo M, Buggiano V, Fededa JP, Petrillo E, Schor I, de la Mata M, Agirre E, Plass M, Eyras E, Elela SA et al (2009) Control of alternative splicing through siRNA-mediated transcriptional gene silencing. Nat Struct Mol Biol 16:717–724

    Article  PubMed  CAS  Google Scholar 

  • Ameur A, Zaghlool A, Halvardson J, Wetterbom A, Gyllensten U, Cavelier L, Feuk L (2011) Total RNA sequencing reveals nascent transcription and widespread co-transcriptional splicing in the human brain. Nat Struct Mol Biol 18:1435–1440

    Article  PubMed  CAS  Google Scholar 

  • Ameyar-Zazoua M, Rachez C, Souidi M, Robin P, Fritsch L, Young R, Morozova N, Fenouil R, Descostes N, Andrau JC et al (2012) Argonaute proteins couple chromatin silencing to alternative splicing. Nat Struct Mol Biol 19:998–1004

    Article  PubMed  CAS  Google Scholar 

  • Andersson R, Enroth S, Rada-Iglesias A, Wadelius C, Komorowski J (2009) Nucleosomes are well positioned in exons and carry characteristic histone modifications. Genome Res 19:1732–1741

    Article  PubMed  CAS  Google Scholar 

  • Barash Y, Calarco JA, Gao W, Pan Q, Wang X, Shai O, Blencowe BJ, Frey BJ (2010) Deciphering the splicing code. Nature 465:53–59

    Article  PubMed  CAS  Google Scholar 

  • Barboric M, Lenasi T, Chen H, Johansen EB, Guo S, Peterlin BM (2009) 7SK snRNP/P-TEFb couples transcription elongation with alternative splicing and is essential for vertebrate development. Proc Natl Acad Sci U S A 106:7798–7803

    Article  PubMed  CAS  Google Scholar 

  • Batsche E, Yaniv M, Muchardt C (2006) The human SWI/SNF subunit Brm is a regulator of alternative splicing. Nat Struct Mol Biol 13:22–29

    Article  PubMed  CAS  Google Scholar 

  • Beckmann JS, Trifonov EN (1991) Splice junctions follow a 205-base ladder. Proc Natl Acad Sci U S A 88:2380–2383

    Article  PubMed  CAS  Google Scholar 

  • Beyer AL, Osheim YN (1988) Splice site selection, rate of splicing, and alternative splicing on nascent transcripts. Genes Dev 2:754–765

    Article  PubMed  CAS  Google Scholar 

  • Bieberstein NI, Carrillo Oesterreich F, Straube K, Neugebauer KM (2012) First exon length controls active chromatin signatures and transcription. Cell Rep 2:62–68

    Article  PubMed  CAS  Google Scholar 

  • Bonnal S, Vigevani L, Valcarcel J (2012) The spliceosome as a target of novel antitumour drugs. Nat Rev Drug Discov 11:847–859

    Article  PubMed  CAS  Google Scholar 

  • Braunschweig U, Gueroussov S, Plocik AM, Graveley BR, Blencowe BJ (2013) Dynamic integration of splicing within gene regulatory pathways. Cell 152:1252–1269

    Article  PubMed  CAS  Google Scholar 

  • Brody Y, Neufeld N, Bieberstein N, Causse SZ, Bohnlein EM, Neugebauer KM, Darzacq X, Shav-Tal Y (2011) The in vivo kinetics of RNA polymerase II elongation during co-transcriptional splicing. PLoS Biol 9:e1000573

    Article  PubMed  CAS  Google Scholar 

  • Carrillo Oesterreich F, Preibisch S, Neugebauer KM (2010) Global analysis of nascent RNA reveals transcriptional pausing in terminal exons. Mol Cell 40:571–581

    Article  PubMed  CAS  Google Scholar 

  • Chen L, Xiao S, Manley NR (2009) Foxn1 is required to maintain the postnatal thymic microenvironment in a dosage-sensitive manner. Blood 113:567–574

