fbpx
Wikipedia

Nucleosome

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[1] 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.

Basic units of chromatin structure

DNA must be compacted into nucleosomes to fit within the cell nucleus.[2] In addition to nucleosome wrapping, eukaryotic chromatin is further compacted by being folded into a series of more complex structures, eventually forming a chromosome. Each human cell contains about 30 million nucleosomes.[3]

Nucleosomes are thought to carry epigenetically inherited information in the form of covalent modifications of their core histones. Nucleosome positions in the genome are not random, and it is important to know where each nucleosome is located because this determines the accessibility of the DNA to regulatory proteins.[4]

Nucleosomes were first observed as particles in the electron microscope by Don and Ada Olins in 1974,[5] and their existence and structure (as histone octamers surrounded by approximately 200 base pairs of DNA) were proposed by Roger Kornberg.[6][7] The role of the nucleosome as a regulator of transcription was demonstrated by Lorch et al. in vitro[8] in 1987 and by Han and Grunstein[9] and Clark-Adams et al.[10] in vivo in 1988.

The nucleosome core particle consists of approximately 146 base pairs (bp) of DNA[11] wrapped in 1.67 left-handed superhelical turns around a histone octamer, consisting of 2 copies each of the core histones H2A, H2B, H3, and H4.[12] Core particles are connected by stretches of linker DNA, which can be up to about 80 bp long. Technically, a nucleosome is defined as the core particle plus one of these linker regions; however the word is often synonymous with the core particle.[13] Genome-wide nucleosome positioning maps are now available for many model organisms and human cells.[14]

Linker histones such as H1 and its isoforms are involved in chromatin compaction and sit at the base of the nucleosome near the DNA entry and exit binding to the linker region of the DNA.[15] Non-condensed nucleosomes without the linker histone resemble "beads on a string of DNA" under an electron microscope.[16]

In contrast to most eukaryotic cells, mature sperm cells largely use protamines to package their genomic DNA, most likely to achieve an even higher packaging ratio.[17] Histone equivalents and a simplified chromatin structure have also been found in Archaea,[18] suggesting that eukaryotes are not the only organisms that use nucleosomes.

Structure edit

Structure of the core particle edit

 
The crystal structure of the nucleosome core particle consisting of H2A , H2B , H3 and H4 core histones, and DNA. The view is from the top through the superhelical axis.

Overview edit

Pioneering structural studies in the 1980s by Aaron Klug's group provided the first evidence that an octamer of histone proteins wraps DNA around itself in about 1.7 turns of a left-handed superhelix.[19] In 1997 the first near atomic resolution crystal structure of the nucleosome was solved by the Richmond group, showing the most important details of the particle. The human alpha satellite palindromic DNA critical to achieving the 1997 nucleosome crystal structure was developed by the Bunick group at Oak Ridge National Laboratory in Tennessee.[20][21][22][23][24] The structures of over 20 different nucleosome core particles have been solved to date,[25] including those containing histone variants and histones from different species. The structure of the nucleosome core particle is remarkably conserved, and even a change of over 100 residues between frog and yeast histones results in electron density maps with an overall root mean square deviation of only 1.6Å.[26]

The nucleosome core particle (NCP) edit

The nucleosome core particle (shown in the figure) consists of about 146 base pair of DNA[11] wrapped in 1.67 left-handed superhelical turns around the histone octamer, consisting of 2 copies each of the core histones H2A, H2B, H3, and H4. Adjacent nucleosomes are joined by a stretch of free DNA termed linker DNA (which varies from 10 - 80 bp in length depending on species and tissue type[18]).The whole structure generates a cylinder of diameter 11 nm and a height of 5.5 nm.

 
Apoptotic DNA laddering. Digested chromatin is in the first lane; the second contains DNA standard to compare lengths.
 
Scheme of nucleosome organization.[27]
 
The crystal structure of the nucleosome core particle (PDB: 1EQZ[28])

Nucleosome core particles are observed when chromatin in interphase is treated to cause the chromatin to unfold partially. The resulting image, via an electron microscope, is "beads on a string". The string is the DNA, while each bead in the nucleosome is a core particle. The nucleosome core particle is composed of DNA and histone proteins.[29]

Partial DNAse digestion of chromatin reveals its nucleosome structure. Because DNA portions of nucleosome core particles are less accessible for DNAse than linking sections, DNA gets digested into fragments of lengths equal to multiplicity of distance between nucleosomes (180, 360, 540 base pairs etc.). Hence a very characteristic pattern similar to a ladder is visible during gel electrophoresis of that DNA.[27] Such digestion can occur also under natural conditions during apoptosis ("cell suicide" or programmed cell death), because autodestruction of DNA typically is its role.[30]

Protein interactions within the nucleosome edit

The core histone proteins contains a characteristic structural motif termed the "histone fold", which consists of three alpha-helices (α1-3) separated by two loops (L1-2). In solution, the histones form H2A-H2B heterodimers and H3-H4 heterotetramers. Histones dimerise about their long α2 helices in an anti-parallel orientation, and, in the case of H3 and H4, two such dimers form a 4-helix bundle stabilised by extensive H3-H3' interaction. The H2A/H2B dimer binds onto the H3/H4 tetramer due to interactions between H4 and H2B, which include the formation of a hydrophobic cluster.[12] The histone octamer is formed by a central H3/H4 tetramer sandwiched between two H2A/H2B dimers. Due to the highly basic charge of all four core histones, the histone octamer is stable only in the presence of DNA or very high salt concentrations.

Histone - DNA interactions edit

The nucleosome contains over 120 direct protein-DNA interactions and several hundred water-mediated ones.[31] Direct protein - DNA interactions are not spread evenly about the octamer surface but rather located at discrete sites. These are due to the formation of two types of DNA binding sites within the octamer; the α1α1 site, which uses the α1 helix from two adjacent histones, and the L1L2 site formed by the L1 and L2 loops. Salt links and hydrogen bonding between both side-chain basic and hydroxyl groups and main-chain amides with the DNA backbone phosphates form the bulk of interactions with the DNA. This is important, given that the ubiquitous distribution of nucleosomes along genomes requires it to be a non-sequence-specific DNA-binding factor. Although nucleosomes tend to prefer some DNA sequences over others,[32] they are capable of binding practically to any sequence, which is thought to be due to the flexibility in the formation of these water-mediated interactions. In addition, non-polar interactions are made between protein side-chains and the deoxyribose groups, and an arginine side-chain intercalates into the DNA minor groove at all 14 sites where it faces the octamer surface. The distribution and strength of DNA-binding sites about the octamer surface distorts the DNA within the nucleosome core. The DNA is non-uniformly bent and also contains twist defects. The twist of free B-form DNA in solution is 10.5 bp per turn. However, the overall twist of nucleosomal DNA is only 10.2 bp per turn, varying from a value of 9.4 to 10.9 bp per turn.

Histone tail domains edit

The histone tail extensions constitute up to 30% by mass of histones, but are not visible in the crystal structures of nucleosomes due to their high intrinsic flexibility, and have been thought to be largely unstructured.[33] The N-terminal tails of histones H3 and H2B pass through a channel formed by the minor grooves of the two DNA strands, protruding from the DNA every 20 bp. The N-terminal tail of histone H4, on the other hand, has a region of highly basic amino acids (16-25), which, in the crystal structure, forms an interaction with the highly acidic surface region of a H2A-H2B dimer of another nucleosome, being potentially relevant for the higher-order structure of nucleosomes. This interaction is thought to occur under physiological conditions also, and suggests that acetylation of the H4 tail distorts the higher-order structure of chromatin.[citation needed]

Higher order structure edit

 
The current chromatin compaction model.

The organization of the DNA that is achieved by the nucleosome cannot fully explain the packaging of DNA observed in the cell nucleus. Further compaction of chromatin into the cell nucleus is necessary, but it is not yet well understood. The current understanding[25] is that repeating nucleosomes with intervening "linker" DNA form a 10-nm-fiber, described as "beads on a string", and have a packing ratio of about five to ten.[18] A chain of nucleosomes can be arranged in a 30 nm fiber, a compacted structure with a packing ratio of ~50[18] and whose formation is dependent on the presence of the H1 histone.

A crystal structure of a tetranucleosome has been presented and used to build up a proposed structure of the 30 nm fiber as a two-start helix.[34] There is still a certain amount of contention regarding this model, as it is incompatible with recent electron microscopy data.[35] Beyond this, the structure of chromatin is poorly understood, but it is classically suggested that the 30 nm fiber is arranged into loops along a central protein scaffold to form transcriptionally active euchromatin. Further compaction leads to transcriptionally inactive heterochromatin.

Dynamics edit

Although the nucleosome is a very stable protein-DNA complex, it is not static and has been shown to undergo a number of different structural re-arrangements including nucleosome sliding and DNA site exposure. Depending on the context, nucleosomes can inhibit or facilitate transcription factor binding. Nucleosome positions are controlled by three major contributions: First, the intrinsic binding affinity of the histone octamer depends on the DNA sequence. Second, the nucleosome can be displaced or recruited by the competitive or cooperative binding of other protein factors. Third, the nucleosome may be actively translocated by ATP-dependent remodeling complexes.[36]

Nucleosome sliding edit

Work performed in the Bradbury laboratory showed that nucleosomes reconstituted onto the 5S DNA positioning sequence were able to reposition themselves translationally onto adjacent sequences when incubated thermally.[37] Later work showed that this repositioning did not require disruption of the histone octamer but was consistent with nucleosomes being able to "slide" along the DNA in cis. In 2008, it was further revealed that CTCF binding sites act as nucleosome positioning anchors so that, when used to align various genomic signals, multiple flanking nucleosomes can be readily identified.[38] Although nucleosomes are intrinsically mobile, eukaryotes have evolved a large family of ATP-dependent chromatin remodelling enzymes to alter chromatin structure, many of which do so via nucleosome sliding. In 2012, Beena Pillai's laboratory has demonstrated that nucleosome sliding is one of the possible mechanism for large scale tissue specific expression of genes. The work shows that the transcription start site for genes expressed in a particular tissue, are nucleosome depleted while, the same set of genes in other tissue where they are not expressed, are nucleosome bound.[39]

DNA site exposure edit

Work from the Widom laboratory has shown that nucleosomal DNA is in equilibrium between a wrapped and unwrapped state. Measurements of these rates using time-resolved FRET revealed that DNA within the nucleosome remains fully wrapped for only 250 ms before it is unwrapped for 10-50 ms and then rapidly rewrapped.[40] This implies that DNA does not need to be actively dissociated from the nucleosome but that there is a significant fraction of time during which it is fully accessible. Indeed, this can be extended to the observation that introducing a DNA-binding sequence within the nucleosome increases the accessibility of adjacent regions of DNA when bound.[41] This propensity for DNA within the nucleosome to "breathe" has important functional consequences for all DNA-binding proteins that operate in a chromatin environment.[40] In particular, the dynamic breathing of nucleosomes plays an important role in restricting the advancement of RNA polymerase II during transcription elongation.[42]

Nucleosome free region edit

Promoters of active genes have nucleosome free regions (NFR). This allows for promoter DNA accessibility to various proteins, such as transcription factors. Nucleosome free region typically spans for 200 nucleotides in S. cerevisiae[43] Well-positioned nucleosomes form boundaries of NFR. These nucleosomes are called +1-nucleosome and −1-nucleosome and are located at canonical distances downstream and upstream, respectively, from transcription start site.[44] +1-nucleosome and several downstream nucleosomes also tend to incorporate H2A.Z histone variant.[44]

Modulating nucleosome structure edit

Eukaryotic genomes are ubiquitously associated into chromatin; however, cells must spatially and temporally regulate specific loci independently of bulk chromatin. In order to achieve the high level of control required to co-ordinate nuclear processes such as DNA replication, repair, and transcription, cells have developed a variety of means to locally and specifically modulate chromatin structure and function. This can involve covalent modification of histones, the incorporation of histone variants, and non-covalent remodelling by ATP-dependent remodeling enzymes.

