BI83CH20-Almouzni

ARI

ANNUAL REVIEWS

30 April 2014

11:12

Further

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Click here for quick links to Annual Reviews content online, including: • Other articles in this volume • Top cited articles • Top downloaded articles • Our comprehensive search

Histone Chaperones: Assisting Histone Traffic and Nucleosome Dynamics Zachary A. Gurard-Levin, Jean-Pierre Quivy, and Genevi`eve Almouzni Institut Curie, Centre de Recherche; CNRS UMR 3664; Equipe Labellis´ee, Ligue contre le Cancer; and Universit´e Pierre et Marie Curie, Paris F-75248, France; email: [email protected]

Annu. Rev. Biochem. 2014. 83:487–517

Keywords

The Annual Review of Biochemistry is online at biochem.annualreviews.org

chromatin, epigenetics, cancer, genome function and instability

This article’s doi: 10.1146/annurev-biochem-060713-035536

Abstract

c 2014 by Annual Reviews. Copyright  All rights reserved

The functional organization of eukaryotic DNA into chromatin uses histones as components of its building block, the nucleosome. Histone chaperones, which are proteins that escort histones throughout their cellular life, are key actors in all facets of histone metabolism; they regulate the supply and dynamics of histones at chromatin for its assembly and disassembly. Histone chaperones can also participate in the distribution of histone variants, thereby defining distinct chromatin landscapes of importance for genome function, stability, and cell identity. Here, we discuss our current knowledge of the known histone chaperones and their histone partners, focusing on histone H3 and its variants. We then place them into an escort network that distributes these histones in various deposition pathways. Through their distinct interfaces, we show how they affect dynamics during DNA replication, DNA damage, and transcription, and how they maintain genome integrity. Finally, we discuss the importance of histone chaperones during development and describe how misregulation of the histone flow can link to disease.

487

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Contents

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

INTRODUCTION . . . . . . . . . . . . . . . . . Discovery and Definition of Histone Chaperones . . . . . . . . . . . Functional Classification of Histone Chaperones . . . . . . . . . . . Histone Chaperones and Their Histone Cargo . . . . . . . . . . . . . . . . Structural Insight into Histone Binding and Variant Selectivity . . . . . . . . . . . . . . . . . . . . . HISTONE CHAPERONES SHUTTLING HISTONES IN VARIOUS PATHWAYS . . . . . . . . . Histone Chaperones Acting on Histones in Transit . . . . . . . . . . . . Protecting H3–H4 from Degradation: NASP . . . . . . . . . . . Directing Histone Traffic: ASF1 . . HISTONE CHAPERONES INVOLVED IN DISTINCT HISTONE DEPOSITION PATHWAYS . . . . . . . . . . . . . . . . . . . . CAF-1 De Novo Deposition of Replicative H3.1–H4 in a DNA Synthesis–Coupled Pathway . . . Histone Chaperone Deposition of Replacement H3.3–H4 in a DNA Synthesis–Independent Pathway . . . . . . . . . . . . . . . . . . . . . .

488 489 490 490

494

496 496 496 498

498

498

499

INTRODUCTION The nucleosome core is the universal repeat unit of chromatin; it contains 147 bp of DNA wrapped around an octamer comprising two copies of the four core histones H3, H4, H2A, and H2B (1, 2). The addition of linker DNA, along with linker histone, completes the nucleosome (3) and connects adjacent nucleosomes. The linker DNA varies in length depending on the cell type and species. As shown by detailed contacts at 2.8-A˚resolution (4), the basic charge of the histones in the core particle is neutralized by the phosphate backbone of 488

Gurard-Levin

·

Quivy

·

Almouzni

HJURP Deposits CENP-A in a Cell Cycle–Dependent Manner . . . . . . . . . . . . . . . . . . . . . . . Histone Chaperones Participate in Cross Talk . . . . . . . . . . . . . . . . . . . . HISTONE CHAPERONES OPERATE AS AN ESCORT NETWORK . . . . . . . . . . . . . . . . . . . . . Histone Chaperones’ Interface with Various DNA Processes at the Cellular Level: Adaptor Domains . . . . . . . . . . . . . . . . . . . . . . Histone Chaperones’ Role During Replication: Handling Histones at the Fork . . . . . . . . . . . . . . . . . . . . Histone Chaperones’ Place During DNA Damage Response . . . . . . . Histone Chaperones’ Participation During Transcription. . . . . . . . . . DISCUSSION AND PERSPECTIVES . . . . . . . . . . . . . . . . Histone Chaperone Integration at the Organism Level During Development and Reprogramming . . . . . . . . . . . . . . . Histone Chaperone Misfunction and Disease . . . . . . . . . . . . . . . . . . . Questions About Histone Chaperones for Future Perspectives . . . . . . . . . . . . . . . . . . .

