CTCF

Last updated
CTCF
Protein CTCF PDB 1x6h.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases CTCF , MRD21, CCCTC-binding factor, FAP108, CFAP108
External IDs OMIM: 604167 MGI: 109447 HomoloGene: 4786 GeneCards: CTCF
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001191022
NM_006565
NM_001363916

NM_181322
NM_001358924

RefSeq (protein)

NP_001177951
NP_006556
NP_001350845

Location (UCSC) Chr 16: 67.56 – 67.64 Mb Chr 8: 105.64 – 105.68 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Transcriptional repressor CTCF also known as 11-zinc finger protein or CCCTC-binding factor is a transcription factor that in humans is encoded by the CTCF gene. [5] [6] CTCF is involved in many cellular processes, including transcriptional regulation, insulator activity, V(D)J recombination [7] and regulation of chromatin architecture. [8]

Discovery

CCCTC-Binding factor or CTCF was initially discovered as a negative regulator of the chicken c-myc gene. This protein was found to be binding to three regularly spaced repeats of the core sequence CCCTC and thus was named CCCTC binding factor. [9]

Function

The primary role of CTCF is thought to be in regulating the 3D structure of chromatin. [8] CTCF binds together strands of DNA, thus forming chromatin loops, and anchors DNA to cellular structures like the nuclear lamina. [10] It also defines the boundaries between active and heterochromatic DNA.

Since the 3D structure of DNA influences the regulation of genes, CTCF's activity influences the expression of genes. CTCF is thought to be a primary part of the activity of insulators, sequences that block the interaction between enhancers and promoters. CTCF binding has also been both shown to promote and repress gene expression. It is unknown whether CTCF affects gene expression solely through its looping activity, or if it has some other, unknown, activity. [8] In a recent study, it has been shown that, in addition to demarcating TADs, CTCF mediates promoter–enhancer loops, often located in promoter-proximal regions, to facilitate the promoter–enhancer interactions within one TAD. [11] This is in line with the concept that a subpopulation of CTCF associates with the RNA polymerase II (Pol II) protein complex to activate transcription. It is likely that CTCF helps to bridge the transcription factor-bound enhancers to transcription start site-proximal regulatory elements and to initiate transcription by interacting with Pol II, thus supporting a role of CTCF in facilitating contacts between transcription regulatory sequences. This model has been demonstrated by the previous work on the beta-globin locus.

Observed activity

The binding of CTCF has been shown to have many effects, which are enumerated below. In each case, it is unknown if CTCF directly evokes the outcome or if it does so indirectly (in particular through its looping role).

Transcriptional regulation

The protein CTCF plays a heavy role in repressing the insulin-like growth factor 2 gene, by binding to the H-19 imprinting control region (ICR) along with differentially-methylated region-1 (DMR1) and MAR3. [12] [13]

Insulation

Binding of targeting sequence elements by CTCF can block the interaction between enhancers and promoters, therefore limiting the activity of enhancers to certain functional domains. Besides acting as enhancer blocking, CTCF can also act as a chromatin barrier [14] by preventing the spread of heterochromatin structures.

Regulation of chromatin architecture

CTCF physically binds to itself to form homodimers, [15] which causes the bound DNA to form loops. [16] CTCF also occurs frequently at the boundaries of sections of DNA bound to the nuclear lamina. [10] Using chromatin immuno-precipitation (ChIP) followed by ChIP-seq, it was found that CTCF localizes with cohesin genome-wide and affects gene regulatory mechanisms and the higher-order chromatin structure. [17] [18] It is currently believed that the DNA loops are formed by the "loop extrusion" mechanism, whereby the cohesin ring is actively being translocated along the DNA until it meets CTCF. CTCF has to be in a proper orientation to stop cohesin. [19] [20]

Regulation of RNA splicing

CTCF binding has been shown to influence mRNA splicing. [21]

DNA binding

CTCF binds to the consensus sequence CCGCGNGGNGGCAG (in IUPAC notation). [22] [23] This sequence is defined by 11 zinc finger motifs in its structure. CTCF's binding is disrupted by CpG methylation of the DNA it binds to. [24] On the other hand, CTCF binding may set boundaries for the spreading of DNA methylation. [25] In recent studies, CTCF binding loss is reported to increase localized CpG methylation, which reflected another epigenetic remodeling role of CTCF in human genome. [26] [27] [28]