Histone post-translational modifications edit

 
Histone tails and their function in chromatin formation

Since they were discovered in the mid-1960s, histone modifications have been predicted to affect transcription.[45] The fact that most of the early post-translational modifications found were concentrated within the tail extensions that protrude from the nucleosome core lead to two main theories regarding the mechanism of histone modification. The first of the theories suggested that they may affect electrostatic interactions between the histone tails and DNA to "loosen" chromatin structure. Later it was proposed that combinations of these modifications may create binding epitopes with which to recruit other proteins.[46] Recently, given that more modifications have been found in the structured regions of histones, it has been put forward that these modifications may affect histone-DNA[47] and histone-histone[48] interactions within the nucleosome core. Modifications (such as acetylation or phosphorylation) that lower the charge of the globular histone core are predicted to "loosen" core-DNA association; the strength of the effect depends on location of the modification within the core.[49] Some modifications have been shown to be correlated with gene silencing; others seem to be correlated with gene activation. Common modifications include acetylation, methylation, or ubiquitination of lysine; methylation of arginine; and phosphorylation of serine. The information stored in this way is considered epigenetic, since it is not encoded in the DNA but is still inherited to daughter cells. The maintenance of a repressed or activated status of a gene is often necessary for cellular differentiation.[18]

Histone variants edit

Although histones are remarkably conserved throughout evolution, several variant forms have been identified. This diversification of histone function is restricted to H2A and H3, with H2B and H4 being mostly invariant. H2A can be replaced by H2AZ (which leads to reduced nucleosome stability) or H2AX (which is associated with DNA repair and T cell differentiation), whereas the inactive X chromosomes in mammals are enriched in macroH2A. H3 can be replaced by H3.3 (which correlates with activate genes and regulatory elements) and in centromeres H3 is replaced by CENPA.[18]

ATP-dependent nucleosome remodeling edit

A number of distinct reactions are associated with the term ATP-dependent chromatin remodeling. Remodeling enzymes have been shown to slide nucleosomes along DNA,[50] disrupt histone-DNA contacts to the extent of destabilizing the H2A/H2B dimer[51][52] and to generate negative superhelical torsion in DNA and chromatin.[53] Recently, the Swr1 remodeling enzyme has been shown to introduce the variant histone H2A.Z into nucleosomes.[54] At present, it is not clear if all of these represent distinct reactions or merely alternative outcomes of a common mechanism. What is shared between all, and indeed the hallmark of ATP-dependent chromatin remodeling, is that they all result in altered DNA accessibility.

Studies looking at gene activation in vivo[55] and, more astonishingly, remodeling in vitro[56] have revealed that chromatin remodeling events and transcription-factor binding are cyclical and periodic in nature. While the consequences of this for the reaction mechanism of chromatin remodeling are not known, the dynamic nature of the system may allow it to respond faster to external stimuli. A recent study indicates that nucleosome positions change significantly during mouse embryonic stem cell development, and these changes are related to binding of developmental transcription factors.[57]

Dynamic nucleosome remodelling across the Yeast genome edit

Studies in 2007 have catalogued nucleosome positions in yeast and shown that nucleosomes are depleted in promoter regions and origins of replication.[58][59][60] About 80% of the yeast genome appears to be covered by nucleosomes[61] and the pattern of nucleosome positioning clearly relates to DNA regions that regulate transcription, regions that are transcribed and regions that initiate DNA replication.[62] Most recently, a new study examined dynamic changes in nucleosome repositioning during a global transcriptional reprogramming event to elucidate the effects on nucleosome displacement during genome-wide transcriptional changes in yeast (Saccharomyces cerevisiae).[63] The results suggested that nucleosomes that were localized to promoter regions are displaced in response to stress (like heat shock). In addition, the removal of nucleosomes usually corresponded to transcriptional activation and the replacement of nucleosomes usually corresponded to transcriptional repression, presumably because transcription factor binding sites became more or less accessible, respectively. In general, only one or two nucleosomes were repositioned at the promoter to effect these transcriptional changes. However, even in chromosomal regions that were not associated with transcriptional changes, nucleosome repositioning was observed, suggesting that the covering and uncovering of transcriptional DNA does not necessarily produce a transcriptional event. After transcription, the rDNA region has to protected from any damage, it suggested HMGB proteins play a major role in protecting the nucleosome free region.[64][65]

DNA Twist Defects edit

DNA twist defects are when the addition of one or a few base pairs from one DNA segment are transferred to the next segment resulting in a change of the DNA twist. This will not only change the twist of the DNA but it will also change the length.[66] This twist defect eventually moves around the nucleosome through the transferring of the base pair, this means DNA twists can cause nucleosome sliding.[67] Nucleosome crystal structures have shown that superhelix location 2 and 5 on the nucleosome are commonly found to be where DNA twist defects occur as these are common remodeler binding sites.[68] There are a variety of chromatin remodelers but all share the existence of an ATPase motor which facilitates chromatin sliding on DNA through the binding and hydrolysis of ATP.[69] ATPase has an open and closed state. When the ATPase motor is changing from open and closed states, the DNA duplex changes geometry and exhibits base pair tilting.[68] The initiation of the twist defects via the ATPase motor causes tension to accumulate around the remodeler site. The tension is released when the sliding of DNA has been completed throughout the nucleosome via the spread of two twist defects (one on each strand) in opposite directions.[69]

Nucleosome assembly in vitro edit

 
Diagram of nucleosome assembly.

Nucleosomes can be assembled in vitro by either using purified native or recombinant histones.[70][71] One standard technique of loading the DNA around the histones involves the use of salt dialysis. A reaction consisting of the histone octamers and a naked DNA template can be incubated together at a salt concentration of 2 M. By steadily decreasing the salt concentration, the DNA will equilibrate to a position where it is wrapped around the histone octamers, forming nucleosomes. In appropriate conditions, this reconstitution process allows for the nucleosome positioning affinity of a given sequence to be mapped experimentally.[72]

Disulfide crosslinked nucleosome core particles edit

A recent advance in the production of nucleosome core particles with enhanced stability involves site-specific disulfide crosslinks.[73] Two different crosslinks can be introduced into the nucleosome core particle. A first one crosslinks the two copies of H2A via an introduced cysteine (N38C) resulting in histone octamer which is stable against H2A/H2B dimer loss during nucleosome reconstitution. A second crosslink can be introduced between the H3 N-terminal histone tail and the nucleosome DNA ends via an incorporated convertible nucleotide.[74] The DNA-histone octamer crosslink stabilizes the nucleosome core particle against DNA dissociation at very low particle concentrations and at elevated salt concentrations.

Nucleosome assembly in vivo edit

 
Steps in nucleosome assembly

Nucleosomes are the basic packing unit of genomic DNA built from histone proteins around which DNA is coiled. They serve as a scaffold for formation of higher order chromatin structure as well as for a layer of regulatory control of gene expression. Nucleosomes are quickly assembled onto newly synthesized DNA behind the replication fork.

H3 and H4 edit

Histones H3 and H4 from disassembled old nucleosomes are kept in the vicinity and randomly distributed on the newly synthesized DNA.[75] They are assembled by the chromatin assembly factor 1 (CAF-1) complex, which consists of three subunits (p150, p60, and p48).[76] Newly synthesized H3 and H4 are assembled by the replication coupling assembly factor (RCAF). RCAF contains the subunit Asf1, which binds to newly synthesized H3 and H4 proteins.[77] The old H3 and H4 proteins retain their chemical modifications which contributes to the passing down of the epigenetic signature. The newly synthesized H3 and H4 proteins are gradually acetylated at different lysine residues as part of the chromatin maturation process.[78] It is also thought that the old H3 and H4 proteins in the new nucleosomes recruit histone modifying enzymes that mark the new histones, contributing to epigenetic memory.

H2A and H2B edit

In contrast to old H3 and H4, the old H2A and H2B histone proteins are released and degraded; therefore, newly assembled H2A and H2B proteins are incorporated into new nucleosomes.[79] H2A and H2B are assembled into dimers which are then loaded onto nucleosomes by the nucleosome assembly protein-1 (NAP-1) which also assists with nucleosome sliding.[80] The nucleosomes are also spaced by ATP-dependent nucleosome-remodeling complexes containing enzymes such as Isw1 Ino80, and Chd1, and subsequently assembled into higher order structure.[81][82]

Gallery edit

The crystal structure of the nucleosome core particle (PDB: 1EQZ[28]) - different views showing details of histone folding and organization. Histones H2A, H2B, H3, H4 and DNA are coloured.

