Histone H1


DEVELOPMENTAL BIOLOGY

Embryonic

The first appearance of H1 takes place during cycles 7 and 8. It is during cycle 7 that the size of the nuclei begins to decrease. By cycles 10-12 a sufficient amount of H1 has accumulated to allow the reorganization of chromatin to a transcriptionally active state. Subsequently, increased zygotic transcription elevates H1 levels even further. This exponential increase, together with the increased number of nuclei, rapidly depletes the relative levels of HMG-D protein. It has been suggested that the chromatin generated in the presence of HMG-D is transcriptionally silent, and that transcription begins only when H1 levels have reached a particular threshold value and overcome the HMG-D effects, around nuclear cycle 10 (Ner, 1994).

The amount of histone H1 relative to core histones (See Histone H4) has been determined in three Drosophila species (D. melanogaster, D. texana and D. virilis) in chromatin from several tissues differing in chromatin structure and genetic activity. Low levels of H1 are found in relatively undifferentiated, early embryos as well as in a line of cultured cells. In late embryos the content of H1 is highest in D. virilis, which possesses larger amounts of and a partially more compacted constitutive heterochromatin than the two other species. Polytene chromatin from larval salivary glands shows increased levels of H1 compared with diploid chromatin; the degree of phosphorylation of this histone is relatively low. The degree of phosphorylation of H2A is found to be drastically reduced in polytene as compared with diploid embryonic chromatin, which parallels the extensive underreplication of constitutive heterochromatin. In diploid chromatin, a qualitative correlation is also observed between the relative amounts of heterochromatin and the levels of H2A phosphorylation. These findings suggest a connection between H2A phosphorylation and heavy compaction of interphase chromatin (Holmgren, 1985).

HMG-D is an abundant chromosomal protein associated with condensed chromatin during the first nuclear cleavage cycles of the developing Drosophila embryo. It previously suggested that HMG-D might substitute for the linker histone H1 in the preblastoderm embryo and that this substitution might result in the characteristic less compacted chromatin. The association of HMG-D with chromatin has been studied using a cell-free system for chromatin reconstitution derived from Drosophila embryos. Association of HMG-D with chromatin, like that of histone H1, increases the nucleosome spacing indicative of binding to the linker DNA between nucleosomes. HMG-D interacts with DNA during the early phases of nucleosome assembly but is gradually displaced as chromatin matures. By contrast, purified chromatin can be loaded with stoichiometric amounts of HMG-D, and this can be displaced upon addition of histone H1. A direct physical interaction between HMG-D and histone H1 was observed in a Far Western analysis. The competitive nature of this interaction is reminiscent of the apparent replacement of HMG-D by H1 during mid-blastula transition. These data are consistent with the hypothesis that HMG-D functions as a specialized linker protein prior to appearance of histone H1 (Ner, 2001).

Histone H1 and HMGB1 proteins could influence chromatin structure in a similar manner by binding to linker DNA sequence. Histone H1 associates with linker DNA sequences and organizes nucleosomal arrays into higher order chromatin structures, such as the 30-nm chromatin fiber. However, little is known about how HMGB1 interacts with the nucleosome and about the consequences in structure and function. H1 and HMGB1 share important features; both protect linker DNA sequences from nuclease digestion, and both bind four-way junctions. Consistent with the idea that interaction of HMGB1 might replace histone H1, in the very early stages of Drosophila embryogenesis histone H1 is absent, but the high mobility group protein D (HMG-D) is present in vast excess. Based on the similarities between HMG-D and H1, a role for HMG-D as a linker protein compatible with and perhaps required for the fast condensation-decondensation cycles associated with the very rapid nuclear division cycles found in preblastoderm embryos has been suggested. An analogous role has been proposed for the Xenopus HMGB1 and B4 proteins; both proteins have been demonstrated to bind di-nucleosomal DNA (Ner, 2001).

