scispace - formally typeset
Search or ask a question

Showing papers on "Histone H4 published in 2002"


Journal ArticleDOI
01 Nov 2002-Cell
TL;DR: It is found that only a small subset of lysines in histones H4 and H3 are acetylated in vivo by the GCN5 acetyltransferase during activation of the IFN-beta gene, which reveals the cascade of gene activation via a point-by-point interpretation of the histone code through the ordered recruitment of bromodomain-containing transcription complexes.

657 citations


Journal ArticleDOI
26 Sep 2002-Nature
TL;DR: The data show that histone tail acetylation is required directly for DNA repair and suggest that a related human HAT complex may function similarly.
Abstract: Although the acetylation of histones has a well-documented regulatory role in transcription, its role in other chromosomal functions remains largely unexplored. Here we show that distinct patterns of histone H4 acetylation are essential in two separate pathways of double-strand break repair. A budding yeast strain with mutations in wild-type H4 acetylation sites shows defects in nonhomologous end joining repair and in a newly described pathway of replication-coupled repair. Both pathways require the ESA1 histone acetyl transferase (HAT), which is responsible for acetylating all H4 tail lysines, including ectopic lysines that restore repair capacity to a mutant H4 tail. Arp4, a protein that binds histone H4 tails and is part of the Esa1-containing NuA4 HAT complex, is recruited specifically to DNA double-strand breaks that are generated in vivo. The purified Esa1-Arp4 HAT complex acetylates linear nucleosomal arrays with far greater efficiency than circular arrays in vitro, indicating that it preferentially acetylates nucleosomes near a break site. Together, our data show that histone tail acetylation is required directly for DNA repair and suggest that a related human HAT complex may function similarly.

581 citations



Journal ArticleDOI
TL;DR: There is a gradient of acetylation of histone H4 at lysine 16 (H4–Lys16) along a yeast chromosome; this gradient ranges from a hypoacetylated state in regions near the telomere to a hyperacetylation state in more distant regions.
Abstract: Genes located in chromosomal regions near telomeres are transcriptionally silent, whereas those located in regions away from telomeres are not. Here we show that there is a gradient of acetylation of histone H4 at lysine 16 (H4-Lys16) along a yeast chromosome; this gradient ranges from a hypoacetylated state in regions near the telomere to a hyperacetylated state in more distant regions. The hyperacetylation is regulated by Sas2p, a member of the MYST-type family of histone acetylases, whereas hypoacetylation is under the control of Sir2p, a histone deacetylase. Loss of hyperacetylation is accompanied by an increase in localization of the telomere protein Sir3p and the inactivation of gene expression in telomere-distal regions. Thus, the Sas2p and Sir2p function in concert to regulate transcription in yeast, by acetylating and deacetylating H4-Lys16 in a mechanism that may be common to all eukaryotes.

484 citations


Journal ArticleDOI
TL;DR: It is shown that the MYST-like acetyltransferase Sas2p is required for the acetylation of H4–Lys16 in euchromatin and disruption of Sir3p binding in a deacetylase-deficient Sir 2Δ strain can be suppressed by sas2Δ.
Abstract: The Sir3 protein helps form telomeric heterochromatin by interacting with hypoacetylated histone H4 lysine 16 (H4–Lys16). The molecular nature of the heterochromatin boundary is still unknown. Here we show that the MYST-like acetyltransferase Sas2p is required for the acetylation (Ac) of H4–Lys16 in euchromatin. In a sas2Δ strain or a phenocopy Lys16Arg mutant, Sir3p spreads from roughly 3 kb to roughly 15 kb, causing hypoacetylation and repression of adjacent chromatin. We also found that disruption of Sir3p binding in a deacetylase-deficient Sir 2Δ strain can be suppressed by sas2Δ. These data indicate that opposing effects of Sir2p and Sas2p on acetylation of H4–Lys16 maintain the boundary at telomeric heterochromatin.