    Article  PubMed  CAS  Google Scholar 

  • Chodavarapu RK, Feng S, Bernatavichute YV, Chen PY, Stroud H, Yu Y, Hetzel JA, Kuo F, Kim J, Cokus SJ et al (2010) Relationship between nucleosome positioning and DNA methylation. Nature 466:388–392

    Article  PubMed  CAS  Google Scholar 

  • Churchman LS, Weissman JS (2011) Nascent transcript sequencing visualizes transcription at nucleotide resolution. Nature 469:368–373

    Article  PubMed  CAS  Google Scholar 

  • Close P, East P, Dirac-Svejstrup AB, Hartmann H, Heron M, Maslen S, Chariot A, Soding J, Skehel M, Svejstrup JQ (2012) DBIRD complex integrates alternative mRNA splicing with RNA polymerase II transcript elongation. Nature 484:386–389

    Article  PubMed  CAS  Google Scholar 

  • Darnell JE Jr (2013) Reflections on the history of pre-mRNA processing and highlights of current knowledge: a unified picture. RNA 19:443–460

    Article  PubMed  CAS  Google Scholar 

  • de Almeida SF, Carmo-Fonseca M (2012) Design principles of interconnections between chromatin and pre-mRNA splicing. Trends Biochem Sci 37:248–253

    Article  PubMed  Google Scholar 

  • de Almeida SF, Grosso AR, Koch F, Fenouil R, Carvalho S, Andrade J, Levezinho H, Gut M, Eick D, Gut I et al (2011) Splicing enhances recruitment of methyltransferase HYPB/Setd2 and methylation of histone H3 Lys36. Nat Struct Mol Biol 18:977–983

    Article  PubMed  Google Scholar 

  • de la Mata M, Alonso CR, Kadener S, Fededa JP, Blaustein MA, Pelisch F, Cramer P, Bentley D, Kornblihtt AR (2003) A slow RNA polymerase II affects alternative splicing in vivo. Mol Cell 12:525–532

    Article  PubMed  Google Scholar 

  • de la Mata M, Lafaille C, Kornblihtt AR (2010) First come, first served revisited: factors affecting the same alternative splicing event have different effects on the relative rates of intron removal. RNA 16:904–912

    Article  PubMed  Google Scholar 

  • Dekker J, Marti-Renom MA, Mirny LA (2013) Exploring the three-dimensional organization of genomes: interpreting chromatin interaction data. Nat Rev Genet 14:390–403

    Google Scholar 

  • Dhami P, Saffrey P, Bruce AW, Dillon SC, Chiang K, Bonhoure N, Koch CM, Bye J, James K, Foad NS et al (2010) Complex exon–intron marking by histone modifications is not determined solely by nucleosome distribution. PLoS One 5:e12339

    Article  PubMed  Google Scholar 

  • Dye MJ, Gromak N, Proudfoot NJ (2006) Exon tethering in transcription by RNA polymerase II. Mol Cell 21:849–859

    Article  PubMed  CAS  Google Scholar 

  • Filipowicz W, Jaskiewicz L, Kolb FA, Pillai RS (2005) Post-transcriptional gene silencing by siRNAs and miRNAs. Curr Opin Struct Biol 15:331–341

    Article  PubMed  CAS  Google Scholar 

  • Fong YW, Zhou Q (2001) Stimulatory effect of splicing factors on transcriptional elongation. Nature 414:929–933

    Article  PubMed  CAS  Google Scholar 

  • Gelfman S, Cohen N, Yearim A, Ast G (2013) DNA-methylation effect on co-transcriptional splicing is dependent on GC-architecture of the exon–intron structure. Genome Res 23(5):789–799

    Article  PubMed  CAS  Google Scholar 

  • Girard C, Will CL, Peng J, Makarov EM, Kastner B, Lemm I, Urlaub H, Hartmuth K, Luhrmann R (2012) Post-transcriptional spliceosomes are retained in nuclear speckles until splicing completion. Nat Commun 3:994