The fact that recombinant HMG-D increases the nucleosome repeat length (NRL) in a cell-free chromatin assembly system strongly supports this hypothesis. The NRL is strongly dependent on the ionic environment such that polycations are particularly effective in increasing the average separation between adjacent nucleosomes. In accordance with these findings the data implicate the polycationic basic region (residues 85-99; net charge, +10) of HMG-D in this function. However, the HMG-D-dependent increase in NRL is mediated both by the full-length protein and by HMG-D100. These forms differ substantially in net charge +7 (for HMG-D) and +17 (for HMG-D100), suggesting that the chromatin DNA can compete effectively with the polyanionic acidic tail of HMG-D. Histone H1 and the HMGN1 and HMGN2 proteins (formerly HMG-14 and HMG-17) are the only other proteins reported to cause such a change, in the case of H1 presumably by binding to the linker DNA. The binding of H1 to the linker sequence appears to differ from that of HMG-D. Increasing concentrations of histone H1 added to the assembly reaction will continue to increase the NRL to well over 220 bp before the regular nucleosomal array is lost. HMG-D, on the other hand, increases the NRL to only ~180 bp. This may reflect the stoichiometry of binding to the linker sequence. Di-nucleosomal DNA reconstituted by dialysis has been shown to be able to bind two molecules of H1 but only a single molecule of HMGB1. Although the exact nature of the binding remains unknown, HMG-D binds ~14 bp of DNA, and consequently 1-2 molecules of HMG-D could potentially occupy the linker space (Ner, 2001).

A tight correlation between nucleosome spacing and the folding of the nucleosomal fiber into a 30-nm fiber has been observed, which led to the suggestion that different NRLs would correspond to particular fiber geometries and, therefore, compaction states. Accordingly, increased nucleosome spacing is indicative of more compacted chromatin. The observation that HMG-D does not increase the NRL beyond 185 bp as H1 may indicate that HMG-D-containing chromatin is folded but is less compacted (Ner, 2001).

Like other HMG domain proteins such as LEF-1 and SRY, HMG-D can introduce sharp bends or kinks into DNA. The current estimates of the magnitude of the DNA kinks induced by HMG-D range from 100-120° for the full-length protein to 60° to >90° for HMG-D100. These values are substantially greater than the average curvature of DNA wrapped around the histone octamer and indicate that HMG-D bound DNA is not smoothly curved. In the context of linker DNA, such a state would be consistent with both the lack of UV-induced thymine dimer formation in the linker and also, with evidence from electric dichroism studies, that the trajectory of linker DNA differs from that of DNA bound to the core histones. Of particular relevance are the observations that, in the presence of histone H1 derivatives containing a major proportion of the basic C-terminal domain, the linker DNA enters and leaves a single chromatosome as a straight rod approximately perpendicular to the superhelical axis. A similar structure has been observed in chromatin fibers. This organization implies that the DNA must bend sharply as it enters and leaves the chromatosome. A possible role for HMG-D would be to induce such sharp bends by kinking the DNA and thereby promoting a higher level of chromatin folding (Ner, 2001).

Evidence has been provided for an interaction of HMGB1 with the nucleosome and it has been suggested that it might replace histone H1 in the nucleosome. Evidence has been provided for interactions between histone H1 and HMGB1. The results are consistent with these observations. (1) In a Far Western analysis, H1 is the predominant protein identified when labeled HMG-D was used as a probe. (2) Using chromatin assembled on DNA attached to paramagnetic beads and preloaded with HMG-D protein, HMG-D is displaced upon titration of histone H1. It is noted that the full-length HMG-D and HMG-D100 both interact with H1 in a Far Western analysis. The alanine-lysine-rich region (amino acids 84-100, AKKRAKPAKKVAKKSKK) is very similar to a region found in histone H1. Far Western analysis suggests that this region, or possibly the region immediately preceding glycine-rich linker, is interacting with H1. In HMG-D this sequence contains a serine residue that is phosphorylated by casein kinase II.

Although it is possible to argue for a structural role for HMG-D and HMGB1 in early embryonic chromatin, in vitro observations show that in the absence of H1 HMG-D, although initially present at high levels, is displaced to below 1 molecule/10-20 nucleosomes as the reaction proceeds and the chromatin matures. This would argue against a purely structural role for HMG-D and suggest that the protein may fulfill a different role. One possibility is that HMG-D functions as a chaperone molecule and preconfigures the DNA to facilitate the chromatin assembly process. HMG-D could participate to bend the DNA at the exit and entry points to the nucleosome, and this bend is then stabilized by histone H1. Under such a scenario, as chromatin assembly proceeds and the core histones are recruited, HMG-D molecules are displaced. The linker sequences would be the only locations where the protein would persist for longer duration. However, this too would be displaced on the addition of other chromatin-associated proteins (transcription factors, assembly factors). Such a mechanism would be very similar to that proposed for the recruitment of transcription factors. Similarly the displacement and competition with histone H1 can be envisaged as part of a process in which the DNA is kinked by HMG-D, and then the binding of the linker histone stabilizes this kink (Ner, 2001).