454 citations


Journal ArticleDOI
01 Nov 2002-Genomics
TL;DR: The multigene family encoding the five classes of replication-dependent histones has been identified from the human and mouse genome sequence, and the complexity of the histone protein complement significantly greater than previously thought.

432 citations


Journal ArticleDOI
TL;DR: Although H4-K20 methylation does not correlate with gene activity, it appears to be regulated during the cell cycle and is demonstrated to have an essential role in Drosophila development.

317 citations


Journal ArticleDOI
TL;DR: It is shown that the Dot1 protein possesses intrinsic histone methyltransferase activity, and it is demonstrated that, like its mammalian homolog PRMT1, Rmt1 specifically dimethylates an arginine residue at position 3 of histone H4 N-terminal tail.

293 citations


Journal ArticleDOI
TL;DR: It is shown that the levels of multiply acetylated histone H3 and H4 in chromatin are decreased in vivo in yeast cells lacking ELP3, and that Elongator can acetylate both core histones and nucleosomal substrates.
Abstract: The elongating, hyperphosphorylated form of RNA polymerase II is associated with the Elongator complex, which has the histone acetyltransferase (HAT) Elp3 as a subunit. Here we show that, in contrast to the isolated Elp3 subunit, the activity of intact Elongator complex is directed specifically toward the amino-terminal tails of histone H3 and H4, and that Elongator can acetylate both core histones and nucleosomal substrates. The predominant acetylation sites are lysine-14 of histone H3 and lysine-8 of histone H4. The three smallest Elongator subunits—Elp4, Elp5, and Elp6—are required for HAT activity, and Elongator binds to both naked and nucleosomal DNA. By using chromatin immunoprecipitation, we show that the levels of multiply acetylated histone H3 and H4 in chromatin are decreased in vivo in yeast cells lacking ELP3.

279 citations


Journal ArticleDOI
TL;DR: Males can be rescued by roX cDNAs from autosomal transgenes, demonstrating the genetic separation of the chromatin entry and RNA‐encoding functions.
Abstract: The roX1 and roX2 genes of Drosophila produce male‐specific non‐coding RNAs that co‐localize with the Male‐Specific Lethal (MSL) protein complex. This complex mediates up‐regulation of the male X chromo some by increasing histone H4 acetylation, thus contributing to the equalization of X‐linked gene expression between the sexes. Both roX genes overlap two of ∼35 chromatin entry sites, DNA sequences proposed to act in cis to direct the MSL complex to the X chromosome. Although dosage compensation is essential in males, an intact roX1 gene is not required by either sex. We have generated flies lacking roX2 and find that this gene is also non‐essential. However, simultaneous removal of both roX RNAs causes a striking male‐specific reduction in viability accompanied by relocation of the MSL proteins and acetylated histone H4 from the X chromosome to autosomal sites and heterochromatin. Males can be rescued by roX cDNAs from autosomal transgenes, demonstrating the genetic separation of the chromatin entry and RNA‐encoding functions. Therefore, the roX1 and roX2 genes produce redundant, male‐specific lethal transcripts required for targeting the MSL complex.

251 citations


Journal ArticleDOI
TL;DR: H4 Lys 20 methylation by PR-Set7 during mitosis acts to antagonize H4 Lys 16 acetylation and to establish a mechanism by which this mark is epigenetically transmitted.
Abstract: We describe distinct patterns of histone methylation during human cell cycle progression. Histone H4 methyltransferase activity was found to be cell cycle-regulated, consistent with increased H4 Lys 20 methylation at mitosis. This increase closely followed the cell cycle-regulated expression of the H4 Lys 20 methyltransferase, PR-Set7. Localization of PR-Set7 to mitotic chromosomes and subsequent increase in H4 Lys 20 methylation were inversely correlated to transient H4 Lys 16 acetylation in early S-phase. These data suggest that H4 Lys 20 methylation by PR-Set7 during mitosis acts to antagonize H4 Lys 16 acetylation and to establish a mechanism by which this mark is epigenetically transmitted.