CTCF binds to an average of about 55,000 DNA sites in 19 diverse cell types (12 normal and 7 immortal) and in total 77,811 distinct binding sites across all 19 cell types. [29] CTCF's ability to bind to multiple sequences through the usage of various combinations of its zinc fingers earned it the status of a “multivalent protein”. [5] More than 30,000 CTCF binding sites have been characterized. [30] The human genome contains anywhere between 15,000 and 40,000 CTCF binding sites depending on cell type, suggesting a widespread role for CTCF in gene regulation. [14] [22] [31] In addition CTCF binding sites act as nucleosome positioning anchors so that, when used to align various genomic signals, multiple flanking nucleosomes can be readily identified. [14] [32] On the other hand, high-resolution nucleosome mapping studies have demonstrated that the differences of CTCF binding between cell types may be attributed to the differences in nucleosome locations. [33] Methylation loss at CTCF-binding site of some genes has been found to be related to human diseases, including male infertility. [23]

Protein-protein interactions

CTCF binds to itself to form homodimers. [15] CTCF has also been shown to interact with Y box binding protein 1. [34] CTCF also co-localizes with cohesin, which extrudes chromatin loops by actively translocating one or two DNA strands through its ring-shaped structure, until it meets CTCF in a proper orientation. [35] CTCF is also known to interact with chromatin remodellers such as Chd4 and Snf2h (SMARCA5). [36]

Related Research Articles

Chromatin is a complex of DNA and protein found in eukaryotic cells. The primary function is to package long DNA molecules into more compact, denser structures. This prevents the strands from becoming tangled and also plays important roles in reinforcing the DNA during cell division, preventing DNA damage, and regulating gene expression and DNA replication. During mitosis and meiosis, chromatin facilitates proper segregation of the chromosomes in anaphase; the characteristic shapes of chromosomes visible during this stage are the result of DNA being coiled into highly condensed chromatin.

<span class="mw-page-title-main">Nucleosome</span> Basic structural unit of DNA packaging in eukaryotes

A nucleosome is the basic structural unit of DNA packaging in eukaryotes. The structure of a nucleosome consists of a segment of DNA wound around eight histone proteins and resembles thread wrapped around a spool. The nucleosome is the fundamental subunit of chromatin. Each nucleosome is composed of a little less than two turns of DNA wrapped around a set of eight proteins called histones, which are known as a histone octamer. Each histone octamer is composed of two copies each of the histone proteins H2A, H2B, H3, and H4.

A regulatory sequence is a segment of a nucleic acid molecule which is capable of increasing or decreasing the expression of specific genes within an organism. Regulation of gene expression is an essential feature of all living organisms and viruses.

In molecular biology and genetics, transcriptional regulation is the means by which a cell regulates the conversion of DNA to RNA (transcription), thereby orchestrating gene activity. A single gene can be regulated in a range of ways, from altering the number of copies of RNA that are transcribed, to the temporal control of when the gene is transcribed. This control allows the cell or organism to respond to a variety of intra- and extracellular signals and thus mount a response. Some examples of this include producing the mRNA that encode enzymes to adapt to a change in a food source, producing the gene products involved in cell cycle specific activities, and producing the gene products responsible for cellular differentiation in multicellular eukaryotes, as studied in evolutionary developmental biology.

An insulator is a type of cis-regulatory element known as a long-range regulatory element. Found in multicellular eukaryotes and working over distances from the promoter element of the target gene, an insulator is typically 300 bp to 2000 bp in length. Insulators contain clustered binding sites for sequence specific DNA-binding proteins and mediate intra- and inter-chromosomal interactions.