    Article  PubMed  Google Scholar 

  • Gunderson FQ, Johnson TL (2009) Acetylation by the transcriptional coactivator Gcn5 plays a novel role in co-transcriptional spliceosome assembly. PLoS Genet 5:e1000682

    Article  PubMed  Google Scholar 

  • Gunderson FQ, Merkhofer EC, Johnson TL (2011) Dynamic histone acetylation is critical for cotranscriptional spliceosome assembly and spliceosomal rearrangements. Proc Natl Acad Sci U S A 108:2004–2009

    Article  PubMed  CAS  Google Scholar 

  • Hnilicova J, Hozeifi S, Duskova E, Icha J, Tomankova T, Stanek D (2011) Histone deacetylase activity modulates alternative splicing. PLoS One 6:e16727

    Article  PubMed  CAS  Google Scholar 

  • Hodges E, Smith AD, Kendall J, Xuan Z, Ravi K, Rooks M, Zhang MQ, Ye K, Bhattacharjee A, Brizuela L et al (2009) High definition profiling of mammalian DNA methylation by array capture and single molecule bisulfite sequencing. Genome Res 19:1593–1605

    Article  PubMed  CAS  Google Scholar 

  • Howe KJ, Kane CM, Ares M Jr (2003) Perturbation of transcription elongation influences the fidelity of internal exon inclusion in Saccharomyces cerevisiae. RNA 2003(8):993–1006

    Article  Google Scholar 

  • Ip JY, Schmidt D, Pan Q, Ramani AK, Fraser AG, Odom DT, Blencowe BJ (2011) Global impact of RNA polymerase II elongation inhibition on alternative splicing regulation. Genome Res 21:390–401

    Article  PubMed  CAS  Google Scholar 

  • Ji X, Zhou Y, Pandit S, Huang J, Li H, Lin CY, Xiao R, Burge CB, Fu XD (2013) SR proteins collaborate with 7SK and promoter-associated nascent RNA to release paused polymerase. Cell 153:855–868

    Article  PubMed  CAS  Google Scholar 

  • Kelemen O, Convertini P, Zhang Z, Wen Y, Shen M, Falaleeva M, Stamm S (2013) Function of alternative splicing. Gene 514:1–30

    Article  PubMed  CAS  Google Scholar 

  • Keren-Shaul H, Lev-Maor G, Ast G (2013) Pre-mRNA splicing is a determinant of nucleosome organization. PLoS One 8:e53506

    Article  PubMed  CAS  Google Scholar 

  • Khare T, Pai S, Koncevicius K, Pal M, Kriukiene E, Liutkeviciute Z, Irimia M, Jia P, Ptak C, Xia M et al (2012) 5-hmC in the brain is abundant in synaptic genes and shows differences at the exon–intron boundary. Nat Struct Mol Biol 19:1037–1043

    Article  PubMed  CAS  Google Scholar 

  • Khodor YL, Rodriguez J, Abruzzi KC, Tang CH, Marr MT 2nd, Rosbash M (2011) Nascent-seq indicates widespread cotranscriptional pre-mRNA splicing in Drosophila. Genes Dev 25:2502–2512

    Article  PubMed  CAS  Google Scholar 

  • Khodor YL, Menet JS, Tolan M, Rosbash M (2012) Cotranscriptional splicing efficiency differs dramatically between Drosophila and mouse. RNA 18:2174–2186

    Article  PubMed  CAS  Google Scholar 

  • Kim S, Kim H, Fong N, Erickson B, Bentley DL (2011) Pre-mRNA splicing is a determinant of histone H3K36 methylation. Proc Natl Acad Sci U S A 108:13564–13569

    Article  PubMed  CAS  Google Scholar 

  • Kolasinska-Zwierz P, Down T, Latorre I, Liu T, Liu XS, Ahringer J (2009) Differential chromatin marking of introns and expressed exons by H3K36me3. Nat Genet 41:376–381