500 501

501

501

503 504 506 506

507 507

508

the DNA. However, before their incorporation into chromatin and after eviction, when histones are soluble, the high basicity may engage promiscuous interactions with any acidic partner in the cell or may form aggregates leading to defects such as those observed when there is an improper histone supply (5–8). Therefore, under most circumstances, dedicated proteins associate with nonnucleosomal histones, some of which directly buffer the positive charge. These factors are known as histone chaperones, a termed coined for nucleoplasmin, the first identified histone chaperone involved

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

in the prevention of aberrant histone–DNA aggregation during nucleosome assembly (9). Notably, the use of chaperone was later adopted to describe a class of proteins that help fold others, such as the heat-shock proteins (10). In this review, we examine the functional network of the histone chaperones at the cellular level (Figure 1). Specifically, we discuss (a) the identification and characterization of the known histone chaperones, (b) the role of histone chaperones acting on histones in transit, (c) histone chaperones in the best-characterized nucleosome assembly pathways, (d ) how the histone chaperones operate as an escort network in various DNA metabolic processes, and finally (e) the link between histone chaperones and development and disease.

Histone traffic Histone synthesis Degradation

Nuclear import

Histones in transit

Storage, buffering

Nucleosome assembly

Eviction, nucleosome dynamics

Exchange, recycling

Nucleosomal histones Specific dynamics

Discovery and Definition of Histone Chaperones In vitro the mixing of purified histones and DNA at physiological ionic strength causes aggregation and precipitation. To overcome this problem, reconstitution experiments either exploited slow dialysis from high (2 M NaCl) to physiological salt concentrations (2, 11) or used anionic polymers such as RNA (12) or polyglutamic acid (13, 14). However, these reconstitution experiments, unless positioning sequences are employed, do not generate regular arrays of nucleosomes. In contrast, assembly in crude extracts provided additional components, including chaperones and remodelers. The large maternal store of histones in Xenopus laevis eggs (15) that enable multiple rounds of cell division prior to the onset of transcription provided a major source of chaperone proteins. Indeed, cell-free extracts generated initially from unfertilized X. laevis eggs (16) or Drosophila embryos (17) efficiently formed nucleosomes by using the histones present in these extracts (18) on exogenously added DNA without noticeable histone–DNA aggregation. The first histone chaperone was thereby purified from X. laevis oocytes in 1978 (9) and named nucleoplasmin (19). In X. laevis eggs, nucleoplasmin has a major storage function for the ma-

Genomic regions Centromere

DNA processes Replication

Telomere

Repair

Genes ...

Transcription

Cellular level

Tissue

Organism and development

Normal Pathological

Figure 1 Histones are escorted by histone chaperones throughout their cellular life and are shuttled into various pathways for their nuclear import, storage, degradation, assembly, and dynamics at chromatin. Together, these processes participate in several DNA metabolic processes that regulate cellular function, which ultimately affects the entire organism.

ternal pool of H2A–H2B dimers, and similarly, the N1 and N2 proteins are in charge of H3–H4 dimers (20, 21). This histone selectivity is consistent with the proposed two-step nucleosome formation in vitro, wherein one chaperone first promotes the deposition of an H3–H4 tetramer [herein noted (H3–H4)2 ] and a different chaperone then deposits H2A–H2B dimers [for a review, see Kaufman & Almouzni (22)]. In this review, we focus on histone chaperones for histone H3 and its variants. For more information about H2A–H2B, please see References 23–26. In somatic cells, which are largely studied in culture, the soluble pool of histones is limited, and other chaperones were identified. The www.annualreviews.org • Histone Chaperones