Preblastoderm embryonic chromatin clearly differs profoundly from post-blastoderm chromatin. Early syncytial nuclei are much larger and contain chromatin that is less compacted than later nuclei. In the early embryo HMG-D is highly abundant, although not all molecules are necessarily available for DNA binding. It is deposited in the egg by the mother but thereafter is maintained at an approximately constant level per embryo. Consequently, with each nuclear division the average number of HMG-D molecules per nucleus falls, although during nuclear cycles 7-14 the amount of H1 rapidly increases. Only during cycle 7 does the size of the nuclei begin to decrease. By cycles 10-12 a sufficient amount of histone H1 has accumulated to allow the reorganization of chromatin to a transcriptionally active state. Subsequently, increased zygotic transcription elevates histone H1 levels further. This exponential increase of histone H1 together with the increasing number of nuclei rapidly deplete HMG-D protein to levels that cannot have global effects on chromatin structure. What could be the physiological significance of different linker proteins? HMG-D- or H1-containing chromatin may differ profoundly in the degree or mode of compaction. The looser structure formed in the absence of H1 could facilitate the rapid condensation and decondensation required during the very short early cleavage cycles (Ner, 2001).

The switch from HMG-D- to H1-containing chromatin correlates with the acquisition of global transcriptional competence. Similar observations have been described in the Xenopus system in which B4, an H1 variant, and HMGB1 disappear during mid-blastula transition, again correlating with a change in the accessibility of embryonic chromatin to class III transcriptional machinery. The cell-free system employed in this study may facilitate the detailed analysis of this major switch in genome function during embryonic development (Ner, 2001).

Adult

A genomic fragment was cloned from a DNA library constructed from a Drosophila enhancer trap line in which reporter gene expression was observed at the anterior-most tip of the ovaries and testes. This genomic clone was identified as the L-repeat of the Drosophila melanogaster histone gene cluster. Northern blotting and in situ hybridization to RNA in tissues with individual cDNAs and PCR-generated probes for each histone confirm that gene expression is greatest at the anterior portion of each ovariole, in the germarium, and is also elevated in a few individual nurse cells and somatic follicle cells within the egg chamber during early developmental stages. Histone H1 and each of the core histones have a similar expression pattern that is correlated to cell division. Maternal stores of histone transcripts are also transported to the mature oocyte from the nurse cells at a later stage of oogenesis (stage 10), when virtually all the nurse cells contain high levels of histone transcripts. The results are consistent with expression of the somatic histone gene cluster during oogenesis as a coordinate unit. There does not seem to be a reduced level of somatic type H1 in the germ-line, as is observed in some other species. The relationship between the P[lacZ] expression pattern in the germarium and the overall expression of the histone cluster suggests there are specific regulatory elements for germ-line expression (Walker, 1998).


REFERENCES

Adenot, P. G., et al. (2000). Somatic linker histone H1 is present throughout mouse embryogenesis and is not replaced by variant H1°. J. Cell Sci. 113(Pt 16): 2897-2907. 10910774

Almouzni, G., et al. (1994). Histone acetylation influences both gene expression and development of Xenopus laevis. Dev. Biol. 165: 654-69. PubMed Citation: 7958429

Andreyeva, E. N., Bernardo, T. J., Kolesnikova, T. D., Lu, X., Yarinich, L. A., Bartholdy, B. A., Guo, X., Posukh, O. V., Healton, S., Willcockson, M. A., Pindyurin, A. V., Zhimulev, I. F., Skoultchi, A. I. and Fyodorov, D. V. (2017). Regulatory functions and chromatin loading dynamics of linker histone H1 during endoreplication in Drosophila. Genes Dev 31(6): 603-616. PubMed ID: 28404631

Barra, J. L., Rhounim, L., Rossignol, J. L. and Faugeron, G. (1999). Histone H1 is dispensable for methylation-associated gene silencing in Ascobolus immersus and essential for long life span. Mol. Cell. Biol. 20: 61-69. PubMed Citation: 10594009