    Article  PubMed  CAS  Google Scholar 

  • Kornblihtt AR (2007) Coupling transcription and alternative splicing. Adv Exp Med Biol 623:175–189

    Article  PubMed  Google Scholar 

  • Kornblihtt AR, Schor IE, Allo M, Dujardin G, Petrillo E, Munoz MJ (2013) Alternative splicing: a pivotal step between eukaryotic transcription and translation. Nat Rev Mol Cell Biol 14:153–165

    Article  PubMed  CAS  Google Scholar 

  • Kouzarides T (2007) Chromatin modifications and their function. Cell 128:693–705

    Article  PubMed  CAS  Google Scholar 

  • Kwak H, Fuda NJ, Core LJ, Lis JT (2013) Precise maps of RNA polymerase reveal how promoters direct initiation and pausing. Science 339:950–953

    Article  PubMed  CAS  Google Scholar 

  • Li B, Carey M, Workman JL (2007) The role of chromatin during transcription. Cell 128:707–719

    Article  PubMed  CAS  Google Scholar 

  • Lin S, Coutinho-Mansfield G, Wang D, Pandit S, Fu XD (2008) The splicing factor SC35 has an active role in transcriptional elongation. Nat Struct Mol Biol 15:819–826

    Article  PubMed  CAS  Google Scholar 

  • Listerman I, Sapra AK, Neugebauer KM (2006) Cotranscriptional coupling of splicing factor recruitment and precursor messenger RNA splicing in mammalian cells. Nat Struct Mol Biol 13:815–822

    Article  PubMed  CAS  Google Scholar 

  • Loomis RJ, Naoe Y, Parker JB, Savic V, Bozovsky MR, Macfarlan T, Manley JL, Chakravarti D (2009) Chromatin binding of SRp20 and ASF/SF2 and dissociation from mitotic chromosomes is modulated by histone H3 serine 10 phosphorylation. Mol Cell 33:450–461

    Article  PubMed  CAS  Google Scholar 

  • Lorincz MC, Dickerson DR, Schmitt M, Groudine M (2004) Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nat Struct Mol Biol 11:1068–1075

    Article  PubMed  CAS  Google Scholar 

  • Luco RF, Pan Q, Tominaga K, Blencowe BJ, Pereira-Smith OM, Misteli T (2010) Regulation of alternative splicing by histone modifications. Science 327:996–1000

    Article  PubMed  CAS  Google Scholar 

  • Luco RF, Allo M, Schor IE, Kornblihtt AR, Misteli T (2011) Epigenetics in alternative pre-mRNA splicing. Cell 144:16–26

    Article  PubMed  CAS  Google Scholar 

  • Lyko F, Foret S, Kucharski R, Wolf S, Falckenhayn C, Maleszka R (2010) The honey bee epigenomes: differential methylation of brain DNA in queens and workers. PLoS Biol 8:e1000506

    Article  PubMed  Google Scholar 

  • Martins SB, Rino J, Carvalho T, Carvalho C, Yoshida M, Klose JM, de Almeida SF, Carmo-Fonseca M (2011) Spliceosome assembly is coupled to RNA polymerase II dynamics at the 3′ end of human genes. Nat Struct Mol Biol 18:1115–1123

    Article  PubMed  CAS  Google Scholar 

  • McCracken S, Fong N, Yankulov K, Ballantyne S, Pan G, Greenblatt J, Patterson SD, Wickens M, Bentley DL (1997) The C-terminal domain of RNA polymerase II couples mRNA processing to transcription. Nature 385:357–361

    Article  PubMed  CAS  Google Scholar 

  • Mercer TR, Edwards SL, Clark MB, Neph SJ, Wang H, Stergachis AB, John S, Sandstrom R, Li G, Sandhu KS, Ruan Y, Nilesen LK, Mattick JS, Stamatoyannopoulos JA (2013) DNase I-hypersensitive exons colocalize with promoters and distal regulatory elements. Nat Genet. doi:10.1038/ng.2677, advanced online publication