See also edit

References edit

  1. ^ Reece J, Campbell N (2006). Biology. San Francisco: Benjamin Cummings. ISBN 978-0-8053-6624-2.
  2. ^ Alberts B (2002). Molecular biology of the cell (4th ed.). New York: Garland Science. p. 207. ISBN 978-0-8153-4072-0.
  3. ^ lberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2002). Chromosomal DNA and Its Packaging in the Chromatin Fiber.
  4. ^ Teif VB, Clarkson CT (2019). "Nucleosome Positioning". Encyclopedia of Bioinformatics and Computational Biology. 2: 308–317. doi:10.1016/B978-0-12-809633-8.20242-2. ISBN 9780128114322. S2CID 43929234.
  5. ^ Olins AL, Olins DE (January 1974). "Spheroid chromatin units (v bodies)". Science. 183 (4122): 330–332. Bibcode:1974Sci...183..330O. doi:10.1126/science.183.4122.330. PMID 4128918. S2CID 83480762.
  6. ^ McDonald D (December 2005). "Milestone 9, (1973-1974) The nucleosome hypothesis: An alternative string theory". Nature Milestones: Gene Expression. doi:10.1038/nrm1798.
  7. ^ Kornberg RD (May 1974). "Chromatin structure: a repeating unit of histones and DNA". Science. 184 (4139): 868–871. Bibcode:1974Sci...184..868K. doi:10.1126/science.184.4139.868. PMID 4825889.
  8. ^ Lorch Y, LaPointe JW, Kornberg RD (April 1987). "Nucleosomes inhibit the initiation of transcription but allow chain elongation with the displacement of histones". Cell. 49 (2): 203–210. doi:10.1016/0092-8674(87)90561-7. PMID 3568125. S2CID 21270171.
  9. ^ Han M, Grunstein M (December 1988). "Nucleosome loss activates yeast downstream promoters in vivo". Cell. 55 (6): 1137–1145. doi:10.1016/0092-8674(88)90258-9. PMID 2849508. S2CID 41520634.
  10. ^ Clark-Adams CD, Norris D, Osley MA, Fassler JS, Winston F (February 1988). "Changes in histone gene dosage alter transcription in yeast". Genes & Development. 2 (2): 150–159. doi:10.1101/gad.2.2.150. PMID 2834270.
  11. ^ a b In different crystals, values of 146 and 147 basepairs were observed
  12. ^ a b Luger K, Mäder AW, Richmond RK, Sargent DF, Richmond TJ (September 1997). "Crystal structure of the nucleosome core particle at 2.8 A resolution". Nature. 389 (6648): 251–260. Bibcode:1997Natur.389..251L. doi:10.1038/38444. PMID 9305837. S2CID 4328827.
  13. ^ Alberts B (2007). Molecular Biology of the Cell (5th ed.). New York: Garland Science. p. 211. ISBN 978-0-8153-4106-2.
  14. ^ Shtumpf M, Piroeva KV, Agrawal SP, Jacob DR, Teif VB (June 2022). "NucPosDB: a database of nucleosome positioning in vivo and nucleosomics of cell-free DNA". Chromosoma. 131 (1–2): 19–28. doi:10.1007/s00412-021-00766-9. PMC 8776978. PMID 35061087.
  15. ^ Zhou YB, Gerchman SE, Ramakrishnan V, Travers A, Muyldermans S (September 1998). "Position and orientation of the globular domain of linker histone H5 on the nucleosome". Nature. 395 (6700): 402–405. Bibcode:1998Natur.395..402Z. doi:10.1038/26521. PMID 9759733. S2CID 204997317.
  16. ^ Thoma F, Koller T, Klug A (November 1979). "Involvement of histone H1 in the organization of the nucleosome and of the salt-dependent superstructures of chromatin". The Journal of Cell Biology. 83 (2 Pt 1): 403–427. doi:10.1083/jcb.83.2.403. PMC 2111545. PMID 387806.
  17. ^ Clarke HJ (1992). "Nuclear and chromatin composition of mammalian gametes and early embryos". Biochemistry and Cell Biology. 70 (10–11): 856–866. doi:10.1139/o92-134. PMID 1297351.
  18. ^ a b c d e f Felsenfeld G, Groudine M (January 2003). "Controlling the double helix". Nature. 421 (6921): 448–453. Bibcode:2003Natur.421..448F. doi:10.1038/nature01411. PMID 12540921.
  19. ^ Richmond TJ, Finch JT, Rushton B, Rhodes D, Klug A (1984). "Structure of the nucleosome core particle at 7 A resolution". Nature. 311 (5986): 532–7. Bibcode:1984Natur.311..532R. doi:10.1038/311532a0. PMID 6482966. S2CID 4355982.
  20. ^ Harp JM, Palmer EL, York MH, Gewiess A, Davis M, Bunick GJ (October 1995). "Preparative separation of nucleosome core particles containing defined-sequence DNA in multiple translational phases". Electrophoresis. 16 (10): 1861–1864. doi:10.1002/elps.11501601305. PMID 8586054. S2CID 20178479.
  21. ^ Palmer EL, Gewiess A, Harp JM, York MH, Bunick GJ (October 1995). "Large-scale production of palindrome DNA fragments". Analytical Biochemistry. 231 (1): 109–114. doi:10.1006/abio.1995.1509. PMID 8678288.
  22. ^ Harp JM, Uberbacher EC, Roberson AE, Palmer EL, Gewiess A, Bunick GJ (March 1996). "X-ray diffraction analysis of crystals containing twofold symmetric nucleosome core particles". Acta Crystallographica. Section D, Biological Crystallography. 52 (Pt 2): 283–288. doi:10.1107/S0907444995009139. PMID 15299701.
  23. ^ Harp JM, Hanson BL, Timm DE, Bunick GJ (December 2000). "Asymmetries in the nucleosome core particle at 2.5 A resolution". Acta Crystallographica. Section D, Biological Crystallography. 56 (Pt 12): 1513–1534. doi:10.1107/s0907444900011847. PMID 11092917.
  24. ^ Hanson BL, Alexander C, Harp JM, Bunick GJ (2004). "Preparation and crystallization of nucleosome core particle". Chromatin and Chromatin Remodeling Enzymes, Part A. Methods in Enzymology. Vol. 375. pp. 44–62. doi:10.1016/s0076-6879(03)75003-4. ISBN 9780121827793. PMID 14870658.
  25. ^ a b Chakravarthy S, Park YJ, Chodaparambil J, Edayathumangalam RS, Luger K (February 2005). "Structure and dynamic properties of nucleosome core particles". FEBS Letters. 579 (4): 895–898. doi:10.1016/j.febslet.2004.11.030. PMID 15680970. S2CID 41706403.
  26. ^ White CL, Suto RK, Luger K (September 2001). "Structure of the yeast nucleosome core particle reveals fundamental changes in internucleosome interactions". The EMBO Journal. 20 (18): 5207–5218. doi:10.1093/emboj/20.18.5207. PMC 125637. PMID 11566884.
  27. ^ a b Stryer L (1995). Biochemistry (fourth ed.). New York - Basingstoke: W. H. Freeman and Company. ISBN 978-0716720096.
  28. ^ a b Harp JM, Hanson BL, Timm DE, Bunick GJ (December 2000). "Asymmetries in the nucleosome core particle at 2.5 A resolution". Acta Crystallographica. Section D, Biological Crystallography. 56 (Pt 12): 1513–1534. doi:10.1107/S0907444900011847. PMID 11092917. PDB ID: 1EQZ.
  29. ^ Alberts B (2009). Essential Cell Biology (2nd ed.). New York: Garland Science.
  30. ^ Allen, Paul D.; Newland, Adrian C. (1 June 1998). "Electrophoretic DNA analysis for the detection of apoptosis". Molecular Biotechnology. 9 (3): 247–251. doi:10.1007/BF02915798. ISSN 1559-0305.
  31. ^ Davey CA, Sargent DF, Luger K, Maeder AW, Richmond TJ (June 2002). "Solvent mediated interactions in the structure of the nucleosome core particle at 1.9 a resolution". Journal of Molecular Biology. 319 (5): 1097–1113. doi:10.1016/S0022-2836(02)00386-8. PMID 12079350.
  32. ^ Segal E, Fondufe-Mittendorf Y, Chen L, Thåström A, Field Y, Moore IK, et al. (August 2006). "A genomic code for nucleosome positioning". Nature. 442 (7104): 772–778. Bibcode:2006Natur.442..772S. doi:10.1038/nature04979. PMC 2623244. PMID 16862119.
  33. ^ Zheng C, Hayes JJ (April 2003). "Structures and interactions of the core histone tail domains". Biopolymers. 68 (4): 539–546. doi:10.1002/bip.10303. PMID 12666178.
  34. ^ Schalch T, Duda S, Sargent DF, Richmond TJ (July 2005). "X-ray structure of a tetranucleosome and its implications for the chromatin fibre". Nature. 436 (7047): 138–141. Bibcode:2005Natur.436..138S. doi:10.1038/nature03686. PMID 16001076. S2CID 4387396.
  35. ^ Robinson PJ, Fairall L, Huynh VA, Rhodes D (April 2006). "EM measurements define the dimensions of the "30-nm" chromatin fiber: evidence for a compact, interdigitated structure". Proceedings of the National Academy of Sciences of the United States of America. 103 (17): 6506–6511. Bibcode:2006PNAS..103.6506R. doi:10.1073/pnas.0601212103. PMC 1436021. PMID 16617109.
  36. ^ Teif VB, Rippe K (September 2009). "Predicting nucleosome positions on the DNA: combining intrinsic sequence preferences and remodeler activities". Nucleic Acids Research. 37 (17): 5641–5655. doi:10.1093/nar/gkp610. PMC 2761276. PMID 19625488.
  37. ^ Pennings S, Muyldermans S, Meersseman G, Wyns L (May 1989). "Formation, stability and core histone positioning of nucleosomes reassembled on bent and other nucleosome-derived DNA". Journal of Molecular Biology. 207 (1): 183–192. doi:10.1016/0022-2836(89)90449-X. PMID 2738923.
  38. ^ Fu Y, Sinha M, Peterson CL, Weng Z (July 2008). Van Steensel B (ed.). "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.
  39. ^ Bargaje R, Alam MP, Patowary A, Sarkar M, Ali T, Gupta S, et al. (October 2012). "Proximity of H2A.Z containing nucleosome to the transcription start site influences gene expression levels in the mammalian liver and brain". Nucleic Acids Research. 40 (18): 8965–8978. doi:10.1093/nar/gks665. PMC 3467062. PMID 22821566.
  40. ^ a b Li G, Levitus M, Bustamante C, Widom J (January 2005). "Rapid spontaneous accessibility of nucleosomal DNA". Nature Structural & Molecular Biology. 12 (1): 46–53. doi:10.1038/nsmb869. PMID 15580276. S2CID 14540078.
  41. ^ Li G, Widom J (August 2004). "Nucleosomes facilitate their own invasion". Nature Structural & Molecular Biology. 11 (8): 763–769. doi:10.1038/nsmb801. PMID 15258568. S2CID 11299024.
  42. ^ Hodges C, Bintu L, Lubkowska L, Kashlev M, Bustamante C (July 2009). "Nucleosomal fluctuations govern the transcription dynamics of RNA polymerase II". Science. 325 (5940): 626–628. Bibcode:2009Sci...325..626H. doi:10.1126/science.1172926. PMC 2775800. PMID 19644123.
  43. ^ Yuan GC, Liu YJ, Dion MF, Slack MD, Wu LF, Altschuler SJ, Rando OJ (July 2005). "Genome-scale identification of nucleosome positions in S. cerevisiae". Science. 309 (5734): 626–630. Bibcode:2005Sci...309..626Y. doi:10.1126/science.1112178. PMID 15961632. S2CID 43625066.
  44. ^ a b Lai WK, Pugh BF (September 2017). "Understanding nucleosome dynamics and their links to gene expression and DNA replication". Nature Reviews. Molecular Cell Biology. 18 (9): 548–562. doi:10.1038/nrm.2017.47. PMC 5831138. PMID 28537572.