Baxevanis, A. D. and Landsman, D. (1996). Histone Sequence Database: a compilation of highly-conserved nucleoprotein sequences. Nucleic Acids Res. 24: 245-247. PubMed Citation: 8594591

Becker, P. B. and Wu, C. (1992). Cell-Free system for assembly of transcriptionally repressed chromatin from Drosophila embryos. Mol. Cell. Biol. 12: 2241-49. PubMed Citation: 1569951

Bouvet, P., Dimitrov, S. and Wolffe, A. P. (1994). Specific regulation of Xenopus chromosomal 5S rRNA gene transcription in vivo by histone H1. Genes Dev. 8: 1147-1159. PubMed Citation: 7926720

Brown, E. J. and Bachtrog, D. (2014). The chromatin landscape of Drosophila: comparisons between species, sexes, and chromosomes. Genome Res 24(7):1125-37. PubMed ID: 24840603

Chadee, D. N., et al. (1995). Increased phosphorylation of histone H1 in mouse fibroblasts transformed with oncogenes or constitutively active mitogen-activated protein kinase kinase. J. Biol. Chem. 270: 20098-20105. PubMed Citation: 7650028

Chen, Z. H., Zhu, M., Yang, J., Liang, H., He, J., He, S., Wang, P., Kang, X., McNutt, M. A., Yin, Y. and Shen, W. H. (2014). PTEN interacts with histone H1 and controls chromatin condensation. Cell Rep 8: 2003-2014. PubMed ID: 25199838

Cheng, M. K. and Shearn, A. (2004). The direct interaction between Ash2, a Drosophila trithorax group protein, and Sktl, a nuclear phosphatidylinositol 4-phosphate 5-kinase, implies a role for phosphatidylinositol 4,5-bisphosphate in maintaining transcriptionally active chromatin. Genetics 167(3): 1213-23. 15280236

Clarke, H. J., Oblin, S. and Bustin, M. (1992). Developmental regulation of chromatin composition during mouse embryogenesis: somatic histone H1 is first detectable at the 4-cell stage. Development 115: 791-9. PubMed Citation: 1425354

Climent-Canto, P., Carbonell, A., Tatarski, M., Reina, O., Bujosa, P., Font-Mateu, J., Bernues, J., Beato, M. and Azorin, F. (2020). The embryonic linker histone dBigH1 alters the functional state of active chromatin. Nucleic Acids Res. PubMed ID: 32103264

Corona, D. F., et al. (2007). ISWI regulates higher-order chromatin structure and histone H1 assembly in vivo. PLoS Biol 5: e232. PubMed citation: 17760505

Dang, W., Kagalwala, M. N. and Bartholomew, B. (2006). Regulation of ISW2 by concerted action of histone H4 tail and extranucleosomal DNA. Mol. Cell Biol. 26: 7388-7396. PubMed citation: 17015471

De Santa, F., et al. (2007). The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell 130(6): 1083-94. Medline abstract: 17825402

Dimitrov, S., et al. (1993). Chromatin transitions during early Xenopus embryogenesis: changes in histone H4 acetylation and in linker histone type. Dev. Biol. 160: 214-27

Dimitrov, S. and Wolffe, A. P. (1996). Remodeling somatic nuclei in Xenopus laevis egg extracts: molecular mechanisms for the selective release of histones H1 and H1(0) from chromatin and the acquisition of transcriptional competence. EMBO J. 15 (21): 5897-5906.

Ding, H. F., Bustin, M. and Hansen, U. (1997). Alleviation of histone H1-mediated transcriptional repression and chromatin compaction by the acidic activation region in chromosomal protein HMG-14. Mol. Cell. Biol. 17(10): 5843-5855.