489

ARI

30 April 2014

11:12

use of an in vitro SV40 DNA replication assay with a cytosolic extract (27) complemented with a salt-extracted nuclear fraction from the same cells (28) revealed an activity that promotes nucleosome formation on newly replicated DNA. Furthermore, fractionation of the nuclear extract demonstrated that a three-subunit protein complex termed chromatin assembly factor 1 (CAF-1) is responsible for this activity (29). Also identified in Xenopus (30), this key H3–H4 histone chaperone factor couples DNA replication to chromatin assembly and is essential for early development in Xenopus. We discuss its function in more detail below in the section titled CAF-1 De Novo Deposition of Replicative H3.1–H4 in a DNA Synthesis–Coupled Pathway. These two examples (nucleoplasmin and CAF-1) illustrate two important concepts for histone chaperones: (a) with nucleoplasmin, a storage function and some degree of histone specificity (H2A–H2B), and (b) with CAF-1, a deposition factor and the concept of coupling with one particular aspect of DNA metabolism, such as replication. Thus, in addition to the interaction with histones, one must also consider the interface aspect. We discuss these factors in conjunction with other chaperones throughout this review.

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

Functional Classification of Histone Chaperones The current definition of a histone chaperone is a protein that associates with histones and is involved in their transfer but is not necessarily part of the final product (31). In vitro, all histone chaperones share the fundamental ability to promote a progressive transfer of purified histones onto naked DNA at physiological ionic strength to reconstitute nucleosomes from purified components (32). However, not all histone chaperones promote nucleosome assembly in vivo. Investigators developed an assay that used X. laevis egg extracts that enabled them to follow chromatin assembly either coupled or uncoupled from DNA synthesis (33–35) to further classify histone chaperones. 490

Gurard-Levin

·

Quivy

·

Almouzni

By depleting chaperones from these extracts, in combination with add-back experiments using wild-type or mutant forms, the experiments helped define the histone chaperones’ role in particular pathways (36). This strategy revealed that CAF-1 and the histone regulator A (HIRA) chaperone are bona fide histone deposition factors that promote assembly that is dependent on and independent of DNA synthesis, respectively (30, 37). The assay also demonstrated that the histone chaperone antisilencing function 1 (ASF1), which directly interacts with histones H3–H4 and can promote nucleosome reconstitution in vitro (38, 39), does not necessarily have a direct role in nucleosome assembly in vivo (39). Rather, current data point to a model in which ASF1 can relay histones to either CAF-1 or HIRA for subsequent deposition, processes that we discuss below in the section titled Histone Chaperones Involved in Distinct Histone Deposition Pathways (reviewed in References 31 and 40). These observations underscore the importance of choosing assay conditions—specifically, nucleosome reconstitution from purified components and assembly using extracts—in a complementary fashion so as to formulate a hypothesis concerning their histone chaperone functions. On this basis, in vivo experiments can then be designed to test and strengthen these hypotheses.

Histone Chaperones and Their Histone Cargo Histone chaperones can be classified according to their selectivity for cognate histones. First, we consider H3–H4 or H2A–H2B chaperones (Table 1). However, because H3, H2A, and H2B can exist as several variants (41–44), their specificity can be further refined. The use of stable cell lines expressing epitope-tagged histone variants, combined with biochemical isolation from soluble extracts of the corresponding histone complexes, proved to be critical to identify associated histone chaperones (45, 46). This approach identified CAF-1 with the replicative H3.1 variant (45); HIRA (45) and death domain–associated protein (DAXX) (47)

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Table 1 Histone chaperones in multiple organisms Classification of histone chaperones

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

H3–H4

Histone chaperonea

Variant selectivity

Conservation

Main function(s)

Reference(s)

ASF1 (Drosophila melanogaster)

H3.1–H4, H3.3–H4

Asf1 (Saccharomyces cerevisiae, Xenopus laevis); Cia1 (Saccharomyces pombe); Sga1, Sga2 (Arabidopsis thaliana); ASF1a, ASF1b (Mus musculus, Homo sapiens)

Histone donor for CAF-1 and HIRA

97–99

CAF-1 complex (H. sapiens)