Chromatin remodeling is the dynamic modification of chromatin architecture to allow access of condensed genomic DNA to the regulatory transcription machinery proteins, and thereby control gene expression. Such remodeling is principally carried out by 1) covalent histone modifications by specific enzymes, e.g., histone acetyltransferases (HATs), deacetylases, methyltransferases, and kinases, and 2) ATP-dependent chromatin remodeling complexes which either move, eject or restructure nucleosomes. Besides actively regulating gene expression, dynamic remodeling of chromatin imparts an epigenetic regulatory role in several key biological processes, egg cells DNA replication and repair; apoptosis; chromosome segregation as well as development and pluripotency. Aberrations in chromatin remodeling proteins are found to be associated with human diseases, including cancer. Targeting chromatin remodeling pathways is currently evolving as a major therapeutic strategy in the treatment of several cancers.

<span class="mw-page-title-main">CTCFL</span> Protein-coding gene in the species Homo sapiens

Transcriptional repressor CTCFL also known as BORIS is a protein that in humans is encoded by the CTCFL gene.

Epigenomics is the study of the complete set of epigenetic modifications on the genetic material of a cell, known as the epigenome. The field is analogous to genomics and proteomics, which are the study of the genome and proteome of a cell. Epigenetic modifications are reversible modifications on a cell's DNA or histones that affect gene expression without altering the DNA sequence. Epigenomic maintenance is a continuous process and plays an important role in stability of eukaryotic genomes by taking part in crucial biological mechanisms like DNA repair. Plant flavones are said to be inhibiting epigenomic marks that cause cancers. Two of the most characterized epigenetic modifications are DNA methylation and histone modification. Epigenetic modifications play an important role in gene expression and regulation, and are involved in numerous cellular processes such as in differentiation/development and tumorigenesis. The study of epigenetics on a global level has been made possible only recently through the adaptation of genomic high-throughput assays.

H3K4me3 is an epigenetic modification to the DNA packaging protein Histone H3 that indicates tri-methylation at the 4th lysine residue of the histone H3 protein and is often involved in the regulation of gene expression. The name denotes the addition of three methyl groups (trimethylation) to the lysine 4 on the histone H3 protein.

<span class="mw-page-title-main">Nuclear organization</span> Spatial distribution of chromatin within a cell nucleus

Nuclear organization refers to the spatial distribution of chromatin within a cell nucleus. There are many different levels and scales of nuclear organisation. Chromatin is a higher order structure of DNA.

<span class="mw-page-title-main">Insulated neighborhood</span>

In mammalian biology, insulated neighborhoods are chromosomal loop structures formed by the physical interaction of two DNA loci bound by the transcription factor CTCF and co-occupied by cohesin. Insulated neighborhoods are thought to be structural and functional units of gene control because their integrity is important for normal gene regulation. Current evidence suggests that these structures form the mechanistic underpinnings of higher-order chromosome structures, including topologically associating domains (TADs). Insulated neighborhoods are functionally important in understanding gene regulation in normal cells and dysregulated gene expression in disease.

H3K9me3 is an epigenetic modification to the DNA packaging protein Histone H3. It is a mark that indicates the tri-methylation at the 9th lysine residue of the histone H3 protein and is often associated with heterochromatin.

Human epigenome is the complete set of structural modifications of chromatin and chemical modifications of histones and nucleotides. These modifications affect according to cellular type and development status. Various studies show that epigenome depends on exogenous factors.

H4K20me is an epigenetic modification to the DNA packaging protein Histone H4. It is a mark that indicates the mono-methylation at the 20th lysine residue of the histone H4 protein. This mark can be di- and tri-methylated. It is critical for genome integrity including DNA damage repair, DNA replication and chromatin compaction.

H4K16ac is an epigenetic modification to the DNA packaging protein Histone H4. It is a mark that indicates the acetylation at the 16th lysine residue of the histone H4 protein.

H4K12ac is an epigenetic modification to the DNA packaging protein histone H4. It is a mark that indicates the acetylation at the 12th lysine residue of the histone H4 protein. H4K12ac is involved in learning and memory. It is possible that restoring this modification could reduce age-related decline in memory.

H3K14ac is an epigenetic modification to the DNA packaging protein Histone H3. It is a mark that indicates the acetylation at the 14th lysine residue of the histone H3 protein.