Dominski, Z., Yang, X.C., Purdy, M. and Marzluff, W. F. (2003). Cloning and characterization of the Drosophila U7 small nuclear RNA. Proc. Natl. Acad. Sci. 100(16): 9422-7. 12872004

Dominski, Z., Yang, X. C., Purdy, M. and Marzluff, W. F. (2005). Differences and similarities between Drosophila and mammalian 3' end processing of histone pre-mRNAs. RNA 11(12): 1835-47. 16251385

Dou, Y. L., Mizzen, C. A., Abrams, M., Allis, C. D. and Gorovsky, M. A. (1999). Phosphorylation of linker histone H1 regulates gene expression in vivo by mimicking H1 removal. Mol. Cell 4: 641-647

Dou, Y. and Gorovsky, M. A. (2000). Phosphorylation of linker histone H1 regulates gene expression in vivo by creating a charge patch. Molec. Cell 6: 225-231. 10983971

Fan, Y., Nikitina, T., Zhao, J., Fleury, T. J., Bhattacharyya, R., Bouhassira, E. E., Stein, A., Woodcock, C. L. and Skoultchi, A. I. (2005). Histone H1 depletion in mammals alters global chromatin structure but causes specific changes in gene regulation. Cell 123: 1199-1212. PubMed citation: 16377562

Filion, G. J., van Bemmel, J. G., Braunschweig, U., Talhout, W., Kind, J., Ward, L. D., Brugman, W., de Castro, I. J., Kerkhoven, R. M., Bussemaker, H. J. and van Steensel, B. (2010). Systematic protein location mapping reveals five principal chromatin types in Drosophila cells. Cell 143: 212-224. PubMed ID: 20888037

Gangaraju, V. K. and Bartholomew, B. (2007). Dependency of ISW1a chromatin remodeling on extranucleosomal DNA. Mol. Cell Biol. 27: 3217-3225. PubMed citation: 17283061

Gasser, R., Koller, T. and Sogo, J. M. (1996). The stability of nucleosomes at the replication fork. J. Mol. Biol. 258: 224-239

Goytisolo, F. A., et al. (1996). Identification of two DNA-binding sites on the globular domain of histone H5. EMBO J. 15: 3421-29

Godfrey, A. C., et al. (2006). U7 snRNA mutations in Drosophila block histone pre-mRNA processing and disrupt oogenesis. RNA 12: 396-409. 16495235

Guglielmi, B., La Rochelle, N. and Tjian, R. (2013). Gene-specific transcriptional mechanisms at the histone gene cluster revealed by single-cell imaging. Mol Cell 51: 480-492. PubMed ID: 23973376

Gunjan, A., and Brown, D.T. (1999a). Overproduction of histone H1 variants in vivo increases basal and induced activity of the mouse mammary tumor virus promoter. Nucleic Acids Res. 27: 3355-3363.

Gunjan, A., Alexander, B. T., Sittman, D. B. and Brown, D. T. (1999b). Effects of H1 histone variant overexpression on chromatin structure. J. Biol. Chem. 1274: 37950-37956. PubMed ID: 10608862

Halmer L. and Gruss, C. (1996). Effects of cell cycle dependent histone H1 phosphorylation on chromatin structure and chromatin replication. Nucleic Acids Res. 24: 1420-1427

Hassan, B. and Vaessin, H. (1997). Daughterless is required for the expression of cell cycle genes in peripheral nervous system precursors of Drosophila embryos. Dev. Genet. 21(2): 117-122.

Hayes, J. J. (1996). Site-directed cleavage of DNA by a linker histone-Fe(II) EDTA conjugate: Localization of a globular domain binding site within a nucleosome. Biochemistry 35: 11931-7

Holmgren, P., et al. (1985). Content of histone H1 and histone phosphorylation in relation to the higher order structures of chromatin in Drosophila. Chromosoma 93: 123-31

Hu, J., Gu, L., Ye, Y., Zheng, M., Xu, Z., Lin, J., Du, Y., Tian, M., Luo, L., Wang, B., Zhang, X., Weng, Z. and Jiang, C. (2018). Dynamic placement of the linker histone H1 associated with nucleosome arrangement and gene transcription in early Drosophila embryonic development. Cell Death Dis 9(7): 765. PubMed ID: 29988149  

Ito, S., Fujiyama-Nakamura, S., Kimura, S., Lim, J., Kamoshida, Y., Shiozaki-Sato, Y., Sawatsubashi, S., Suzuki, E., Tanabe, M., Ueda, T., Murata, T., Kato, H., Ohtake, F., Fujiki, R., Miki, T., Kouzmenko, A., Takeyama, K., Kato, S. (2012). Epigenetic Silencing of Core Histone Genes by HERS in Drosophila. Mol. Cell 45(4): 494-504. PubMed Citation: 22365829