H3.1–H4



Deposition factor coupled to DNA synthesis: replication, DNA repair

29

p150

H3.1–H4

Rif2/Cac1 (S. cerevisiae), SPBC29A10.03C (S. pombe), Fas1 (A. thaliana), p180 (D. melanogaster), p150 (X. laevis, M. musculus)

p60

H3.1–H4

Cac2 (S. cerevisiae), SPAC26H5.03 (S. pombe), Fas2 (A. thaliana), p150 (D. melanogaster), p60 (X. laevis, M. musculus)

RbAp48b

H3–H4

Msi1/Cac3 (S. cerevisiae), Msi16 (S. pombe), Msi1 (A. thaliana), p55 (D. melanogaster), p48 (X. laevis), RbAp48 (M. musculus)

DAXX (with ATRX)

H3.3–H4



Deposition factor independent of DNA synthesis: telomere maintenance, ribosomal DNA, pericentric heterochromatin

DEK (D. melanogaster)

H3.3–H4

DEK (H. sapiens)

Transcriptional coactivator

Hif1 (S. cerevisiae)

H3–H4



Assists HAT

93

HIRA complex

H3.3–H4



Deposition factor independent of DNA synthesis

37

HIRA

H3.3–H4

HIR1/HIR2 (S. cerevisiae); Hip1, Sim2 (S. pombe), HIRA (A. thaliana, D. melanogaster, H. sapiens, M. musculus, X. laevis

Cabin1

H3.3–H4

Hir3 (S. cerevisiae), Hip3 (S. pombe), Cabin1 (M. musculus), AT4G32820 (A. thaliana)

129, 131

217

(Continued ) www.annualreviews.org • Histone Chaperones

491

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Table 1 (Continued) Classification of histone chaperones

Variant selectivity

Histone chaperonea

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

UBN1

H2A–H2B

Conservation

Main function(s)

Reference(s)

H3.3–H4

Hpc4 (S. cerevisiae), Hip4 (S. pombe), yemanuclein-A (D. melanogaster), T14N5.16 (A. thaliana), mCG.1031012 (M. musculus)

HJURP (H. sapiens)

CENP-A–H4

Scm3 (S. cerevisiae), HJURP (M. musculus, X. laevis), CAL1 (D. melanogaster)f

Deposition factor, centromere maintenance

N1/N2 (X. laevis)

H3–H4

tNASP, sNASP (M. musculus, H. sapiens)

H3–H4 storage in X. laevis oocytes; NASP protects H3–H4 from degradation in human cells

Rsf-1 (H. sapiens)

H3–H4

Rsf-1 (M. musculus)

Assists RC

214

Rtt106 (S. cerevisiae)

H3–H4

SPAC6G9.03c (S. pombe)

Heterochromatic silencing

204

Spt6 (S. cerevisiae)

H3–H4

Spt6 (S. pombe, D. melanogaster, X. laevis, M. musculus, H. sapiens)

Transcription initiation and elongation

205

SSRP1c (FACT complex)

H3–H4

Pob3 (S. cerevisiae), Pob3 (S. pombe), SSRP (A. thaliana), SSRP1 (D. melanogaster, X. laevis, M. musculus)

Transcription elongation, assistance in chromatin remodeling

58

Chz1 (S. cerevisiae)

H2A.Z–H2B

HIRIP3? (H. sapiens)

H2AZ incorporation by SWR1

206

Nap1d (X. laevis); Nap1-related proteins: Nap1L2 (M. musculus), SET/TAF1b (H. sapiens), CINAP (H. sapiens, M. musculus), Vps75 (S. cerevisiae)

H2A–H2B

Nap1 (S. cerevisiae, D. melanogaster, M. musculus, H. sapiens); NRP1, NRP2 (A. thaliana); Nap1, Nap1.2 (S. pombe)

Cytosolic-nuclear transport, replication, transcription

9, 210–213

Nucleoline (H. sapiens)

macroH2A– H2B

NSR1 (S. cerevisiae), nucleolin (A. thaliana, X. laevis, M. musculus)

Transcription elongation, assistance in chromatin remodeling

Nucleoplasmin (X. laevis), nucleophosmin 1 (NPM1) (H. sapiens)

H2A–H2B

Nip (D. melanogaster); NPM1, NPM2, NPM3 (M. musculus, H. sapiens)

Storage in X. laevis oocytes, cytosolicnuclear transport, replication, transcription