Journal ArticleDOI
TL;DR: It is found that acetylation of histone H4 in rat hippocampal CA3 neurons was reduced at the glutamate receptor 2 (GluR2; GRIA2) glutamate receptor promoter but increased at brain-derived neurotrophic factor promoter P2 as soon as 3 hr after induction of status epilepticus by pilocarpine, indicating that status epileptus rapidly activates different signal pathways to modulate histone acetylations in a promoter-specific manner.
Abstract: The mechanisms underlying seizure-induced changes in gene expression are unclear. Using a chromatin immunoprecipitation assay, we found that acetylation of histone H4 in rat hippocampal CA3 neurons was reduced at the glutamate receptor 2 (GluR2; GRIA2) glutamate receptor promoter but increased at brain-derived neurotrophic factor promoter P2 as soon as 3 hr after induction of status epilepticus by pilocarpine. This result indicates that status epilepticus rapidly activates different signal pathways to modulate histone acetylation in a promoter-specific manner. H4 deacetylation preceded seizure-induced GluR2 mRNA downregulation. The histone deacetylase inhibitor trichostatin A prevented and quickly reversed deacetylation of GluR2-associated histones. Trichostatin A also blunted seizure-induced downregulation of GluR2 mRNA in CA3. Thus, rapid gene-specific changes in histone acetylation patterns may be a key early step in the pathological processes triggered by status epilepticus.

Journal ArticleDOI
TL;DR: It is shown that blocking H4K16 acetylation suppresses the X chromosome defects resulting from loss of ISWI function in males and directly counteracts chromatin compaction mediated by the ISWI ATPase.
Abstract: Mutations in Drosophila ISWI, a member of the SWI2/SNF2 family of chromatin remodeling ATPases, alter the global architecture of the male X chromosome. The transcription of genes on this chromosome is increased 2-fold relative to females due to dosage compensation, a process involving the acetylation of histone H4 at lysine 16 (H4K16). Here we show that blocking H4K16 acetylation suppresses the X chromosome defects resulting from loss of ISWI function in males. In contrast, the forced acetylation of H4K16 in ISWI mutant females causes X chromosome defects indistinguishable from those seen in ISWI mutant males. Increased expression of MOF, the histone acetyltransferase that acetylates H4K16, strongly enhances phenotypes resulting from the partial loss of ISWI function. Peptide competition assays revealed that H4K16 acetylation reduces the ability of ISWI to interact productively with its substrate. These findings suggest that H4K16 acetylation directly counteracts chromatin compaction mediated by the ISWI ATPase.

Journal ArticleDOI
TL;DR: This work investigated the methylation pattern of histone H4 both in different mammalian organs of various ages and in cell lines by applying mass spectrometric analysis and a newly developed hydrophilic-interaction liquid chromatographic method enabling the simultaneous separation of methylated and acetylated forms, which obviates the need to work with radioactive materials.

Journal ArticleDOI
TL;DR: Ch Chromatin immunoprecipitations show that PRMT5 is associated specifically with the transcription start site region of the cyclin E1 promoter, providing the first direct experimental evidence that a type II arginine methylase is involved in the control of transcription and proliferation.
Abstract: We have identified previously a repressor element in the transcription start site region of the cyclin E1 promoter that periodically associates with an atypical, high molecular weight E2F complex, termed CERC. Purification of native CERC reveals the presence of the type II arginine methyltransferase PRMT5, which can mono- or symetrically dimethylate arginine residues in proteins. Chromatin immunoprecipitations (ChIPs) show that PRMT5 is associated specifically with the transcription start site region of the cyclin E1 promoter. ChIP analyses also show that this correlates with the presence on the same promoter region of arginine-methylated proteins including histone H4, an in vitro substrate of PRMT5. Consistent with its presence within the repressor complex, forced expression of PRMT5 negatively affects cyclin E1 promoter activity and cellular proliferation, effects that require its methyltransferase activity. These data provide the first direct experimental evidence that a type II arginine methylase is involved in the control of transcription and proliferation.