Isogai, Y. et al. (2007). Transcription of histone gene cluster by differential core-promoter factors. Genes Dev. 21: 2936-49. PubMed Citation: 17978101

Kagalwala, M. N., Glaus, B. J., Dang, W., Zofall, M. and Bartholomew, B. (2004), Topography of the ISW2-nucleosome complex: insights into nucleosome spacing and chromatin remodeling. EMBO J 23: 2092-2104. PubMed citation: 15131696

Kaludov, N. K., et al. (1997). A mouse histone H1 variant, h1b, binds preferentially to a regulatory sequence within a mouse H3.2 replication-dependent histone gene. J. Biol. Chem. 272 (24): 15120-15127.

Kas, E., et al. (1993). A model for chromatin opening: stimulation of topoisomerase II and restriction enzyme cleavage of chromatin by distamycin. EMBO J. 12: 115-26

Kermekchiev, M., Workman, J. L. and Pikaard, C. S., (1997). Nucleosome binding by the polymerase I transactivator upstream binding factor displaces linker histone H1. Mol. Cell. Biol. 17(10): 5833-5842.

Kim, M. Y., et al. (2004). NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell 119: 803-814. 15607977

Kraus, W.L. and Lis, J.T. (2003). PARP goes transcription. Cell 113: 677-683. 12809599

Lee, N, Maurange, C., Ringrose, L. and Paro, R. (2005). Suppression of Polycomb group proteins by JNK signalling induces transdetermination in Drosophila imaginal discs. Nature 438: 234-237. Medline abstract: 16281037

Lemercier, C., et al. (2000). Involvement of retinoblastoma protein and HBP1 in histone H1(0) gene expression. Mol. Cell Biol. 20(18): 6627-37

Lifton, R. P., et al. (1977). The organization of histone genes in Drosophila melanogaster: Functional and evolutionary implications. Cold Spring Harbor Symposia on Quantitative Biology XLII: 1047-51

Lone, I. N., Shukla, M. S., Charles Richard, J. L., Peshev, Z. Y., Dimitrov, S. and Angelov, D. (2013). Binding of NF-κB to nucleosomes: effect of translational positioning, nucleosome remodeling and linker histone H1. PLoS Genet 9: e1003830. PubMed ID: 24086160

Lu, X., et al. (2009). Linker histone H1 is essential for Drosophila development, the establishment of pericentric heterochromatin, and a normal polytene chromosome structure. Genes Dev. 23(4): 452-65. PubMed Citation: 19196654

Lu, Z. H., et al. (1998). Histone H1 reduces the frequency of initiation in Xenopus egg extract by limiting the assembly of prereplication complexes on sperm chromatin. Mol. Biol. Cell 9(5): 1163-1176.

Maier, V. K., Chioda, M., Rhodes, D. and Becker, P. B. (2008). ACF catalyses chromatosome movements in chromatin fibres. EMBO J. 27(6): 817-26. PubMed citation: 17962805

Maresca, T. J., Freedman, B. S. and Heald, R. (2005). Histone H1 is essential for mitotic chromosome architecture and segregation in Xenopus laevis egg extracts. J. Cell Biol. 169: 859-869. PubMed citation: 15967810

Mishima, Y., Kaizu, H. and Kominami, R. (1997). Pairing of DNA fragments containing (GGA:TCC)n repeats and promotion by high mobility group protein 1 and histone H1. J. Biol. Chem. 272(42): 26578-26584.

Mizzen, C. A., et al. (1999). Identification and mutation of phosphorylation sites in a linker histone. Phosphorylation of macronuclear H1 is not essential for viability in tetrahymena. J. Biol. Chem. 274(21): 14533-6.