H3R17me2 is an epigenetic modification to the DNA packaging protein histone H3. It is a mark that indicates the di-methylation at the 17th arginine residue of the histone H3 protein. In epigenetics, arginine methylation of histones H3 and H4 is associated with a more accessible chromatin structure and thus higher levels of transcription. The existence of arginine demethylases that could reverse arginine methylation is controversial.

H3R8me2 is an epigenetic modification to the DNA packaging protein histone H3. It is a mark that indicates the di-methylation at the 8th arginine residue of the histone H3 protein. In epigenetics, arginine methylation of histones H3 and H4 is associated with a more accessible chromatin structure and thus higher levels of transcription. The existence of arginine demethylases that could reverse arginine methylation is controversial.

H3R2me2 is an epigenetic modification to the DNA packaging protein histone H3. It is a mark that indicates the di-methylation at the 2nd arginine residue of the histone H3 protein. In epigenetics, arginine methylation of histones H3 and H4 is associated with a more accessible chromatin structure and thus higher levels of transcription. The existence of arginine demethylases that could reverse arginine methylation is controversial.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000102974 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000005698 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 Filippova GN, Fagerlie S, Klenova EM, Myers C, Dehner Y, Goodwin G, Neiman PE, Collins SJ, Lobanenkov VV (June 1996). "An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes". Mol. Cell. Biol. 16 (6): 2802–13. doi:10.1128/mcb.16.6.2802. PMC   231272 . PMID   8649389.
  6. Rubio ED, Reiss DJ, Welcsh PL, Disteche CM, Filippova GN, Baliga NS, Aebersold R, Ranish JA, Krumm A (June 2008). "CTCF physically links cohesin to chromatin". Proc. Natl. Acad. Sci. U.S.A. 105 (24): 8309–14. Bibcode:2008PNAS..105.8309R. doi: 10.1073/pnas.0801273105 . PMC   2448833 . PMID   18550811.
  7. Chaumeil J, Skok JA (April 2012). "The role of CTCF in regulating V(D)J recombination". Curr. Opin. Immunol. 24 (2): 153–9. doi:10.1016/j.coi.2012.01.003. PMC   3444155 . PMID   22424610.
  8. 1 2 3 Phillips JE; Corces VG (June 2009). "CTCF: master weaver of the genome". Cell. 137 (7): 1194–211. doi:10.1016/j.cell.2009.06.001. PMC   3040116 . PMID   19563753.
  9. Lobanenkov VV, Nicolas RH, Adler VV, Paterson H, Klenova EM, Polotskaja AV, Goodwin GH (December 1990). "A novel sequence-specific DNA binding protein which interacts with three regularly spaced direct repeats of the CCCTC-motif in the 5'-flanking sequence of the chicken c-myc gene". Oncogene. 5 (12): 1743–53. PMID   2284094.
  10. 1 2 Guelen L, Pagie L, Brasset E, Meuleman W, Faza MB, Talhout W, Eussen BH, de Klein A, Wessels L, de Laat W, van Steensel B (June 2008). "Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions". Nature. 453 (7197): 948–51. Bibcode:2008Natur.453..948G. doi:10.1038/nature06947. PMID   18463634. S2CID   4429401.
  11. Qu J, Yi G, Zhou H (June 2019). "p63 cooperates with CTCF to modulate chromatin architecture in skin keratinocytes". Epigenetics & Chromatin. 12 (1): 31. doi: 10.1186/s13072-019-0280-y . PMC   6547520 . PMID   31164150.
  12. Ohlsson R, Renkawitz R, Lobanenkov V (2001). "CTCF is a uniquely versatile transcription regulator linked to epigenetics and disease". Trends Genet. 17 (9): 520–7. doi:10.1016/S0168-9525(01)02366-6. PMID   11525835.