492

Gurard-Levin

·

Quivy

·

48, 49

21, 51, 52

26

9, 209

(Continued ) Almouzni

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Table 1 (Continued) Classification of histone chaperones

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Not determined or multiple

Histone chaperonea

Variant selectivity

Conservation

Main function(s)

Reference(s)

Spt16 (FACT complex)

H2A–H2B

Spt16 (S. cerevisiae, S. pombe, A. thaliana, D. melanogaster, X. laevis, M. musculus)

Transcription elongation, assistance in chromatin remodeling

58

Arp4 (S. cerevisiae)

Not determined

Alp5 (S. pombe), Arp4 (A. thaliana), BAP55 (D. melanogaster), BAF53 (M. musculus, H. sapiens)

Assists RC

207

Arp7, Arp9 (S. cerevisiae)

Not determined

Arp7, Arp9 (A. thaliana); BAP55 (D. melanogaster); BAF53 (M. musculus, H. sapiens)

Assists RC

215

Arp8 (S. cerevisiae)

H3–H4

BAP55 (D. melanogaster), BAF53 (M. musculus, H. sapiens)

Assists RC

208

Acf1 (D. melanogaster)

H2A–H2B, H3–H4

Itc1 (S. cerevisiae), Acf1 (M. musculus, H. sapiens)

Assists RC

216

a

Histone chaperones are classified as H3–H4, H2A–H2B, or not determined/multiple histone chaperones, and then listed alphabetically. Specifically preferred histone variants, when known, are in bold. b RbAp48 (also known as Rbbp4) and its homologs can act as single chaperones, as part of the multichaperone complex CAF-1, or within other complexes (RC, HAT, HDAC, or PC). c SSRP1, through its interaction with H3–H4, may assist Spt16 in nucleosome binding. d Both Nap1 and sNASP also interact with linker histones. e Nucleolin facilitates remodeling of nucleosomes containing macroH2A, suggesting specificity for this variant. f Proposed to be a functional homolog. Abbreviations: ATRX, α-thalassemia/mental retardation X-linked; CENP-A, centromere protein A; DAXX, death domain–associated protein; FACT, facilitates chromatin transcription; HAT, histone acetyltransferase; HDAC, histone deacetylase complex; HIRA, histone regulator A; HJURP, Holliday junction recognition protein; PC, Polycomb complex; PHC, pericentric heterochromatin; RC, remodeling complex; sNASP, somatic nuclear autoantigenic sperm protein (NASP); SSRP1, structure-specific recognition protein 1; tNASP, testicular NASP; UBN1, ubinuclein 1.

with the replacement variant H3.3; and Holliday junction recognition protein (HJURP) with the human centromeric H3 variant centromere protein A (CENP-A) (48, 49). In contrast to this selective behavior, the histone chaperones ASF1 and nuclear autoantigenic sperm protein (NASP) are present in both H3.1 and H3.3 soluble complexes (45). This finding shows that histone chaperone–histone interactions are not necessarily strict. That the histone chaperone NASP associates with H1 in mammals (50) and is also an H3–H4 chaperone also illustrates this point (45, 51, 52). Furthermore, immunoprecipitation experiments showed that Nap1, an H2A–H2B chaperone, binds the B4 linker his-

tone in X. laevis eggs (53). Thus, the selectivity of histone chaperone–histone interactions should be considered with caution and in a context-dependent manner, given that changes (including alterations of histone provision or histone chaperone expression) can affect these interactions. As stated above, beyond in vitro assays the ability to test chaperone activity in vivo is critical. In cell culture, the use of a system that allows expression of histone variants fused with a DNA alkyltransferase hAGT mutant is a powerful in vivo tool for histone deposition. In this assay, referred to as SNAP-tag technology, the DNA alkyltransferase protein reacts rapidly www.annualreviews.org • Histone Chaperones

493

ARI

30 April 2014

11:12

and specifically in cell culture with exogenously added benzylguanine derivatives. This process enables covalent labeling of the histone variant fusion molecule with a synthetic probe (54) that may or may not be fluorescent, thus enabling pulse-chase experiments. The first application of quench-chase-pulse protocols allowed investigators to distinguish newly synthesized CENP-A from parental CENP-A and to determine the timing of new CENP-A deposition during the cell cycle (55); this approach later enabled the discovery that new CENP-A deposition is HJURP dependent (48, 49). Subsequent adaptations of this assay with other histone variants are described below.