Journal ArticleDOI
TL;DR: By affecting SIR3-H4 binding, acetylation may regulate the formation of heterochromatin and help explain the hypoacetylated state of histone H4 in heterochromaatin of eukaryotes.

Journal ArticleDOI
TL;DR: It is shown that human CDY and mouse CDYL proteins exhibit histone acetyltransferase activity in vitro, with a strong preference for histone H4, which offers a plausible mechanism to account for spermatogenic failure in patients bearing deletions of the CDY genes.
Abstract: During spermiogenesis (the maturation of spermatids into spermatozoa) in many vertebrate species, protamines replace histones to become the primary DNA-packaging protein.It has long been thought that this process is facilitated by the hyperacetylation of histone H4. However, the responsible histone acetyltransferase enzymes are yet to be identified. CDY is a human Y-chromosomal gene family expressed exclusively in the testis and implicated in male infertility. Its mouse homolog Cdyl, which is autosomal, is expressed abundantly in the testis. Proteins encoded by CDY and its homologs bear the “chromodomain,” a motif implicated in chromatin binding. Here, we show that (i) human CDY and mouse CDYL proteins exhibit histone acetyltransferase activity in vitro, with a strong preference for histone H4; (ii) expression of human CDY and mouse Cdyl genes during spermatogenesis correlates with the occurrence of H4 hyperacetylation; and (iii) CDY and CDYL proteins are localized to the nuclei of maturing spermatids where H4 hyperacetylation takes place. Taken together, these data link human CDY and mouse CDYL to the histone-to-protamine transition in mammalian spermiogenesis. This link offers a plausible mechanism to account for spermatogenic failure in patients bearing deletions of the CDY genes.

Journal ArticleDOI
TL;DR: It is shown that there is a graded spectrum of histone H4 acetylation that is proportional to CGG repeat length and that correlates with responsiveness of the gene to DNA demethylation but not with chromatin condensation, and that DNA methylation is tightly coupled to the histone modifications associated with euchromatin.
Abstract: Fragile X syndrome is caused by an expansion of a polymorphic CGG triplet repeat that results in silencing of FMR1 expression. This expansion triggers methylation of FMR1's CpG island, hypoacetylation of associated histones, and chromatin condensation, all characteristics of a transcriptionally inactive gene. Here, we show that there is a graded spectrum of histone H4 acetylation that is proportional to CGG repeat length and that correlates with responsiveness of the gene to DNA demethylation but not with chromatin condensation. We also identify alterations in patient cells of two recently identified histone H3 modifications: methylation of histone H3 at lysine 4 and methylation of histone H3 at lysine 9, which are marks for euchromatin and heterochromatin, respectively. In fragile X cells, there is a decrease in methylation of histone H3 at lysine 4 with a large increase in methylation at lysine 9, a change that is consistent with the model of FMR1's switch from euchromatin to heterochromatin in the disease state. The high level of histone H3 methylation at lysine 9 may account for the failure of H3 to be acetylated after treatment of fragile X cells with inhibitors of histone deacetylases, a treatment that fully restores acetylation to histone H4. Using 5-aza-2′-deoxycytidine, we show that DNA methylation is tightly coupled to the histone modifications associated with euchromatin but not to the heterochromatic mark of methylation of histone H3 at lysine 9, consistent with recent findings that this histone modification may direct DNA methylation. Despite the drug-induced accumulation of mRNA in patient cells to 35% of the wild-type level, FMR1 protein remained undetectable. The identification of intermediates in the heterochromatinization of FMR1 has enabled us to begin to dissect the epigenetics of silencing of a disease-related gene in its natural chromosomal context.

Journal ArticleDOI
TL;DR: The acetylation isoforms of histone H4 from butyrate-treated HeLa cells were separated by C4 reverse-phase high pressure liquid chromatography and by polyacrylamide gel electrophoresis and revealed that lysine 20 is di-methylated in all modified isoforms, as well as the non-acetylated isoform of H4.