    PubMed  Google Scholar 

  • Munoz MJ, Perez Santangelo MS, Paronetto MP, de la Mata M, Pelisch F, Boireau S, Glover-Cutter K, Ben-Dov C, Blaustein M, Lozano JJ et al (2009) DNA damage regulates alternative splicing through inhibition of RNA polymerase II elongation. Cell 137:708–720

    Article  PubMed  CAS  Google Scholar 

  • Munoz MJ, de la Mata M, Kornblihtt AR (2010) The carboxy terminal domain of RNA polymerase II and alternative splicing. Trends Biochem Sci 35:497–504

    Article  PubMed  CAS  Google Scholar 

  • Nahkuri S, Taft RJ, Mattick JS (2009) Nucleosomes are preferentially positioned at exons in somatic and sperm cells. Cell Cycle 8:3420–3424

    Article  PubMed  CAS  Google Scholar 

  • Nogues G, Kadener S, Cramer P, Bentley D, Kornblihtt AR (2002) Transcriptional activators differ in their abilities to control alternative splicing. J Biol Chem 277:43110–43114

    Article  PubMed  CAS  Google Scholar 

  • Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ (2008) Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet 40:1413–1415

    Article  PubMed  CAS  Google Scholar 

  • Pandya-Jones A, Black DL (2009) Co-transcriptional splicing of constitutive and alternative exons. RNA 15:1896–1908

    Article  PubMed  CAS  Google Scholar 

  • Pradeepa MM, Sutherland HG, Ule J, Grimes GR, Bickmore WA (2012) Psip1/Ledgf p52 binds methylated histone H3K36 and splicing factors and contributes to the regulation of alternative splicing. PLoS Genet 8:e1002717

    Article  PubMed  CAS  Google Scholar 

  • Rauch TA, Wu X, Zhong X, Riggs AD, Pfeifer GP (2009) A human B cell methylome at 100-base pair resolution. Proc Natl Acad Sci U S A 106:671–678

    Article  PubMed  CAS  Google Scholar 

  • Saint-Andre V, Batsche E, Rachez C, Muchardt C (2011) Histone H3 lysine 9 trimethylation and HP1gamma favor inclusion of alternative exons. Nat Struct Mol Biol 18:337–344

    Article  PubMed  CAS  Google Scholar 

  • Sanchez G, Bittencourt D, Laud K, Barbier J, Delattre O, Auboeuf D, Dutertre M (2008) Alteration of cyclin D1 transcript elongation by a mutated transcription factor up-regulates the oncogenic D1b splice isoform in cancer. Proc Natl Acad Sci U S A 105:6004–6009

    Article  PubMed  CAS  Google Scholar 

  • Schor IE, Rascovan N, Pelisch F, Allo M, Kornblihtt AR (2009) Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing. Proc Natl Acad Sci U S A 106:4325–4330

    Article  PubMed  CAS  Google Scholar 

  • Schwartz S, Meshorer E, Ast G (2009) Chromatin organization marks exon–intron structure. Nat Struct Mol Biol 16:990–995

    Article  PubMed  CAS  Google Scholar 

  • Shieh GS, Pan CH, Wu JH, Sun YJ, Wang CC, Hsiao WC, Lin CY, Tung L, Chang TH, Fleming AB et al (2011) H2B ubiquitylation is part of chromatin architecture that marks exon–intron structure in budding yeast. BMC genomics 12:627

    Article  PubMed  CAS  Google Scholar 

  • Shukla S, Oberdoerffer S (2012) Co-transcriptional regulation of alternative pre-mRNA splicing. Biochim Biophys Acta 1819:673–683

    Article  PubMed  CAS  Google Scholar 

  • Shukla S, Kavak E, Gregory M, Imashimizu M, Shutinoski B, Kashlev M, Oberdoerffer P, Sandberg R, Oberdoerffer S (2011) CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature 479:74–79