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

Structural Insight into Histone Binding and Variant Selectivity The identification of acidic patches on several histone chaperones accords with the idea that histone chaperones play a general role in neutralizing the basicity of histones (56–58). Posttranslational modifications (PTMs) can also alter the electrostatic potential of the chaperone and affect histone binding, as exemplified by polyglutamylation of Nap1 in Drosophila melanogaster (59). However, in some cases charge neutralization alone is dispensable for some histone chaperone functions (60–62). Beyond these general charge properties, a characteristic histone-binding motif can be found as a signature on several H3–H4 and H2A–H2B chaperones (56, 60, 62–67). These shared features may relate to common proper-

ties, yet they do not necessarily imply that all histone chaperones have the same partners, so a refined analysis is required. High-resolution structures aid in identifying key domains and determining how histones may interact with their cognate partners. For example, the structure of ASF1 with a dimer of histones H3–H4 shows that it occludes the tetramerization of H3–H4 by binding critical residues at the H3–H3 interface (Figure 2a). This finding accords with results from in vitro experiments, in which ASF1 acts to promote the disruption of (H3–H4)2 tetramers (68). A current model for ASF1 function at the replication fork proposes that (a) ahead of the fork, ASF1 handles histones from disrupted nucleosomes and (b) behind the fork it delivers dimers to other deposition factors involved in assembly for both recycling and de novo deposition (69). Furthermore, the structure shows that the divergent residues between histones H3.1 and H3.3 (41) remain exposed to the solvent (68, 70, 71), enabling other chaperones to differentiate the variants bound to ASF1 and target them to particular deposition pathways, such as HIRA and CAF-1. The solved structure of ASF1 bound to a small peptide corresponding to the HIRA B domain shows that a region within the N-terminal 156 residues of ASF1 mediates the interaction (72). CAF-1 p60 also contains B-domain motifs that recognize the same ASF1-binding site, a finding that explains why ASF1 binds these proteins in a mutually exclusive manner (72). These structural observations help illustrate a scheme in which ASF1

−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−→ Figure 2 Structural insights into histone chaperone–histone binding, variant selectivity, and in vivo function. (a) (Left) A soluble H3–H4 dimer bound to ASF1 [Protein Data Bank (PDB) identifier 2I05] (68). (Right) A nucleosomal (H3–H4)2 tetramer (PDB 1AOI) (4). Common H3 residues that bind antisilencing function 1 (ASF1) and mediate the (H3–H4)2 tetramer are colored red. The H4 C terminus (brown) undergoes a conformational change following binding to ASF1. Divergent residues between H3.1 and H3.3 ( purple) remain exposed to the solvent in the ASF1 complex. (b) (Left) Death domain–associated protein (DAXX) in complex with H3.3–H4. (Right) Holliday junction recognition protein (HJURP) in complex with centromere protein A (CENP-A)–H4. Red boxes indicate critical residues for variant selectivity. Similar binding motifs are depicted (bottom), and conserved residues are indicated with side chains and labels. DAXX, PDB 4H9N (74); HJURP, PDB 3R45 (75). Abbreviations: aa, amino acids; CAF-1, chromatin assembly factor 1; CBD, CENP-A-binding domain; HIRA, histone regulator A; HBD, histone-binding domain. 494

Gurard-Levin

·

Quivy

·

Almouzni

BI83CH20-Almouzni

ARI

30 April 2014

11:12

can act as a histone donor that functions with CAF-1 and HIRA (22). Interestingly, this binding site for the B domain is conserved between human ASF1a and ASF1b, suggesting possible redundancy (73). However, as discussed below, this is not necessarily the case.

a

The solved structures of DAXX and HJURP (Scm3 in yeast) with H3.3–H4 and CENP-A–H4 (Cse4 in yeast), respectively, revealed additional histone chaperones that bind dimers (74–76), although not all chaperones share this property (60, 77). The DAXX

ASF1 binds a dimer of H3–H4 Nucleosomal (H3–H4)2

Soluble H3–H4 dimer

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

ASF1 (aa1–156)