  13. Dunn KL, Davie JR (2003). "The many roles of the transcriptional regulator CTCF". Biochem. Cell Biol. 81 (3): 161–7. doi:10.1139/o03-052. PMID   12897849.
  14. 1 2 3 Cuddapah S, Jothi R, Schones DE, Roh TY, Cui K, Zhao K (2009). "Global analysis of the insulator binding protein CTCF in chromatin barrier regions reveals demarcation of active and repressive domains". Genome Res. 19 (1): 24–32. doi:10.1101/gr.082800.108. PMC   2612964 . PMID   19056695.
  15. 1 2 Yusufzai TM, Tagami H, Nakatani Y, Felsenfeld G (January 2004). "CTCF tethers an insulator to subnuclear sites, suggesting shared insulator mechanisms across species". Mol. Cell. 13 (2): 291–8. doi: 10.1016/S1097-2765(04)00029-2 . PMID   14759373.
  16. Hou C, Zhao H, Tanimoto K, Dean A (December 2008). "CTCF-dependent enhancer-blocking by alternative chromatin loop formation". Proc. Natl. Acad. Sci. U.S.A. 105 (51): 20398–403. Bibcode:2008PNAS..10520398H. doi: 10.1073/pnas.0808506106 . PMC   2629272 . PMID   19074263.
  17. Sofueva S, Yaffe E, Chan WC, Georgopoulou D, Vietri Rudan M, Mira-Bontenbal H, et al. (December 2013). "Cohesin-mediated interactions organize chromosomal domain architecture". The EMBO Journal. 32 (24): 3119–3129. doi:10.1038/emboj.2013.237. PMC   4489921 . PMID   24185899.
  18. Lee BK, Iyer VR (September 2012). "Genome-wide studies of CCCTC-binding factor (CTCF) and cohesin provide insight into chromatin structure and regulation". J. Biol. Chem. 287 (37): 30906–13. doi: 10.1074/jbc.R111.324962 . PMC   3438923 . PMID   22952237.
  19. Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. (December 2014). "A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping". Cell. 159 (7): 1665–1680. doi:10.1016/j.cell.2014.11.021. PMC   5635824 . PMID   25497547.
  20. Vietri Rudan M, Barrington C, Henderson S, Ernst C, Odom DT, Tanay A, Hadjur S (March 2015). "Comparative Hi-C reveals that CTCF underlies evolution of chromosomal domain architecture". Cell Reports. 10 (8): 1297–1309. doi:10.1016/j.celrep.2015.02.004. PMC   4542312 . PMID   25732821.
  21. Shukla S, Kavak E, Gregory M, Imashimizu M, Shutinoski B, Kashlev M, Oberdoerffer P, Sandberg R, Oberdoerffer S (November 2011). "CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing". Nature. 479 (7371): 74–9. Bibcode:2011Natur.479...74S. doi:10.1038/nature10442. PMC   7398428 . PMID   21964334.
  22. 1 2 Kim TH, Abdullaev ZK, Smith AD, Ching KA, Loukinov DI, Green RD, Zhang MQ, Lobanenkov VV, Ren B (March 2007). "Analysis of the vertebrate insulator protein CTCF-binding sites in the human genome". Cell. 128 (6): 1231–45. doi:10.1016/j.cell.2006.12.048. PMC   2572726 . PMID   17382889.
  23. 1 2 Rotondo JC, Selvatici R, Di Domenico M, Marci R, Vesce F, Tognon M, Martini F (September 2013). "Methylation loss at H19 imprinted gene correlates with methylenetetrahydrofolate reductase gene promoter hypermethylation in semen samples from infertile males". Epigenetics. 8 (9): 990–7. doi:10.4161/epi.25798. PMC   3883776 . PMID   23975186.
  24. Bell AC, Felsenfeld G (May 2000). "Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene". Nature. 405 (6785): 482–5. Bibcode:2000Natur.405..482B. doi:10.1038/35013100. PMID   10839546. S2CID   4387329.
  25. Wiehle L, Thorn GJ, Raddatz G, Clarkson CT, Rippe K, Lyko F, Breiling A, Teif VB (May 2019). "DNA de-methylation in embryonic stem cells controls CTCF-dependent chromatin boundaries". Genome Research. 29 (5): 750–61. doi:10.1101/gr.239707.118. PMC   6499307 . PMID   30948436.