Murphy, T.J. and Blumenfeld, M. (1986). Nucleotide sequence of a Drosophila melanogaster H1 histone gene. Nucleic Acids Res. 14:5563

Nabirochkin, S., Ossokina, M. and Heidmann, T. (1998). A nuclear matrix/scaffold attachment region co-localizes with the gypsy retrotransposon insulator sequence. J. Biol. Chem. 273(4): 2473-2479. Medline reference:

Ner, S. S. and Travers, A. A. (1994). HMG-D, the Drosophila melanogaster homologue of HMG 1 protein, is associated with early embryonic chromatin in the absence of histone H1. EMBO J. 13: 1817-22

Ner, S. S., et al. (2001). HMG-D and Histone H1 interplay during chromatin assembly and early embryogenesis. J. Biol. Chem. 276: 37569-37576. 11473125

Nielsen, A. L., et al. (2001). Heterochromatin formation in mammalian cells: Interaction between histones and HP1 proteins. Molec. Cell 7: 729-739

Patterton, H. G., Landel, C. C., Landsman, D., Peterson, C. L. and Simpson, R. T. (1998). The biochemical and phenotypic characterization of Hho1p, the putative linker histone H1 of Saccharomyces cerevisiae. J. Biol. Chem. 273: 7268-7276.

Pham, A. D. and Sauer, F. (2000). Ubiquitin-activating/conjugating activity of TAFII250, a mediator of activation of gene expression in Drosophila. Science 289(5488): 2357-60. 11009423

Pruss, D., et al. (1996). An asymmetric model for the nucleosome: A binding site for linker histones inside the DNA gyres. Science 274: 614-617. PubMed ID: 8849453

Raghuram, N., Strickfaden, H., McDonald, D., Williams, K., Fang, H., Mizzen, C., Hayes, J. J., Th'ng, J. and Hendzel, M. J. (2013). Pin1 promotes histone H1 dephosphorylation and stabilizes its binding to chromatin. J Cell Biol 203: 57-71. PubMed ID: 24100296

Ramakrishnan, V., et al. (1993). Crystal structure of globular domain of histone H5 and its implications for nucleosome binding. Nature 362: 219-223

Sandaltzopoulos, R., Blank, T., and & Becker, P. B. (1994). Transcriptional repression by nucleosomes but not H1 in reconstituted preblastoderm Drosophila chromatin. EMBO J. 13: 373-9. PubMed Citation: 8313882

Seigneurin, D., et al. (1995). Developmentally regulated chromatin acetylation and histone H1(0) accumulation. Int. J. Dev. Biol. 39: 597-603. PubMed Citation: 8619958

Shen, X., et al. (1995). Linker histones are not essentioal and affect chromatin condensation in vivo. Cell 82: 47-56. PubMed Citation: 7606784

Shen, X. and Gorovsky, M. A. (1996). Linker Histone H1 regulates specific gene expression but not global transcription in vivo. Cell 86: 475-83. PubMed Citation: 8756729

Shevelyov, Y. Y., Lavrov, S. A., Mikhaylova, L. M., Nurminsky, I. D., Kulathinal, R. J., Egorova, K. S., Rozovsky, Y. M. and Nurminsky, D. I. (2009). The B-type lamin is required for somatic repression of testis-specific gene clusters. Proc Natl Acad Sci U S A 106: 3282-3287. PubMed ID: 19218438

Siriaco, G., Deuring, R., Chioda, M., Becker, P. B. and Tamkun, J. W. (2009). Drosophila ISWI regulates the association of histone H1 with interphase chromosomes in vivo. Genetics 182(3): 661-9. PubMed Citation: 19380479

Smith, L. C., et al. (1995). Assembly of somatic histone H1 onto chromatin during bovine early embryogenesis. J. Exp. Zool. 273: 317-326. PubMed Citation: 8530913

Smith, M. B. and Weiler, K. S. (2010). Drosophila D1 overexpression induces ectopic pairing of polytene chromosomes and is deleterious to development. Chromosoma 119: 287-309. PubMed ID: 20127347

Steinbach, O. C., Wolffe, A. P. and Rupp, R. A. (1997). Somatic linker histones cause loss of mesodermal competence in Xenopus. Nature 389: 395-399

Syed, S. H., Goutte-Gattat, D., Becker, N., Meyer, S., Shukla, M. S., Hayes, J. J., Everaers, R., Angelov, D., Bednar, J. and Dimitrov, S. (2010). Single-base resolution mapping of H1-nucleosome interactions and 3D organization of the nucleosome. Proc Natl Acad Sci U S A 107: 9620-9625. PubMed ID: 20457934

Thomas, J. O., Rees, C., and Finch, J. T. (1992). Cooperative binding of the globular domains of histones H1 and H5 to DNA. Nucleic Acids Res. 20: 187-194