  45. ^ Allfrey VG, Faulkner R, Mirsky AE (May 1964). "Acetylation and Methylation of Histones and Their Possible Role in the Regulation of RNA Synthesis". Proceedings of the National Academy of Sciences of the United States of America. 51 (5): 786–794. Bibcode:1964PNAS...51..786A. doi:10.1073/pnas.51.5.786. PMC 300163. PMID 14172992.
  46. ^ Strahl BD, Allis CD (January 2000). "The language of covalent histone modifications". Nature. 403 (6765): 41–45. Bibcode:2000Natur.403...41S. doi:10.1038/47412. PMID 10638745. S2CID 4418993.
  47. ^ Cosgrove MS, Boeke JD, Wolberger C (November 2004). "Regulated nucleosome mobility and the histone code". Nature Structural & Molecular Biology. 11 (11): 1037–1043. doi:10.1038/nsmb851. PMID 15523479. S2CID 34704745.
  48. ^ Ye J, Ai X, Eugeni EE, Zhang L, Carpenter LR, Jelinek MA, et al. (April 2005). "Histone H4 lysine 91 acetylation a core domain modification associated with chromatin assembly". Molecular Cell. 18 (1): 123–130. doi:10.1016/j.molcel.2005.02.031. PMC 2855496. PMID 15808514.
  49. ^ Fenley AT, Adams DA, Onufriev AV (September 2010). "Charge state of the globular histone core controls stability of the nucleosome". Biophysical Journal. 99 (5): 1577–1585. Bibcode:2010BpJ....99.1577F. doi:10.1016/j.bpj.2010.06.046. PMC 2931741. PMID 20816070.
  50. ^ Whitehouse I, Flaus A, Cairns BR, White MF, Workman JL, Owen-Hughes T (August 1999). "Nucleosome mobilization catalysed by the yeast SWI/SNF complex". Nature. 400 (6746): 784–787. Bibcode:1999Natur.400..784W. doi:10.1038/23506. PMID 10466730. S2CID 2841873.
  51. ^ Kassabov SR, Zhang B, Persinger J, Bartholomew B (February 2003). "SWI/SNF unwraps, slides, and rewraps the nucleosome". Molecular Cell. 11 (2): 391–403. doi:10.1016/S1097-2765(03)00039-X. PMID 12620227.
  52. ^ Bruno M, Flaus A, Stockdale C, Rencurel C, Ferreira H, Owen-Hughes T (December 2003). "Histone H2A/H2B dimer exchange by ATP-dependent chromatin remodeling activities". Molecular Cell. 12 (6): 1599–1606. doi:10.1016/S1097-2765(03)00499-4. PMC 3428624. PMID 14690611.
  53. ^ Havas K, Flaus A, Phelan M, Kingston R, Wade PA, Lilley DM, Owen-Hughes T (December 2000). "Generation of superhelical torsion by ATP-dependent chromatin remodeling activities". Cell. 103 (7): 1133–1142. doi:10.1016/S0092-8674(00)00215-4. PMID 11163188. S2CID 7911590.
  54. ^ Mizuguchi G, Shen X, Landry J, Wu WH, Sen S, Wu C (January 2004). "ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex". Science. 303 (5656): 343–348. Bibcode:2004Sci...303..343M. doi:10.1126/science.1090701. PMID 14645854. S2CID 9881829.
  55. ^ Métivier R, Penot G, Hübner MR, Reid G, Brand H, Kos M, Gannon F (December 2003). "Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter". Cell. 115 (6): 751–763. doi:10.1016/S0092-8674(03)00934-6. PMID 14675539. S2CID 145525.
  56. ^ Nagaich AK, Walker DA, Wolford R, Hager GL (April 2004). "Rapid periodic binding and displacement of the glucocorticoid receptor during chromatin remodeling". Molecular Cell. 14 (2): 163–174. doi:10.1016/S1097-2765(04)00178-9. PMID 15099516.
  57. ^ Teif VB, Vainshtein Y, Caudron-Herger M, Mallm JP, Marth C, Höfer T, Rippe K (November 2012). "Genome-wide nucleosome positioning during embryonic stem cell development". Nature Structural & Molecular Biology. 19 (11): 1185–1192. doi:10.1038/nsmb.2419. PMID 23085715. S2CID 34509771.
  58. ^ Albert I, Mavrich TN, Tomsho LP, Qi J, Zanton SJ, Schuster SC, Pugh BF (March 2007). "Translational and rotational settings of H2A.Z nucleosomes across the Saccharomyces cerevisiae genome". Nature. 446 (7135): 572–576. Bibcode:2007Natur.446..572A. doi:10.1038/nature05632. PMID 17392789. S2CID 4416890.
  59. ^ Li B, Carey M, Workman JL (February 2007). "The role of chromatin during transcription". Cell. 128 (4): 707–719. doi:10.1016/j.cell.2007.01.015. PMID 17320508. S2CID 1773333.
  60. ^ Whitehouse I, Rando OJ, Delrow J, Tsukiyama T (December 2007). "Chromatin remodelling at promoters suppresses antisense transcription". Nature. 450 (7172): 1031–1035. Bibcode:2007Natur.450.1031W. doi:10.1038/nature06391. PMID 18075583. S2CID 4305576.
  61. ^ Lee W, Tillo D, Bray N, Morse RH, Davis RW, Hughes TR, Nislow C (October 2007). "A high-resolution atlas of nucleosome occupancy in yeast". Nature Genetics. 39 (10): 1235–1244. doi:10.1038/ng2117. PMID 17873876. S2CID 12816925.
  62. ^ Eaton ML, Galani K, Kang S, Bell SP, MacAlpine DM (April 2010). "Conserved nucleosome positioning defines replication origins". Genes & Development. 24 (8): 748–753. doi:10.1101/gad.1913210. PMC 2854390. PMID 20351051.
  63. ^ Shivaswamy S, Bhinge A, Zhao Y, Jones S, Hirst M, Iyer VR (March 2008). "Dynamic remodeling of individual nucleosomes across a eukaryotic genome in response to transcriptional perturbation". PLOS Biology. 6 (3): e65. doi:10.1371/journal.pbio.0060065. PMC 2267817. PMID 18351804.
  64. ^ Murugesapillai D, McCauley MJ, Huo R, Nelson Holte MH, Stepanyants A, Maher LJ, et al. (August 2014). "DNA bridging and looping by HMO1 provides a mechanism for stabilizing nucleosome-free chromatin". Nucleic Acids Research. 42 (14): 8996–9004. doi:10.1093/nar/gku635. PMC 4132745. PMID 25063301.
  65. ^ Murugesapillai D, McCauley MJ, Maher LJ, Williams MC (February 2017). "Single-molecule studies of high-mobility group B architectural DNA bending proteins". Biophysical Reviews. 9 (1): 17–40. doi:10.1007/s12551-016-0236-4. PMC 5331113. PMID 28303166.
  66. ^ Winger J, Nodelman IM, Levendosky RF, Bowman GD (May 2018). "A twist defect mechanism for ATP-dependent translocation of nucleosomal DNA". eLife. 7: e34100. doi:10.7554/eLife.34100. PMC 6031429. PMID 29809147.
  67. ^ Bowman GD (August 2019). "Uncovering a New Step in Sliding Nucleosomes". Trends in Biochemical Sciences. 44 (8): 643–645. doi:10.1016/j.tibs.2019.05.001. PMC 7092708. PMID 31171402.
  68. ^ a b Nodelman IM, Bowman GD (May 2021). "Biophysics of Chromatin Remodeling". Annual Review of Biophysics. 50 (1): 73–93. doi:10.1146/annurev-biophys-082520-080201. PMC 8428145. PMID 33395550.
  69. ^ a b Brandani GB, Takada S (November 2018). Onufriev A (ed.). "Chromatin remodelers couple inchworm motion with twist-defect formation to slide nucleosomal DNA". PLOS Computational Biology. 14 (11): e1006512. Bibcode:2018PLSCB..14E6512B. doi:10.1371/journal.pcbi.1006512. PMC 6237416. PMID 30395604.
  70. ^ Hayes JJ, Lee KM (May 1997). "In vitro reconstitution and analysis of mononucleosomes containing defined DNAs and proteins". Methods. 12 (1): 2–9. doi:10.1006/meth.1997.0441. PMID 9169189.
  71. ^ Dyer PN, Edayathumangalam RS, White CL, Bao Y, Chakravarthy S, Muthurajan UM, Luger K (2004). "Reconstitution of nucleosome core particles from recombinant histones and DNA". Chromatin and Chromatin Remodeling Enzymes, Part A. Methods in Enzymology. Vol. 375. pp. 23–44. doi:10.1016/s0076-6879(03)75002-2. ISBN 9780121827793. PMID 14870657.
  72. ^ Yenidunya A, Davey C, Clark D, Felsenfeld G, Allan J (April 1994). "Nucleosome positioning on chicken and human globin gene promoters in vitro. Novel mapping techniques". Journal of Molecular Biology. 237 (4): 401–414. doi:10.1006/jmbi.1994.1243. PMID 8151701.
  73. ^ Frouws TD, Barth PD, Richmond TJ (January 2018). "Site-Specific Disulfide Crosslinked Nucleosomes with Enhanced Stability". Journal of Molecular Biology. 430 (1): 45–57. doi:10.1016/j.jmb.2017.10.029. PMC 5757783. PMID 29113904.
  74. ^ Ferentz AE, Verdine GL (1994). "The Convertible Nucleoside Approach: Structural Engineering of Nucleic Acids by Disulfide Cross-Linking". In Eckstein F, Lilley DM (eds.). Nucleic Acids and Molecular Biology. Vol. 8. pp. 14–40. doi:10.1007/978-3-642-78666-2_2. ISBN 978-3-642-78668-6.
  75. ^ Yamasu K, Senshu T (January 1990). "Conservative segregation of tetrameric units of H3 and H4 histones during nucleosome replication". Journal of Biochemistry. 107 (1): 15–20. doi:10.1093/oxfordjournals.jbchem.a122999. PMID 2332416.
  76. ^ Kaufman PD, Kobayashi R, Kessler N, Stillman B (June 1995). "The p150 and p60 subunits of chromatin assembly factor I: a molecular link between newly synthesized histones and DNA replication". Cell. 81 (7): 1105–1114. doi:10.1016/S0092-8674(05)80015-7. PMID 7600578. S2CID 13502921.
  77. ^ Tyler JK, Adams CR, Chen SR, Kobayashi R, Kamakaka RT, Kadonaga JT (December 1999). "The RCAF complex mediates chromatin assembly during DNA replication and repair". Nature. 402 (6761): 555–560. Bibcode:1999Natur.402..555T. doi:10.1038/990147. PMID 10591219. S2CID 205097512.
  78. ^ Benson LJ, Gu Y, Yakovleva T, Tong K, Barrows C, Strack CL, et al. (April 2006). "Modifications of H3 and H4 during chromatin replication, nucleosome assembly, and histone exchange". The Journal of Biological Chemistry. 281 (14): 9287–9296. doi:10.1074/jbc.M512956200. PMID 16464854.
  79. ^ Louters L, Chalkley R (June 1985). "Exchange of histones H1, H2A, and H2B in vivo". Biochemistry. 24 (13): 3080–3085. doi:10.1021/bi00334a002. PMID 4027229.
  80. ^ Park YJ, Chodaparambil JV, Bao Y, McBryant SJ, Luger K (January 2005). "Nucleosome assembly protein 1 exchanges histone H2A-H2B dimers and assists nucleosome sliding". The Journal of Biological Chemistry. 280 (3): 1817–1825. doi:10.1074/jbc.M411347200. PMID 15516689.
  81. ^ Vincent JA, Kwong TJ, Tsukiyama T (May 2008). "ATP-dependent chromatin remodeling shapes the DNA replication landscape". Nature Structural & Molecular Biology. 15 (5): 477–484. doi:10.1038/nsmb.1419. PMC 2678716. PMID 18408730.
  82. ^ Yadav T, Whitehouse I (April 2016). "Replication-Coupled Nucleosome Assembly and Positioning by ATP-Dependent Chromatin-Remodeling Enzymes". Cell Reports. 15 (4): 715–723. doi:10.1016/J.CELREP.2016.03.059. PMC 5063657. PMID 27149855.