Journal ArticleDOI
TL;DR: It is shown that the N-terminal 'tail' of histone H4 is part of a more complex recognition epitope which is destroyed by grafting the H4 N- terminus onto other histones, and the ISWI ATPase is stimulated by isolated H4 tail peptides ISWI only in the presence of DNA.
Abstract: The ATPase ISWI is the catalytic core of several nucleosome remodeling complexes, which are able to alter histone–DNA interactions within nucleosomes such that the sliding of histone octamers on DNA is facilitated. Dynamic nucleosome repositioning may be involved in the assembly of chromatin with regularly spaced nucleosomes and accessible regulatory sequence elements. The mechanism that underlies nucleosome sliding is largely unresolved. We recently discovered that the N-terminal ‘tail’ of histone H4 is critical for nucleosome remodeling by ISWI. If deleted, nucleosomes are no longer recognized as substrates and do not stimulate the ATPase activity of ISWI. We show here that the H4 tail is part of a more complex recognition epitope which is destroyed by grafting the H4 N-terminus onto other histones. We mapped the H4 tail requirement to a hydrophilic patch consisting of the amino acids R17H18R19 localized at the base of the tail. These residues have been shown earlier to contact nucleosomal DNA, suggesting that ISWI recognizes an ‘epitope’ consisting of the DNA-bound H4 tail. Consistent with this hypothesis, the ISWI ATPase is stimulated by isolated H4 tail peptides ISWI only in the presence of DNA. Acetylation of the adjacent K12 and K16 residues impairs substrate recognition by ISWI.

Journal ArticleDOI
TL;DR: It is demonstrated that the decrease of histone acetylation in spermatids associated with impaired sperMatogenesis corresponds with the well known reduction of protamine expression in these cells and confirms the essential role of hist one hyperacetylation for correct histone‐to‐protamine exchange.
Abstract: Histone-to-protamine exchange in haploid spermatids is preceded by hyperacetylation of core histones resulting in decreased DNA-histone interaction. During normal spermatogenesis, immunohistochemistry with a polyclonal antihyperacetylated histone H4 antibody displayed a strong signal in nuclei of elongating spermatids and, in addition, spermatogonia. Quantitative analysis revealed 98.2 +/- 1.1% of immunopositive spermatids. The percentage of positive spermatids was significantly reduced in infertile men exhibiting at least qualitatively normal spermatogenesis (scores 10-8, 93.1 +/- 6.6%) and impaired spermatogenesis (scores 7-1, 74.9 +/- 23.4%). In seminiferous tubules showing spermatogenic arrest at the level of round spermatids, only 59.5 +/- 16.5% of spermatids were immunopositive for hyperacetylated histone H4. These data demonstrate that the decrease of histone acetylation in spermatids associated with impaired spermatogenesis corresponds with the well known reduction of protamine expression in these cells and confirms the essential role of histone hyperacetylation for correct histone-to-protamine exchange. In seminiferous tubules exhibiting round spermatid maturation arrest, there was an additional signal in nuclei of spermatocytes, suggesting that premature hyperacetylation of histone H4 may result in precocious histone-to-protamine exchange followed by infertility. This is in accordance with data from transgenic mice, where it has been demonstrated that premature expression of protamine-1 results in precocious chromatin condensation followed by sterility.

Journal ArticleDOI
TL;DR: HDRAC10 extends the HDAC superfamily and adds to a growing number of HDACs that have been found to have splice variants, suggesting that RNA processing may play a role in mediating the activity ofHDACs.