    Article  PubMed  CAS  Google Scholar 

  • Sims RJ 3rd, Millhouse S, Chen CF, Lewis BA, Erdjument-Bromage H, Tempst P, Manley JL, Reinberg D (2007) Recognition of trimethylated histone H3 lysine 4 facilitates the recruitment of transcription postinitiation factors and pre-mRNA splicing. Mol Cell 28:665–676

    Article  PubMed  CAS  Google Scholar 

  • Smallwood A, Hon GC, Jin F, Henry RE, Espinosa JM, Ren B (2012) CBX3 regulates efficient RNA processing genome-wide. Genome Res 22:1426–1436

    Article  PubMed  CAS  Google Scholar 

  • Spies N, Nielsen CB, Padgett RA, Burge CB (2009) Biased chromatin signatures around polyadenylation sites and exons. Mol Cell 36:245–254

    Article  PubMed  CAS  Google Scholar 

  • Taliaferro JM, Aspden JL, Bradley T, Marwha D, Blanchette M, Rio DC (2013) Two new and distinct roles for Drosophila Argonaute-2 in the nucleus: alternative pre-mRNA splicing and transcriptional repression. Genes Dev 27:378–389

    Article  PubMed  CAS  Google Scholar 

  • Tilgner H, Nikolaou C, Althammer S, Sammeth M, Beato M, Valcarcel J, Guigo R (2009) Nucleosome positioning as a determinant of exon recognition. Nat Struct Mol Biol 16:996–1001

    Article  PubMed  CAS  Google Scholar 

  • Tilgner H, Knowles DG, Johnson R, Davis CA, Chakrabortty S, Djebali S, Curado J, Snyder M, Gingeras TR, Guigo R (2012) Deep sequencing of subcellular RNA fractions shows splicing to be predominantly co-transcriptional in the human genome but inefficient for lncRNAs. Genome Res 22:1616–1625

    Article  PubMed  CAS  Google Scholar 

  • Vargas DY, Shah K, Batish M, Levandoski M, Sinha S, Marras SA, Schedl P, Tyagi S (2011) Single-molecule imaging of transcriptionally coupled and uncoupled splicing. Cell 147:1054–1065

    Article  PubMed  CAS  Google Scholar 

  • Wahl MC, Will CL, Luhrmann R (2009) The spliceosome: design principles of a dynamic RNP machine. Cell 136:701–718

    Article  PubMed  CAS  Google Scholar 

  • Wang Z, Burge CB (2008) Splicing regulation: from a parts list of regulatory elements to an integrated splicing code. RNA 14:802–813

    Article  PubMed  CAS  Google Scholar 

  • Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB (2008) Alternative isoform regulation in human tissue transcriptomes. Nature 456:470–476

    Article  PubMed  CAS  Google Scholar 

  • Zhong XY, Wang P, Han J, Rosenfeld MG, Fu XD (2009) SR proteins in vertical integration of gene expression from transcription to RNA processing to translation. Mol Cell 35:1–10

    Article  PubMed  CAS  Google Scholar 

  • Zhou B, Yan H, Li Y, Wang R, Chen K, Zhou Z, Sun X (2012) PNAS-4 expression and its relationship to p53 in colorectal cancer. Mol Biol Rep 39:243–249

    Article  PubMed  CAS  Google Scholar 

  • Zhu H, Hasman RA, Barron VA, Luo G, Lou H (2006) A nuclear function of Hu proteins as neuron-specific alternative RNA processing regulators. Mol Biol Cell 17:5105–5114

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We thank Miguel Beato, Luciano Di Croce, Roderic Guigó, Maria Paola Paronetto, and members of our laboratory for discussions and comments on the manuscript. CI was supported by a la Caixa PhD fellowship. Work in our lab is supported by Fundación Botín, Ministerio de Economía y Competitividad, and Consolider RNAREG.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juan Valcárcel.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Iannone, C., Valcárcel, J. Chromatin’s thread to alternative splicing regulation. Chromosoma 122, 465–474 (2013). https://doi.org/10.1007/s00412-013-0425-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00412-013-0425-x

Keywords

Navigation