Tulin, A. and Spradling, A. (2003). Chromatin loosening by poly(ADP)-ribose polymerase (PARP) at Drosophila puff loci. Science 299: 560-562. 12543974

Ushinsky, S. C., Bussey, H., Ahmed, A. A., Wang, Y., Friesen, J., Williams, B. A. and Storms, R. K. (1997). Histone H1 in Saccharomyces cerevisiae. Yeast 13: 151-161

Varga-Weisz, P. D., Blank, T. A. and Becker, P. B. (1995). Energy-dependent chromatin accessibility and nucleosome mobility in a cell-free system. EMBO J. 14: 2209-2216

Vermaak, D., et al. (1998). The globular domain of histone H1 is sufficient to direct specific gene repression in early Xenopus embryos. Curr. Biol. 8(9): 533-536

Vicent, G. P., et al. (2011). Four enzymes cooperate to displace histone H1 during the first minute of hormonal gene activation. Genes Dev. 25(8): 845-62. PubMed Citation: 21447625

Walker, J. and Bownes, M. (1998). The expression of histone genes during Drosophila melanogaster oogenesis. Dev. Genes Evol. 207(8): 535-41

Wiekowski, M., et al. (1997). Changes in histone synthesis and modification at the beginning of mouse development correlate with the establishment of chromatin mediated repression of transcription. J. Cell Sci. 110: 1147-1158. 9191039

Yaneva, J., et al. (1997). The major chromatin protein histone H1 binds preferentially to cis-platinum-damaged DNA. Proc. Natl. Acad. Sci. 94(25): 13448-13451. PubMed Citation: 9391045

Yang, J. G., Madrid, T. S., Sevastopoulos, E. and Narlikar, G. J. (2006). The chromatin-remodeling enzyme ACF is an ATP-dependent DNA length sensor that regulates nucleosome spacing. Nat. Struct. Mol. Biol. 13: 1078-1083. PubMed citation: 17099699

Yao, C., Ding, Y., Cai, W., Wang, C., Girton, J., Johansen, K. M. and Johansen, J. (2012). The chromodomain-containing NH(2)-terminus of Chromator interacts with histone H1 and is required for correct targeting to chromatin. Chromosoma 121: 209-220. PubMed ID: 22203189

Yarosh, W. and Spradling, A. C. (2014). Incomplete replication generates somatic DNA alterations within Drosophila polytene salivary gland cells. Genes Dev 28(16): 1840-1855. PubMed ID: 25128500

Zhang, J., Tan, D., DeRose, E. F., Perera, L., Dominski, Z., Marzluff, W. F., Tong, L. and Hall, T. M. (2014). Molecular mechanisms for the regulation of histone mRNA stem-loop-binding protein by phosphorylation. Proc Natl Acad Sci U S A 111(29): E2937-46. PubMed ID: 25002523

Zhao, K., et al. (1993). SAR-dependent mobilization of histone H1 by HMG-I/Y in vitro: HMG-I/Y is enriched in H1-depleted chromatin. EMBO J. 12: 3237-47. PubMed Citation: 8344261

Zhimulev, I. F., Belyaeva, E. S., Makunin, I. V., Pirrotta, V., Volkova, E. I., Alekseyenko, A. A., Andreyeva, E. N., Makarevich, G. F., Boldyreva, L. V., Nanayev, R. A. and Demakova, O. V. (2003). Influence of the SuUR gene on intercalary heterochromatin in Drosophila melanogaster polytene chromosomes. Chromosoma 111: 377-398. PubMed ID: 12644953

Zhou, B. R., Feng, H., Kato, H., Dai, L., Yang, Y., Zhou, Y. and Bai, Y. (2013). Structural insights into the histone H1-nucleosome complex. Proc Natl Acad Sci U S A 110: 19390-19395. PubMed ID: 24218562

Zhou, B. R., Feng, H., Ghirlando, R., Li, S., Schwieters, C. D. and Bai, Y. (2016). A small number of residues can determine if linker histones are bound on or off dyad in the chromatosome. J Mol Biol 428(20):3948-3959. PubMed ID: 27558112


Histone H1: Biological Overview | Evolutionary Homologs | Regulation | Developmental Biology

date revised: 1 September 2020

Home page: The Interactive Fly © 1997 Thomas B. Brody, Ph.D.

The Interactive Fly resides on the
Society for Developmental Biology's Web server.