Journal ArticleDOI
TL;DR: The inviability of gcn5 yng2 double mutants suggests overlapping roles for H3 and H4 acetylation in DNA replication and repair, and implicate nucleosomal histone acetylations in maintaining genomic integrity during chromosomal replication.
Abstract: In eukaryotes, DNA is assembled into nucleosomes, the basic structural unit of chromatin. Factors that target nucleosomes to modulate chromatin structure and provide access to nucleosomal DNA are required to facilitate processes such as transcription, replication, and DNA repair. Covalent modification of core histone N-terminal tails by acetylation of conserved lysine residues is thought to relax chromatin structure and allow access of nonhistone factors to nucleosomal DNA (8, 43). Seminal studies by Smerdon and colleagues showed that histone acetylation might be upregulated to facilitate DNA damage repair in mammalian cells after UV irradiation (40, 41, 46). Recent biochemical results support this model. The mammalian TATA-binding protein-free Taf II complex is recruited along with nucleotide excision repair proteins to UV-damaged DNA (5). Importantly, via its Gcn5 histone acetyltransferase (HAT) subunit, the TATA-binding protein-free Taf II complex preferentially acetylates histone H3 in nucleosomes containing UV-damaged DNA. In turn, acetylation of histone H4 also appears to function in DNA damage repair, as expression of a dominant-negative form of the Tip60 H4 HAT in mammalian cells blocks repair of double strand breaks (23). Studies of histone N-terminal tail acetylation in the budding yeast Saccharomyces cerevisiae also support a role for histone acetylation in DNA repair and genomic integrity. Cells expressing H4 mutants lacking N-terminal acetylation sites activate DNA damage repair signaling and perform a G2/M delay even during unperturbed vegetative growth (33). In turn, conditional mutants in the essential yeast Tip60 homolog Esa1, the catalytic subunit of NuA4, lose nucleosomal histone H4 acetylation and accumulate in G2/M at the nonpermissive temperature. The terminal arrest is partly relieved by abrogating DNA damage checkpoint arrest (13). Analogously, mutants deficient in the Gcn5 or Sas3 histone H3 HAT display a G2/M delay which may also reflect DNA damage checkpoint activation (22, 51). The molecular pathway leading to DNA damage checkpoint activation in histone acetylation mutants remains poorly understood. Perhaps transcription of genes necessary for DNA metabolism is critically impaired, reflecting the known requirement for targeted histone acetylation in transcriptional activation (6, 28). Nonetheless, transcription defects are unlikely to be the only underlying mechanism. Studies of genome-wide expression in yeast histone H4 acetylation mutants revealed surprisingly few significant changes (10, 42). In addition to gene-specific regulation, Esa1 has been implicated in modification of histone H4 across large segments of chromosomes (50). One interpretation is that the DNA-damage-dependent cell cycle arrest with nonacetylatable H4 and esa1 mutants may arise from the effects of decreased genome-wide acetylation per se rather than decreased expression of specific transcripts. Like Esa1, virtually every other NuA4 subunit is essential for cell viability (2, 20). However, mutants lacking the NuA4 subunit Yng2 remain viable and yet are specifically compromised in global nucleosomal histone H4 acetylation (10, 30, 35). Mutants lacking Yng2 display a G2/M delay (10) and are sensitized to methylmethane sulfonate (35). Thus, we have used the yng2 mutant as a tool to study the role of NuA4 activity in DNA damage responses. As for esa1, the yng2 G2/M delay is DNA damage checkpoint dependent. Further, we demonstrate that Yng2 is required specifically to respond to DNA damage during S phase, suggesting a critical role for NuA4 in maintaining genomic integrity during DNA replication.

Journal ArticleDOI
TL;DR: The long-term evolution of histone H4 genes is examined to determine whether concerted evolution or purifying selection was the major factor for maintaining sequence homogeneity and suggest that the members of this gene family evolve according to the birth-and-death model of evolution under strong purify selection.
Abstract: Histones are small basic proteins encoded by a multigene family and are responsible for the nucleosomal organization of chromatin in eukaryotes. Because of the high degree of protein sequence conservation, it is generally believed that histone genes are subject to concerted evolution. However, purifying selection can also generate a high degree of sequence homogeneity. In this study, we examined the long-term evolution of histone H4 genes to determine whether concerted evolution or purifying selection was the major factor for maintaining sequence homogeneity. We analyzed the proportion (p(S)) of synonymous nucleotide differences between the H4 genes from 59 species of fungi, plants, animals, and protists and found that p(S) is generally very high and often close to the saturation level (p(S) ranging from 0.3 to 0.6) even though protein sequences are virtually identical for all H4 genes. A small proportion of genes showed a low level of p(S) values, but this appeared to be caused by recent gene duplication. Our findings suggest that the members of this gene family evolve according to the birth-and-death model of evolution under strong purifying selection. Using histone-like genes in archaebacteria as outgroups, we also showed that H1, H2A, H2B, H3, and H4 histone genes in eukaryotes form separate clusters and that these classes of genes diverged nearly at the same time, before the eukaryotic kingdoms diverged.