External links edit

  • MBInfo - What are nucleosomes
  • Nucleosomes on the Richmond Lab website
  • Proteopedia Nucleosomes
  • Nucleosome positioning data and tools online (annotated list, constantly updated)
  • Histone protein structure
  • HistoneDB 2.0 - Database of histones and variants at NCBI

nucleosome, nucleosome, basic, structural, unit, packaging, eukaryotes, structure, nucleosome, consists, segment, wound, around, eight, histone, proteins, resembles, thread, wrapped, around, spool, nucleosome, fundamental, subunit, chromatin, each, nucleosome,. 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 1 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 Basic units of chromatin structureDNA must be compacted into nucleosomes to fit within the cell nucleus 2 In addition to nucleosome wrapping eukaryotic chromatin is further compacted by being folded into a series of more complex structures eventually forming a chromosome Each human cell contains about 30 million nucleosomes 3 Nucleosomes are thought to carry epigenetically inherited information in the form of covalent modifications of their core histones Nucleosome positions in the genome are not random and it is important to know where each nucleosome is located because this determines the accessibility of the DNA to regulatory proteins 4 Nucleosomes were first observed as particles in the electron microscope by Don and Ada Olins in 1974 5 and their existence and structure as histone octamers surrounded by approximately 200 base pairs of DNA were proposed by Roger Kornberg 6 7 The role of the nucleosome as a regulator of transcription was demonstrated by Lorch et al in vitro 8 in 1987 and by Han and Grunstein 9 and Clark Adams et al 10 in vivo in 1988 The nucleosome core particle consists of approximately 146 base pairs bp of DNA 11 wrapped in 1 67 left handed superhelical turns around a histone octamer consisting of 2 copies each of the core histones H2A H2B H3 and H4 12 Core particles are connected by stretches of linker DNA which can be up to about 80 bp long Technically a nucleosome is defined as the core particle plus one of these linker regions however the word is often synonymous with the core particle 13 Genome wide nucleosome positioning maps are now available for many model organisms and human cells 14 Linker histones such as H1 and its isoforms are involved in chromatin compaction and sit at the base of the nucleosome near the DNA entry and exit binding to the linker region of the DNA 15 Non condensed nucleosomes without the linker histone resemble beads on a string of DNA under an electron microscope 16 In contrast to most eukaryotic cells mature sperm cells largely use protamines to package their genomic DNA most likely to achieve an even higher packaging ratio 17 Histone equivalents and a simplified chromatin structure have also been found in Archaea 18 suggesting that eukaryotes are not the only organisms that use nucleosomes Contents 1 Structure 1 1 Structure of the core particle 1 1 1 Overview 1 1 2 The nucleosome core particle NCP 1 1 2 1 Protein interactions within the nucleosome 1 1 2 2 Histone DNA interactions 1 1 3 Histone tail domains 1 2 Higher order structure 2 Dynamics 2 1 Nucleosome sliding 2 2 DNA site exposure 2 3 Nucleosome free region 3 Modulating nucleosome structure 3 1 Histone post translational modifications 3 2 Histone variants 3 3 ATP dependent nucleosome remodeling 3 4 Dynamic nucleosome remodelling across the Yeast genome 3 5 DNA Twist Defects 4 Nucleosome assembly in vitro 4 1 Disulfide crosslinked nucleosome core particles 5 Nucleosome assembly in vivo 5 1 H3 and H4 5 2 H2A and H2B 6 Gallery 7 See also 8 References 9 External linksStructure editStructure of the core particle edit nbsp The crystal structure of the nucleosome core particle consisting of H2A H2B H3 and H4 core histones and DNA The view is from the top through the superhelical axis Overview edit Pioneering structural studies in the 1980s by Aaron Klug s group provided the first evidence that an octamer of histone proteins wraps DNA around itself in about 1 7 turns of a left handed superhelix 19 In 1997 the first near atomic resolution crystal structure of the nucleosome was solved by the Richmond group showing the most important details of the particle The human alpha satellite palindromic DNA critical to achieving the 1997 nucleosome crystal structure was developed by the Bunick group at Oak Ridge National Laboratory in Tennessee 20 21 22 23 24 The structures of over 20 different nucleosome core particles have been solved to date 25 including those containing histone variants and histones from different species The structure of the nucleosome core particle is remarkably conserved and even a change of over 100 residues between frog and yeast histones results in electron density maps with an overall root mean square deviation of only 1 6A 26 The nucleosome core particle NCP edit The nucleosome core particle shown in the figure consists of about 146 base pair of DNA 11 wrapped in 1 67 left handed superhelical turns around the histone octamer consisting of 2 copies each of the core histones H2A H2B H3 and H4 Adjacent nucleosomes are joined by a stretch of free DNA termed linker DNA which varies from 10 80 bp in length depending on species and tissue type 18 The whole structure generates a cylinder of diameter 11 nm and a height of 5 5 nm nbsp Apoptotic DNA laddering Digested chromatin is in the first lane the second contains DNA standard to compare lengths nbsp Scheme of nucleosome organization 27 nbsp The crystal structure of the nucleosome core particle PDB 1EQZ 28 Nucleosome core particles are observed when chromatin in interphase is treated to cause the chromatin to unfold partially The resulting image via an electron microscope is beads on a string The string is the DNA while each bead in the nucleosome is a core particle The nucleosome core particle is composed of DNA and histone proteins 29 Partial DNAse digestion of chromatin reveals its nucleosome structure Because DNA portions of nucleosome core particles are less accessible for DNAse than linking sections DNA gets digested into fragments of lengths equal to multiplicity of distance between nucleosomes 180 360 540 base pairs etc Hence a very characteristic pattern similar to a ladder is visible during gel electrophoresis of that DNA 27 Such digestion can occur also under natural conditions during apoptosis cell suicide or programmed cell death because autodestruction of DNA typically is its role 30 Protein interactions within the nucleosome edit The core histone proteins contains a characteristic structural motif termed the histone fold which consists of three alpha helices a1 3 separated by two loops L1 2 In solution the histones form H2A H2B heterodimers and H3 H4 heterotetramers Histones dimerise about their long a2 helices in an anti parallel orientation and in the case of H3 and H4 two such dimers form a 4 helix bundle stabilised by extensive H3 H3 interaction The H2A H2B dimer binds onto the H3 H4 tetramer due to interactions between H4 and H2B which include the formation of a hydrophobic cluster 12 The histone octamer is formed by a central H3 H4 tetramer sandwiched between two H2A H2B dimers Due to the highly basic charge of all four core histones the histone octamer is stable only in the presence of DNA or very high salt concentrations Histone DNA interactions edit The nucleosome contains over 120 direct protein DNA interactions and several hundred water mediated ones 31 Direct protein DNA interactions are not spread evenly about the octamer surface but rather located at discrete sites These are due to the formation of two types of DNA binding sites within the octamer the a1a1 site which uses the a1 helix from two adjacent histones and the L1L2 site formed by the L1 and L2 loops Salt links and hydrogen bonding between both side chain basic and hydroxyl groups and main chain amides with the DNA backbone phosphates form the bulk of interactions with the DNA This is important given that the ubiquitous distribution of nucleosomes along genomes requires it to be a non sequence specific DNA binding factor Although nucleosomes tend to prefer some DNA sequences over others 32 they are capable of binding practically to any sequence which is thought to be due to the flexibility in the formation of these water mediated interactions In addition non polar interactions are made between protein side chains and the deoxyribose groups and an arginine side chain intercalates into the DNA minor groove at all 14 sites where it faces the octamer surface The distribution and strength of DNA binding sites about the octamer surface distorts the DNA within the nucleosome core The DNA is non uniformly bent and also contains twist defects The twist of free B form DNA in solution is 10 5 bp per turn However the overall twist of nucleosomal DNA is only 10 2 bp per turn varying from a value of 9 4 to 10 9 bp per turn Histone tail domains edit The histone tail extensions constitute up to 30 by mass of histones but are not visible in the crystal structures of nucleosomes due to their high intrinsic flexibility and have been thought to be largely unstructured 33 The N terminal tails of histones H3 and H2B pass through a channel formed by the minor grooves of the two DNA strands protruding from the DNA every 20 bp The N terminal tail of histone H4 on the other hand has a region of highly basic amino acids 16 25 which in the crystal structure forms an interaction with the highly acidic surface region of a H2A H2B dimer of another nucleosome being potentially relevant for the higher order structure of nucleosomes This interaction is thought to occur under physiological conditions also and suggests that acetylation of the H4 tail distorts the higher order structure of chromatin citation needed Higher order structure edit nbsp The current chromatin compaction model The organization of the DNA that is achieved by the nucleosome cannot fully explain the packaging of DNA observed in the cell nucleus Further compaction of chromatin into the cell nucleus is necessary but it is not yet well understood The current understanding 25 is that repeating nucleosomes with intervening linker DNA form a 10 nm fiber described as beads on a string and have a packing ratio of about five to ten 18 A chain of nucleosomes can be arranged in a 30 nm fiber a compacted structure with a packing ratio of 50 18 and whose formation is dependent on the presence of the H1 histone A crystal structure of a tetranucleosome has been presented and used to build up a proposed structure of the 30 nm fiber as a two start helix 34 There is still a certain amount of contention regarding this model as it is incompatible with recent electron microscopy data 35 Beyond this the structure of