  26. Tian Y, Soupir A, Liu Q, Wu L, Huang CC, Park JY, Wang L (May 2022). "Novel role of prostate cancer risk variant rs7247241 on PPP1R14A isoform transition through allelic TF binding and CpG methylation". Human Molecular Genetics. 31 (10): 1610–1621. doi:10.1093/hmg/ddab347. PMC   9122641 . PMID   34849858.
  27. Damaschke NA, Gawdzik J, Avilla M, Yang B, Svaren J, Roopra A, et al. (June 2020). "CTCF loss mediates unique DNA hypermethylation landscapes in human cancers". Clinical Epigenetics. 12 (1): 80. doi: 10.1186/s13148-020-00869-7 . PMC   7275597 . PMID   32503656.
  28. Kemp CJ, Moore JM, Moser R, Bernard B, Teater M, Smith LE, et al. (May 2014). "CTCF haploinsufficiency destabilizes DNA methylation and predisposes to cancer". Cell Reports. 7 (4): 1020–1029. doi:10.1016/j.celrep.2014.04.004. PMC   4040130 . PMID   24794443.
  29. Wang H, Maurano MT, Qu H, Varley KE, Gertz J, Pauli F, Lee K, Canfield T, Weaver M, Sandstrom R, Thurman RE, Kaul R, Myers RM, Stamatoyannopoulos JA (September 2012). "Widespread plasticity in CTCF occupancy linked to DNA methylation". Genome Res. 22 (9): 1680–8. doi:10.1101/gr.136101.111. PMC   3431485 . PMID   22955980.
  30. Bao L, Zhou M, Cui Y (January 2008). "CTCFBSDB: a CTCF-binding site database for characterization of vertebrate genomic insulators". Nucleic Acids Res. 36 (Database issue): D83–7. doi:10.1093/nar/gkm875. PMC   2238977 . PMID   17981843.
  31. Xie X, Mikkelsen TS, Gnirke A, Lindblad-Toh K, Kellis M, Lander ES (2007). "Systematic discovery of regulatory motifs in conserved regions of the human genome, including thousands of CTCF insulator sites". Proc. Natl. Acad. Sci. U.S.A. 104 (17): 7145–50. Bibcode:2007PNAS..104.7145X. doi: 10.1073/pnas.0701811104 . PMC   1852749 . PMID   17442748.
  32. Fu Y, Sinha M, Peterson CL, Weng Z (2008). "The insulator binding protein CTCF positions 20 nucleosomes around its binding sites across the human genome". PLOS Genetics. 4 (7): e1000138. doi:10.1371/journal.pgen.1000138. PMC   2453330 . PMID   18654629.
  33. Teif VB, Vainshtein Y, Caudron-Herger M, Mallm JP, Marth C, Höfer T, Rippe K (2012). "Genome-wide nucleosome positioning during embryonic stem cell development". Nat Struct Mol Biol. 19 (11): 1185–92. doi:10.1038/nsmb.2419. PMID   23085715. S2CID   34509771.
  34. Chernukhin IV, Shamsuddin S, Robinson AF, Carne AF, Paul A, El-Kady AI, Lobanenkov VV, Klenova EM (September 2000). "Physical and functional interaction between two pluripotent proteins, the Y-box DNA/RNA-binding factor, YB-1, and the multivalent zinc finger factor, CTCF". J. Biol. Chem. 275 (38): 29915–21. doi:10.1074/jbc.M001538200. PMID   10906122.
  35. Kagey MH; Newman JJ; Bilodeau S; Zhan Y; Orlando DA; van Berkum NL; Ebmeier CC; Goossens J; Rahl PB; Levine SS; Taatjes DJ; Dekker J; Young RA (September 2010). "Mediator and cohesin connect gene expression and chromatin architecture". Nature. 467 (7314): 430–5. Bibcode:2010Natur.467..430K. doi:10.1038/nature09380. PMC   2953795 . PMID   20720539.
  36. Clarkson CT, Deeks EA, Samarista R, Mamayusupova H, Zhurkin VB, Teif VB (September 2019). "CTCF-dependent chromatin boundaries formed by asymmetric nucleosome arrays with decreased linker length". Nucleic Acids Research. 47 (21): 11181–11196. doi:10.1093/nar/gkz908. PMC   6868436 . PMID   31665434.

Further reading