Journal ArticleDOI
TL;DR: The deregulated hair cycle and severely diminished fertility in Cutl1tm2Ejn/tm2 Ejn mice suggest that CDP/Cux is required for the developmental control of dermal and reproductive functions.
Abstract: Murine CDP/Cux, a homologue of the Drosophila Cut homeoprotein, modulates the promoter activity of cell cycle-related and cell-type-specific genes. CDP/Cux interacts with histone gene promoters as the DNA binding subunit of a large nuclear complex (HiNF-D). CDP/Cux is a ubiquitous protein containing four conserved DNA binding domains: three Cut repeats and a homeodomain. In this study, we analyzed genetically targeted mice (Cutl1(tm2Ejn), referred to as Delta C) that express a mutant CDP/Cux protein with a deletion of the C terminus, including the homeodomain. In comparison to the wild-type protein, indirect immunofluorescence showed that the mutant protein exhibited significantly reduced nuclear localization. Consistent with these data, DNA binding activity of HiNF-D was lost in nuclear extracts derived from mouse embryonic fibroblasts (MEFs) or adult tissues of homozygous mutant (Delta C(-/-)) mice, indicating the functional loss of CDP/Cux protein in the nucleus. No significant difference in growth characteristics or total histone H4 mRNA levels was observed between wild-type and Delta C(-/-) MEFs in culture. However, specific histone genes (H4.1 and H1) containing CDP/Cux binding sites have reduced expression levels in homozygous mutant MEFs. Stringent control of growth and differentiation appears to be compromised in vivo. Homozygous mutant mice have stunted growth (20 to 50% weight reduction), a high postnatal death rate of 60 to 70%, sparse abnormal coat hair, and severely reduced fertility. The deregulated hair cycle and severely diminished fertility in Cutl1(tm2Ejn/tm2Ejn) mice suggest that CDP/Cux is required for the developmental control of dermal and reproductive functions.

Journal ArticleDOI
TL;DR: A method for identifying protein acetylation at lysine residues by analysis of digested protein using HPLC/ESI-MS with a new modification-specific marker ion that was found to be more specific and approximately 9 times more sensitive than the immonium ion at m/z 143.1.
Abstract: Posttranslational acetylation of proteins regulates many diverse functions, including DNA recognition, protein-protein interaction, and protein stability. The identification of enzymes that regulate protein acetylation has revealed broader use of this modification than was previously suspected. In this study, we describe a method for identifying protein acetylation at lysine residues by analysis of digested protein using HPLC/ESI-MS with a new modification-specific marker ion. Collision-induced dissociation with capillary or nano-LC/ESI-TOF-MS was used to obtain a fragment ion useful as a marker for acetylated lysine. Although the acetylated lysine immonium ion at m/z 143.1 has been used as a marker ion for detecting acetylated lysine, it can be confused with internal fragment ion in some peptides, producing false positive results. We have found a novel marker ion at m/z 126.1, which is a further fragment ion induced by the loss of NH3 from the acetylated lysine immonium ions at m/z 143.1. This novel marker ion was found to be more specific and approximately 9 times more sensitive than the immonium ion at m/z 143.1. In addition, no interfering ions for acetylated peptides were found in the extracted ion chromatogram at m/z 126.1. The utility of this method was demonstrated with acetylated cytochrome c as a model compound. After the modification was probed by the new marker ion, the acetylated lysine site was determined by the CID-MS spectrum. This method was applied to identify histone H4 acetylation in HeLa cells treated with trichostatin A. Three protein bands separated by acid-urea-Triton gel electrophoresis were confirmed as tetra, tri, and diacetylated histone H4 at lysines 5, 8, 12, and 16. This method may be useful for assaying for lysine acetylation, which is an important regulatory process for a range of biological functions.