chromatin is poorly understood but it is classically suggested that the 30 nm fiber is arranged into loops along a central protein scaffold to form transcriptionally active euchromatin Further compaction leads to transcriptionally inactive heterochromatin Dynamics editAlthough the nucleosome is a very stable protein DNA complex it is not static and has been shown to undergo a number of different structural re arrangements including nucleosome sliding and DNA site exposure Depending on the context nucleosomes can inhibit or facilitate transcription factor binding Nucleosome positions are controlled by three major contributions First the intrinsic binding affinity of the histone octamer depends on the DNA sequence Second the nucleosome can be displaced or recruited by the competitive or cooperative binding of other protein factors Third the nucleosome may be actively translocated by ATP dependent remodeling complexes 36 Nucleosome sliding edit Work performed in the Bradbury laboratory showed that nucleosomes reconstituted onto the 5S DNA positioning sequence were able to reposition themselves translationally onto adjacent sequences when incubated thermally 37 Later work showed that this repositioning did not require disruption of the histone octamer but was consistent with nucleosomes being able to slide along the DNA in cis In 2008 it was further revealed that CTCF binding sites act as nucleosome positioning anchors so that when used to align various genomic signals multiple flanking nucleosomes can be readily identified 38 Although nucleosomes are intrinsically mobile eukaryotes have evolved a large family of ATP dependent chromatin remodelling enzymes to alter chromatin structure many of which do so via nucleosome sliding In 2012 Beena Pillai s laboratory has demonstrated that nucleosome sliding is one of the possible mechanism for large scale tissue specific expression of genes The work shows that the transcription start site for genes expressed in a particular tissue are nucleosome depleted while the same set of genes in other tissue where they are not expressed are nucleosome bound 39 DNA site exposure edit Work from the Widom laboratory has shown that nucleosomal DNA is in equilibrium between a wrapped and unwrapped state Measurements of these rates using time resolved FRET revealed that DNA within the nucleosome remains fully wrapped for only 250 ms before it is unwrapped for 10 50 ms and then rapidly rewrapped 40 This implies that DNA does not need to be actively dissociated from the nucleosome but that there is a significant fraction of time during which it is fully accessible Indeed this can be extended to the observation that introducing a DNA binding sequence within the nucleosome increases the accessibility of adjacent regions of DNA when bound 41 This propensity for DNA within the nucleosome to breathe has important functional consequences for all DNA binding proteins that operate in a chromatin environment 40 In particular the dynamic breathing of nucleosomes plays an important role in restricting the advancement of RNA polymerase II during transcription elongation 42 Nucleosome free region edit Promoters of active genes have nucleosome free regions NFR This allows for promoter DNA accessibility to various proteins such as transcription factors Nucleosome free region typically spans for 200 nucleotides in S cerevisiae 43 Well positioned nucleosomes form boundaries of NFR These nucleosomes are called 1 nucleosome and 1 nucleosome and are located at canonical distances downstream and upstream respectively from transcription start site 44 1 nucleosome and several downstream nucleosomes also tend to incorporate H2A Z histone variant 44 Modulating nucleosome structure editEukaryotic genomes are ubiquitously associated into chromatin however cells must spatially and temporally regulate specific loci independently of bulk chromatin In order to achieve the high level of control required to co ordinate nuclear processes such as DNA replication repair and transcription cells have developed a variety of means to locally and specifically modulate chromatin structure and function This can involve covalent modification of histones the incorporation of histone variants and non covalent remodelling by ATP dependent remodeling enzymes Histone post translational modifications edit nbsp Histone tails and their function in chromatin formationSince they were discovered in the mid 1960s histone modifications have been predicted to affect transcription 45 The fact that most of the early post translational modifications found were concentrated within the tail extensions that protrude from the nucleosome core lead to two main theories regarding the mechanism of histone modification The first of the theories suggested that they may affect electrostatic interactions between the histone tails and DNA to loosen chromatin structure Later it was proposed that combinations of these modifications may create binding epitopes with which to recruit other proteins 46 Recently given that more modifications have been found in the structured regions of histones it has been put forward that these modifications may affect histone DNA 47 and histone histone 48 interactions within the nucleosome core Modifications such as acetylation or phosphorylation that lower the charge of the globular histone core are predicted to loosen core DNA association the strength of the effect depends on location of the modification within the core 49 Some modifications have been shown to be correlated with gene silencing others seem to be correlated with gene activation Common modifications include acetylation methylation or ubiquitination of lysine methylation of arginine and phosphorylation of serine The information stored in this way is considered epigenetic since it is not encoded in the DNA but is still inherited to daughter cells The maintenance of a repressed or activated status of a gene is often necessary for cellular differentiation 18 Histone variants edit Although histones are remarkably conserved throughout evolution several variant forms have been identified This diversification of histone function is restricted to H2A and H3 with H2B and H4 being mostly invariant H2A can be replaced by H2AZ which leads to reduced nucleosome stability or H2AX which is associated with DNA repair and T cell differentiation whereas the inactive X chromosomes in mammals are enriched in macroH2A H3 can be replaced by H3 3 which correlates with activate genes and regulatory elements and in centromeres H3 is replaced by CENPA 18 ATP dependent nucleosome remodeling edit A number of distinct reactions are associated with the term ATP dependent chromatin remodeling Remodeling enzymes have been shown to slide nucleosomes along DNA 50 disrupt histone DNA contacts to the extent of destabilizing the H2A H2B dimer 51 52 and to generate negative superhelical torsion in DNA and chromatin 53 Recently the Swr1 remodeling enzyme has been shown to introduce the variant histone H2A Z into nucleosomes 54 At present it is not clear if all of these represent distinct reactions or merely alternative outcomes of a common mechanism What is shared between all and indeed the hallmark of ATP dependent chromatin remodeling is that they all result in altered DNA accessibility Studies looking at gene activation in vivo 55 and more astonishingly remodeling in vitro 56 have revealed that chromatin remodeling events and transcription factor binding are cyclical and periodic in nature While the consequences of this for the reaction mechanism of chromatin remodeling are not known the dynamic nature of the system may allow it to respond faster to external stimuli A recent study indicates that nucleosome positions change significantly during mouse embryonic stem cell development and these changes are related to binding of developmental transcription factors 57 Dynamic nucleosome remodelling across the Yeast genome edit Studies in 2007 have catalogued nucleosome positions in yeast and shown that nucleosomes are depleted in promoter regions and origins of replication 58 59 60 About 80 of the yeast genome appears to be covered by nucleosomes 61 and the pattern of nucleosome positioning clearly relates to DNA regions that regulate transcription regions that are transcribed and regions that initiate DNA replication 62 Most recently a new study examined dynamic changes in nucleosome repositioning during a global transcriptional reprogramming event to elucidate the effects on nucleosome displacement during genome wide transcriptional changes in yeast Saccharomyces cerevisiae 63 The results suggested that nucleosomes that were localized to promoter regions are displaced in response to stress like heat shock In addition the removal of nucleosomes usually corresponded to transcriptional activation and the replacement of nucleosomes usually corresponded to transcriptional repression presumably because transcription factor binding sites became more or less accessible respectively In general only one or two nucleosomes were repositioned at the promoter to effect these transcriptional changes However even in chromosomal regions that were not associated with transcriptional changes nucleosome repositioning was observed suggesting that the covering and uncovering of transcriptional DNA does not necessarily produce a transcriptional event After transcription the rDNA region has to protected from any damage it suggested HMGB proteins play a major role in protecting the nucleosome free region 64 65 DNA Twist Defects edit DNA twist defects are when the addition of one or a few base pairs from one DNA segment are transferred to the next segment resulting in a change of the DNA twist This will not only change the twist of the DNA but it will also change the length 66 This twist defect eventually moves around the nucleosome through the transferring of the base pair this means DNA twists can cause nucleosome sliding 67 Nucleosome crystal structures have shown that superhelix location 2 and 5 on the nucleosome are commonly found to be where DNA twist defects occur as these are common remodeler binding sites 68 There are a variety of chromatin remodelers but all share the existence of an ATPase motor which facilitates chromatin sliding on DNA through the binding and hydrolysis of ATP 69 ATPase has an open and closed state When the ATPase motor is changing from open and closed states the DNA duplex changes geometry and exhibits base pair tilting 68 The initiation of the twist defects via the ATPase motor causes tension to accumulate around the remodeler site The tension is released when the sliding of DNA has been completed throughout the nucleosome via the spread of two twist defects one on each strand in opposite directions 69 Nucleosome assembly in vitro edit