Journal ArticleDOI
TL;DR: A genetic screen to isolate mutant alleles of the histones H3 and H4 genes that would “lock” telomeric marker genes into a silenced state discovered that each of these mutants had a dramatic reduction in the level of acetylation at lysine 12 within the histone H4 tail, which is proposed to serve as a “memory mark” for propagating the expression state of a telomersic gene.
Abstract: Telomeric position effect in Saccharomyces cerevisiae is a chromatin-mediated phenomenon in which telomere proximal genes are repressed (silenced) in a heritable, but reversible, fashion. Once a transcriptional state (active or silenced) is established, however, there is a strong tendency for that state to be propagated. Twenty-five years ago, H. Weintraub and colleagues suggested that such heritability could be mediated by posttranslational modification of chromatin [Weintraub, H., Flint, S. J., Leffak, I. M., Groudine, M. & Grainger, R. M. (1977) Cold Spring Harbor Symp. Quant. Biol. 42, 401–407]. To identify potential sites within the chromatin that might act as sources of “memory” for the heritable transmission, we performed a genetic screen to isolate mutant alleles of the histones H3 and H4 genes that would “lock” telomeric marker genes into a silenced state. We identified mutations in the NH2-terminal tail and core of both histones; most of the amino acid changes mapped adjacent to lysines that are known sites of acetylation or methylation. We developed a method using MS to quantify the level of acetylation at each lysine within the histone H4 NH2-terminal tail in these mutants. We discovered that each of these mutants had a dramatic reduction in the level of acetylation at lysine 12 within the histone H4 tail. We propose that this lysine serves as a “memory mark” for propagating the expression state of a telomeric gene: when it is unacetylated, silent chromatin will be inherited; when it is acetylated an active state will be inherited.

Journal ArticleDOI
TL;DR: It is demonstrated that chromatin components associated with inactive CDH1 chromatin is heterogeneously modified and suggests the presence of multiple pathways for the formation of inactive chromatin.
Abstract: The structural alteration of chromatin has a key role in regulating gene expression. The alteration of chromatin is mediated by modification of its components. Detailed understanding of the relationship between these modifications, notably, methylation of the full-length CpG island, the association of methyl-CpG binding proteins (MBPs), and the acetylation and methylation of histones in gene silencing is vitally important. Currently, however, the manner in which chromatin components, associated with a specific gene, are modified is poorly understood. Here we provide in vivo evidence in cancer cells of the differential association between CpG methylation, MBPs, and histone modification in the entire CpG island of the human E-cadherin (CDH1) gene. Of the cell lines with CDH1 transcriptional repression, the distribution of methyl-CpGs in the CpG island differed markedly. In a cell line with gene silencing, the promoter region was almost methylation-free. Chromatin immunoprecipitation analysis revealed that the acetylation status of histone H4 differed between cell lines. However, deacetylated histone H3 was associated with the CpG island in all silenced cell lines. Binding of MeCP2 was also detected in all silenced cell lines. Additional binding of MBD1 protein was detected in a cell line in which the promoter region was poorly methylated and only histone H3 was deacetylated. Binding of MBD2 protein was detected in all other silenced cell lines. Histone H3 lysine 9 was methylated in all silenced cells, while histone H3 lysine 4 was methylated in some silenced cell lines. These results demonstrate that chromatin components associated with inactive CDH1 chromatin is heterogeneously modified and suggests the presence of multiple pathways for the formation of inactive chromatin.

Journal ArticleDOI
TL;DR: Results suggest that a variety of transcriptional activators, coactivators, and histone deacetylases participate in the regulation of HTLV-I transcription in infected T-cells.

Journal ArticleDOI
TL;DR: In this article, full-length masses of histones were analyzed by mass spectrometry to characterize post-translational modifications of bulk histones and their changes induced by cell stimulation.