nbsp Diagram of nucleosome assembly Nucleosomes can be assembled in vitro by either using purified native or recombinant histones 70 71 One standard technique of loading the DNA around the histones involves the use of salt dialysis A reaction consisting of the histone octamers and a naked DNA template can be incubated together at a salt concentration of 2 M By steadily decreasing the salt concentration the DNA will equilibrate to a position where it is wrapped around the histone octamers forming nucleosomes In appropriate conditions this reconstitution process allows for the nucleosome positioning affinity of a given sequence to be mapped experimentally 72 Disulfide crosslinked nucleosome core particles edit A recent advance in the production of nucleosome core particles with enhanced stability involves site specific disulfide crosslinks 73 Two different crosslinks can be introduced into the nucleosome core particle A first one crosslinks the two copies of H2A via an introduced cysteine N38C resulting in histone octamer which is stable against H2A H2B dimer loss during nucleosome reconstitution A second crosslink can be introduced between the H3 N terminal histone tail and the nucleosome DNA ends via an incorporated convertible nucleotide 74 The DNA histone octamer crosslink stabilizes the nucleosome core particle against DNA dissociation at very low particle concentrations and at elevated salt concentrations Nucleosome assembly in vivo edit nbsp Steps in nucleosome assemblyNucleosomes are the basic packing unit of genomic DNA built from histone proteins around which DNA is coiled They serve as a scaffold for formation of higher order chromatin structure as well as for a layer of regulatory control of gene expression Nucleosomes are quickly assembled onto newly synthesized DNA behind the replication fork H3 and H4 edit Histones H3 and H4 from disassembled old nucleosomes are kept in the vicinity and randomly distributed on the newly synthesized DNA 75 They are assembled by the chromatin assembly factor 1 CAF 1 complex which consists of three subunits p150 p60 and p48 76 Newly synthesized H3 and H4 are assembled by the replication coupling assembly factor RCAF RCAF contains the subunit Asf1 which binds to newly synthesized H3 and H4 proteins 77 The old H3 and H4 proteins retain their chemical modifications which contributes to the passing down of the epigenetic signature The newly synthesized H3 and H4 proteins are gradually acetylated at different lysine residues as part of the chromatin maturation process 78 It is also thought that the old H3 and H4 proteins in the new nucleosomes recruit histone modifying enzymes that mark the new histones contributing to epigenetic memory H2A and H2B edit In contrast to old H3 and H4 the old H2A and H2B histone proteins are released and degraded therefore newly assembled H2A and H2B proteins are incorporated into new nucleosomes 79 H2A and H2B are assembled into dimers which are then loaded onto nucleosomes by the nucleosome assembly protein 1 NAP 1 which also assists with nucleosome sliding 80 The nucleosomes are also spaced by ATP dependent nucleosome remodeling complexes containing enzymes such as Isw1 Ino80 and Chd1 and subsequently assembled into higher order structure 81 82 Gallery edit nbsp nbsp nbsp The crystal structure of the nucleosome core particle PDB 1EQZ 28 different views showing details of histone folding and organization Histones H2A H2B H3 H4 and DNA are coloured See also editChromomereReferences edit Reece J Campbell N 2006 Biology San Francisco Benjamin Cummings ISBN 978 0 8053 6624 2 Alberts B 2002 Molecular biology of the cell 4th ed New York Garland Science p 207 ISBN 978 0 8153 4072 0 lberts B Johnson A Lewis J Raff M Roberts K Walter P 2002 Chromosomal DNA and Its Packaging in the Chromatin Fiber Teif VB Clarkson CT 2019 Nucleosome Positioning Encyclopedia of Bioinformatics and Computational Biology 2 308 317 doi 10 1016 B978 0 12 809633 8 20242 2 ISBN 9780128114322 S2CID 43929234 Olins AL Olins DE January 1974 Spheroid chromatin units v bodies Science 183 4122 330 332 Bibcode 1974Sci 183 330O doi 10 1126 science 183 4122 330 PMID 4128918 S2CID 83480762 McDonald D December 2005 Milestone 9 1973 1974 The nucleosome hypothesis An alternative string theory Nature Milestones Gene Expression doi 10 1038 nrm1798 Kornberg RD May 1974 Chromatin structure a repeating unit of histones and DNA Science 184 4139 868 871 Bibcode 1974Sci 184 868K doi 10 1126 science 184 4139 868 PMID 4825889 Lorch Y LaPointe JW Kornberg RD April 1987 Nucleosomes inhibit the initiation of transcription but allow chain elongation with the displacement of histones Cell 49 2 203 210 doi 10 1016 0092 8674 87 90561 7 PMID 3568125 S2CID 21270171 Han M Grunstein M December 1988 Nucleosome loss activates yeast downstream promoters in vivo Cell 55 6 1137 1145 doi 10 1016 0092 8674 88 90258 9 PMID 2849508 S2CID 41520634 Clark Adams CD Norris D Osley MA Fassler JS Winston F February 1988 Changes in histone gene dosage alter transcription in yeast Genes amp Development 2 2 150 159 doi 10 1101 gad 2 2 150 PMID 2834270 a b In different crystals values of 146 and 147 basepairs were observed a b Luger K Mader AW Richmond RK Sargent DF Richmond TJ September 1997 Crystal structure of the nucleosome core particle at 2 8 A resolution Nature 389 6648 251 260 Bibcode 1997Natur 389 251L doi 10 1038 38444 PMID 9305837 S2CID 4328827 Alberts B 2007 Molecular Biology of the Cell 5th ed New York Garland Science p 211 ISBN 978 0 8153 4106 2 Shtumpf M Piroeva KV Agrawal SP Jacob DR Teif VB June 2022 NucPosDB a database of nucleosome positioning in vivo and nucleosomics of cell free DNA Chromosoma 131 1 2 19 28 doi 10 1007 s00412 021 00766 9 PMC 8776978 PMID 35061087 Zhou YB Gerchman SE Ramakrishnan V Travers A Muyldermans S September 1998 Position and orientation of the globular domain of linker histone H5 on the nucleosome Nature 395 6700 402 405 Bibcode 1998Natur 395 402Z doi 10 1038 26521 PMID 9759733 S2CID 204997317 Thoma F Koller T Klug A November 1979 Involvement of histone H1 in the organization of the nucleosome and of the salt dependent superstructures of chromatin The Journal of Cell Biology 83 2 Pt 1 403 427 doi 10 1083 jcb 83 2 403 PMC 2111545 PMID 387806 Clarke HJ 1992 Nuclear and chromatin composition of mammalian gametes and early embryos Biochemistry and Cell Biology 70 10 11 856 866 doi 10 1139 o92 134 PMID 1297351 a b c d e f Felsenfeld G Groudine M January 2003 Controlling the double helix Nature 421 6921 448 453 Bibcode 2003Natur 421 448F doi 10 1038 nature01411 PMID 12540921 Richmond TJ Finch JT Rushton B Rhodes D Klug A 1984 Structure of the nucleosome core particle at 7 A resolution Nature 311 5986 532 7 Bibcode 1984Natur 311 532R doi 10 1038 311532a0 PMID 6482966 S2CID 4355982 Harp JM Palmer EL York MH Gewiess A Davis M Bunick GJ October 1995 Preparative separation of nucleosome core particles containing defined sequence DNA in multiple translational phases Electrophoresis 16 10 1861 1864 doi 10 1002 elps 11501601305 PMID 8586054 S2CID 20178479 Palmer EL Gewiess A Harp JM York MH Bunick GJ October 1995 Large scale production of palindrome DNA fragments Analytical Biochemistry 231 1 109 114 doi 10 1006 abio 1995 1509 PMID 8678288 Harp JM Uberbacher EC Roberson AE Palmer EL Gewiess A Bunick GJ March 1996 X ray diffraction analysis of crystals containing twofold symmetric nucleosome core particles Acta Crystallographica Section D Biological Crystallography 52 Pt 2 283 288 doi 10 1107 S0907444995009139 PMID 15299701 Harp JM Hanson BL Timm DE Bunick GJ December 2000 Asymmetries in the nucleosome core particle at 2 5 A resolution Acta Crystallographica Section D Biological Crystallography 56 Pt 12 1513 1534 doi 10 1107 s0907444900011847 PMID 11092917 Hanson BL Alexander C Harp JM Bunick GJ 2004 Preparation and crystallization of nucleosome core particle Chromatin and Chromatin Remodeling Enzymes Part A Methods in Enzymology Vol 375 pp 44 62 doi 10 1016 s0076 6879 03 75003 4 ISBN 9780121827793 PMID 14870658 a b Chakravarthy S Park YJ Chodaparambil J Edayathumangalam RS Luger K February 2005 Structure and dynamic properties of nucleosome core particles FEBS Letters 579 4 895 898 doi 10 1016 j febslet 2004 11 030 PMID 15680970 S2CID 41706403 White CL Suto RK Luger K September 2001 Structure of the yeast nucleosome core particle reveals fundamental changes in internucleosome interactions The EMBO Journal 20 18 5207 5218 doi 10 1093 emboj 20 18 5207 PMC 125637 PMID 11566884 a b Stryer L 1995 Biochemistry fourth ed New York Basingstoke W H Freeman and Company ISBN 978 0716720096 a b Harp JM Hanson BL Timm DE Bunick GJ December 2000 Asymmetries in the nucleosome core particle at 2 5 A resolution Acta Crystallographica Section D Biological Crystallography 56 Pt 12 1513 1534 doi 10 1107 S0907444900011847 PMID 11092917 PDB ID 1EQZ Alberts B 2009 Essential Cell Biology 2nd ed New York Garland Science Allen Paul D Newland Adrian C 1 June 1998 Electrophoretic DNA analysis for the detection of apoptosis Molecular Biotechnology 9 3 247 251 doi 10 1007 BF02915798 ISSN 1559 0305 Davey CA Sargent DF Luger K Maeder AW Richmond TJ June 2002 Solvent mediated interactions in the structure of the nucleosome core particle at 1 9 a resolution Journal of Molecular Biology 319 5 1097 1113 doi 10 1016 S0022 2836 02 00386 8 PMID 12079350 Segal E Fondufe Mittendorf Y Chen L Thastrom A Field Y Moore IK et al August 2006 A genomic code for nucleosome positioning Nature 442 7104 772 778 Bibcode 2006Natur 442 772S doi 10 1038 nature04979 PMC 2623244 PMID 16862119 Zheng C Hayes JJ April 2003 Structures and interactions of the core histone tail domains Biopolymers 68 4 539 546 doi 10 1002 bip 10303 PMID 12666178 Schalch T Duda S Sargent DF Richmond TJ July 2005 X ray structure of a tetranucleosome and its implications for the chromatin fibre Nature 436 7047 138 141 Bibcode 2005Natur 436 138S doi 10 1038 nature03686 PMID 16001076 S2CID 4387396 Robinson PJ Fairall L Huynh VA Rhodes D April 2006 EM measurements define the dimensions of the 30 nm chromatin fiber evidence for a compact interdigitated structure Proceedings of the National Academy of Sciences of the United States of America 103 17 6506 6511 Bibcode 2006PNAS 103 6506R doi 10 1073 pnas 0601212103 PMC 1436021 PMID 16617109 Teif VB Rippe K September 2009 Predicting nucleosome positions on the DNA combining intrinsic sequence preferences and remodeler activities Nucleic Acids Research 37 17 5641 5655 doi 10 1093 nar gkp610 PMC 2761276 PMID 19625488 Pennings S Muyldermans S Meersseman G Wyns L May 1989 Formation stability and core histone positioning of nucleosomes reassembled on bent and other nucleosome derived DNA Journal of Molecular Biology 207 1 183 192 doi 10 1016 0022 2836 89 90449 X PMID 2738923 Fu Y Sinha M Peterson CL Weng Z July 2008 Van Steensel B ed 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 Bargaje R Alam MP Patowary A Sarkar M Ali T Gupta S et al October 2012 Proximity of H2A Z containing nucleosome to the transcription start site influences gene expression levels in the mammalian liver and brain Nucleic Acids Research 40 18 8965 8978 doi 10 1093 nar gks665 PMC 3467062 PMID 22821566 a b Li G Levitus M Bustamante C Widom J January 2005 Rapid spontaneous accessibility of nucleosomal DNA Nature Structural amp Molecular Biology 12 1 46 53 doi 10 1038 nsmb869 PMID 15580276 S2CID 14540078 Li G Widom J August 2004 Nucleosomes facilitate their own invasion Nature Structural amp Molecular Biology 11 8 763 769 doi 10 1038 nsmb801 PMID 15258568 S2CID 11299024 Hodges C Bintu L Lubkowska L Kashlev M Bustamante C July 2009 Nucleosomal fluctuations govern the transcription dynamics of RNA polymerase II Science 325 5940 626 628 Bibcode 2009Sci 325 626H doi 10 1126 science 1172926 PMC 2775800 PMID 19644123 Yuan GC Liu YJ Dion MF Slack MD Wu LF Altschuler SJ Rando OJ July 2005 Genome scale identification of nucleosome positions in S cerevisiae Science 309 5734 626 630 Bibcode 2005Sci 309 626Y doi 10 1126 science 1112178 PMID 15961632 S2CID 43625066 a b Lai WK Pugh BF September 2017 Understanding nucleosome dynamics and their links to gene expression and DNA replication Nature Reviews Molecular Cell Biology 18 9 548 562 doi 10 1038 nrm 2017 47 PMC 5831138 PMID 28537572 Allfrey VG Faulkner R Mirsky AE May 1964 Acetylation and Methylation of Histones and Their Possible Role in the Regulation of RNA Synthesis Proceedings of the National Academy of Sciences of the United States of America 51 5 786 794 Bibcode 1964PNAS 51 786A doi 10 1073 pnas 51 5 786 PMC 300163 PMID 14172992 Strahl BD Allis CD January 2000 The language of covalent histone modifications Nature 403 6765 41 45 Bibcode 2000Natur 403 41S doi 10 1038 47412 PMID 10638745 S2CID 4418993 Cosgrove MS Boeke JD Wolberger C November 2004 Regulated nucleosome mobility and the histone code Nature Structural amp Molecular Biology 11 11 1037 1043 doi 10 1038 nsmb851 PMID 15523479 S2CID 34704745 Ye J Ai X Eugeni EE Zhang L Carpenter LR Jelinek MA et al April 2005 Histone H4 lysine 91 acetylation a core domain modification associated with chromatin assembly Molecular Cell 18 1 123 130 doi 10 1016 j molcel 2005 02 031 PMC 2855496 PMID 15808514 Fenley AT Adams DA Onufriev AV September 2010 Charge state of the globular histone core controls stability of the nucleosome Biophysical Journal 99 5 1577 1585 Bibcode 2010BpJ 99 1577F doi 10 1016 j bpj 2010 06 046 PMC 2931741 PMID 20816070 Whitehouse I Flaus A Cairns BR White MF Workman JL Owen Hughes T August 1999 Nucleosome mobilization catalysed by the yeast SWI SNF complex Nature 400 6746 784 787 Bibcode 1999Natur 400 784W doi 10 1038 23506 PMID 10466730 S2CID 2841873 Kassabov SR Zhang B Persinger J Bartholomew B February 2003 SWI SNF unwraps slides and rewraps the nucleosome Molecular Cell 11 2 391 403 doi 10 1016 S1097 2765 03 00039 X PMID 12620227 Bruno M Flaus A Stockdale C Rencurel C Ferreira H Owen Hughes T December 2003 Histone H2A H2B dimer exchange by ATP dependent chromatin remodeling activities Molecular Cell 12 6 1599 1606 doi 10 1016 S1097 2765 03 00499 4 PMC 3428624 PMID 14690611 Havas K Flaus A Phelan M Kingston R Wade PA Lilley DM Owen Hughes T December 2000 Generation of superhelical torsion by ATP dependent chromatin remodeling activities Cell 103 7 1133 1142 doi 10 1016 S0092 8674 00 00215 4 PMID 11163188 S2CID 7911590 Mizuguchi G Shen X Landry J Wu WH Sen S Wu C January 2004 ATP driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex Science 303 5656 343 348 Bibcode 2004Sci 303 343M doi 10 1126 science 1090701 PMID 14645854 S2CID 9881829 Metivier R Penot G Hubner MR Reid G Brand H Kos M Gannon F December 2003 Estrogen receptor alpha directs ordered cyclical and combinatorial recruitment of cofactors on a natural target promoter Cell 115 6 751 763 doi 10 1016 S0092 8674 03 00934 6 PMID 14675539 S2CID 145525 Nagaich AK Walker DA Wolford R Hager GL April 2004 Rapid periodic binding and displacement of the glucocorticoid receptor during chromatin remodeling Molecular Cell 14 2 163 174 doi 10 1016 S1097 2765 04 00178 9 PMID 15099516 Teif VB Vainshtein Y Caudron Herger M Mallm JP Marth C Hofer T Rippe K November 2012 Genome wide nucleosome positioning during embryonic stem cell development Nature Structural amp Molecular Biology 19 11 1185 1192 doi 10 1038 nsmb 2419 PMID 23085715 S2CID 34509771 Albert I Mavrich TN Tomsho LP Qi J Zanton SJ Schuster SC Pugh BF March 2007 Translational and rotational settings of H2A Z nucleosomes across the Saccharomyces cerevisiae genome Nature 446 7135 572 576 Bibcode 2007Natur 446 572A doi 10 1038 nature05632 PMID 17392789 S2CID 4416890 Li B Carey M Workman JL February 2007 The role of chromatin during transcription Cell 128 4 707 719 doi 10 1016 j cell 2007 01 015 PMID 17320508 S2CID 1773333 Whitehouse I Rando OJ Delrow J Tsukiyama T December 2007 Chromatin remodelling at promoters suppresses antisense transcription Nature 450 7172 1031 1035 Bibcode 2007Natur 450 1031W doi 10 1038 nature06391 PMID 18075583 S2CID 4305576 Lee W Tillo D Bray N Morse RH Davis RW Hughes TR Nislow C October 2007 A high resolution atlas of nucleosome occupancy in yeast Nature Genetics 39 10 1235 1244 doi 10 1038 ng2117 PMID 17873876 S2CID 12816925 Eaton ML Galani K Kang S Bell SP MacAlpine DM April 2010 Conserved nucleosome positioning defines replication origins Genes amp Development 24 8 748 753 doi 10 1101 gad 1913210 PMC 2854390 PMID 20351051 Shivaswamy S Bhinge A Zhao Y Jones S Hirst M Iyer VR March 2008 Dynamic remodeling of individual nucleosomes across a eukaryotic genome in response to transcriptional perturbation PLOS Biology 6 3 e65 doi 10 1371 journal pbio 0060065 PMC 2267817 PMID 18351804 Murugesapillai D McCauley MJ Huo R Nelson Holte MH Stepanyants A Maher LJ et al August 2014 DNA bridging and looping by HMO1 provides a mechanism for stabilizing nucleosome free chromatin Nucleic Acids Research 42 14 8996 9004 doi 10 1093 nar gku635 PMC 4132745 PMID 25063301 Murugesapillai D McCauley MJ Maher LJ Williams MC February 2017 Single molecule studies of high mobility group B architectural DNA bending proteins Biophysical Reviews 9 1 17 40 doi 10 1007 s12551 016 0236 4 PMC 5331113 PMID 28303166 Winger J Nodelman IM Levendosky RF Bowman GD May 2018 A twist defect mechanism for ATP dependent translocation of nucleosomal DNA eLife 7 e34100 doi 10 7554 eLife 34100 PMC 6031429 PMID 29809147 Bowman GD August 2019 Uncovering a New Step in Sliding Nucleosomes Trends in Biochemical Sciences 44 8 643 645 doi 10 1016 j tibs 2019 05 001 PMC 7092708 PMID 31171402 a b Nodelman IM Bowman GD May 2021 Biophysics of Chromatin Remodeling Annual Review of Biophysics 50 1 73 93 doi 10 1146 annurev biophys 082520 080201 PMC 8428145 PMID 33395550 a b Brandani GB Takada S November 2018 Onufriev A ed Chromatin remodelers couple inchworm motion with twist defect formation to slide nucleosomal DNA PLOS Computational Biology 14 11 e1006512 Bibcode 2018PLSCB 14E6512B doi 10 1371 journal pcbi 1006512 PMC 6237416 PMID 30395604 Hayes JJ Lee KM May 1997 In vitro reconstitution and analysis of mononucleosomes containing defined DNAs and proteins Methods 12 1 2 9 doi 10 1006 meth 1997 0441 PMID 9169189 Dyer PN Edayathumangalam RS White CL Bao Y Chakravarthy S Muthurajan UM Luger K 2004 Reconstitution of nucleosome core particles from recombinant histones and DNA Chromatin and Chromatin Remodeling Enzymes Part A Methods in Enzymology Vol 375 pp 23 44 doi 10 1016 s0076 6879 03 75002 2 ISBN 9780121827793 PMID 14870657 Yenidunya A Davey C Clark D Felsenfeld G Allan J April 1994 Nucleosome positioning on chicken and human globin gene promoters in vitro Novel mapping techniques Journal of Molecular Biology 237 4 401 414 doi 10 1006 jmbi 1994 1243 PMID 8151701 Frouws TD Barth PD Richmond TJ January 2018 Site Specific Disulfide Crosslinked Nucleosomes with Enhanced Stability Journal of Molecular Biology 430 1 45 57 doi 10 1016 j jmb 2017 10 029 PMC 5757783 PMID 29113904 Ferentz AE Verdine GL 1994 The Convertible Nucleoside Approach Structural Engineering of Nucleic Acids by Disulfide Cross Linking In Eckstein F Lilley DM eds Nucleic Acids and Molecular Biology Vol 8 pp 14 40 doi 10 1007 978 3 642 78666 2 2 ISBN 978 3 642 78668 6 Yamasu K Senshu T January 1990 Conservative segregation of tetrameric units of H3 and H4 histones during nucleosome replication Journal of Biochemistry 107 1 15 20 doi 10 1093 oxfordjournals jbchem a122999 PMID 2332416 Kaufman PD Kobayashi R Kessler N Stillman B June 1995 The p150 and p60 subunits of chromatin assembly factor I a molecular link between newly synthesized histones and DNA replication Cell 81 7 1105 1114 doi 10 1016 S0092 8674 05 80015 7 PMID 7600578 S2CID 13502921 Tyler JK Adams CR Chen SR Kobayashi R Kamakaka RT Kadonaga JT December 1999 The RCAF complex mediates chromatin assembly during DNA replication and repair Nature 402 6761 555 560 Bibcode 1999Natur 402 555T doi 10 1038 990147 PMID 10591219 S2CID 205097512 Benson LJ Gu Y Yakovleva T Tong K Barrows C Strack CL et al April 2006 Modifications of H3 and H4 during chromatin replication nucleosome assembly and histone exchange The Journal of Biological Chemistry 281 14 9287 9296 doi 10 1074 jbc M512956200 PMID 16464854 Louters L Chalkley R June 1985 Exchange of histones H1 H2A and H2B in vivo Biochemistry 24 13 3080 3085 doi 10 1021 bi00334a002 PMID 4027229 Park YJ Chodaparambil JV Bao Y McBryant SJ Luger K January 2005 Nucleosome assembly protein 1 exchanges histone H2A H2B dimers and assists nucleosome sliding The Journal of Biological Chemistry 280 3 1817 1825 doi 10 1074 jbc M411347200 PMID 15516689 Vincent JA Kwong TJ Tsukiyama T May 2008 ATP dependent chromatin remodeling shapes the DNA replication landscape Nature Structural amp Molecular Biology 15 5 477 484 doi 10 1038 nsmb 1419 PMC 2678716 PMID 18408730 Yadav T Whitehouse I April 2016 Replication Coupled Nucleosome Assembly and Positioning by ATP Dependent Chromatin Remodeling Enzymes Cell Reports 15 4 715 723 doi 10 1016 J CELREP 2016 03 059 PMC 5063657 PMID 27149855 External links editMBInfo What are nucleosomes Nucleosomes on the Richmond Lab website Proteopedia Nucleosomes Nucleosome at the PDB Dynamic Remodeling of Individual Nucleosomes Across a Eukaryotic Genome in Response to Transcriptional Perturbation Nucleosome positioning data and tools online annotated list constantly updated Histone protein structure HistoneDB 2 0 Database of histones and variants at NCBI Retrieved from https en wikipedia org w index php title Nucleosome amp oldid 1213707675, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.