H3–H3’ interface

H4 HIRA/CAF-1binding site H3 H4 C terminus

H3–H3’ interface

b

H4 C terminus

H3 aa 87, 89–90

H3 aa 87, 89–90

Similar binding motifs: DAXX with H3.3–H4 and HJURP with CENP-A–H4 DAXX (HBD)

HJURP (CBD)

H3.3

CENP-A Ser68 H4 H3.3 Gly90 CENP-A

H4

CENP-A

H4

Y

DAXX

K

L

L

L

Y

K

L

L

L

HJURP

www.annualreviews.org • Histone Chaperones

495

ARI

30 April 2014

11:12

structure demonstrates an interaction with a region of H3.3 that is also recognized by ASF1 (Figure 2b). This finding suggests that the formation of DAXX and ASF1 complexes with H3.3 represent concurrent events in vivo, a hypothesis supported by pull-down experiments in vitro and in vivo (74). The HJURP structure underscores the importance of the CENP-A targeting domain and CENP-A Ser68 to distinguish this variant from canonical H3 (Figure 2b, right) (75). Interestingly, the structures of DAXX with H3.3– H4 and HJURP with CENP-A–H4 reveal a striking similarity in terms of how they associate with their respective histone partners (Figure 2b). Therefore, cross talk may play a role in the ability of these chaperones to bind different variants, particularly in the case of an imbalance in histone provision (78). Therefore, how cross talk could lead to errors in the timing and/or placement of these variants throughout the genome that affect cell function is important to consider. The association of the FACT (facilitates chromatin transcription) subunit Spt16 with an H2A–H2B dimer provides evidence for the first chaperone that interacts through H2B (79), which may explain its broad association with various H2A variants. For a comprehensive review of histone chaperone structures, see Reference 80. Together with the examples discussed above, this section illustrates how structural analyses help explain how different chaperones bind histones and their potential selectivity for histone variants, thereby providing an entry point to investigate the effect of mutations on histone chaperone– histone interactions and gain insight into chaperone function in vivo.

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

HISTONE CHAPERONES SHUTTLING HISTONES IN VARIOUS PATHWAYS In this section, we discuss histone chaperones that act on histones in transit (Figure 1). We focus on the best-characterized histones, H3 and H4 (for reviews on H2A–H2B chaperones, see References 81–83). We begin by describing the 496

Gurard-Levin

·

Quivy

·

Almouzni

presence of histone chaperones in histone complexes following their synthesis; we then discuss how the chaperones NASP and ASF1 shuttle histones into pathways for nucleosome assembly, storage, and degradation in response to cellular needs.

Histone Chaperones Acting on Histones in Transit Soluble histone complexes from cell lines that stably express epitope-tagged histones (45, 46) were purified to characterize subcomplexes of histones H3 and H4 in transit. A series of complexes, along with histone PTMs, were characterized (Figure 3a) (46, 51, 52, 84). Notably, these complexes are dynamic. Thus, additional transient or low-abundance subcomplexes and PTMs that are not detectable under the conditions of the complex purification and/or characterization may exist. Histone chaperones may aid nuclear import; some may increase the affinity for karyopherins that are responsible for transport (85; for a review, see Reference 81). Once in the nucleus, histone chaperones do not necessarily distribute histones to deposition factors for immediate incorporation into chromatin. Histones may persist in a soluble state depending on the variant and cell-cycle phase. In the next two sections, we describe the roles of the chaperones NASP and ASF1 in regulating histone provision and the storing and buffering of soluble histones H3.1–H4 and H3.3–H4.

Protecting H3–H4 from Degradation: NASP NASP was identified in a screen for histone binding proteins in testes. It exists in two isoforms: somatic NASP, which is ubiquitously expressed, and testicular NASP, which is highly expressed in testes and ovaries, as well as in transformed cells (50, 86). Although originally considered a chaperone for the linker histone H1 (50, 87–89), NASP has H3–H4 chaperone activity in vitro (90, 91) and its homologs in Saccharomyces cerevisiae (Hif1p) and in Saccharomyces pombe (Sim3) have H3–H4 chaperone activity in vivo (92, 93). The high sequence

BI83CH20-Almouzni

a

ARI

30 April 2014

11:12

Histone chaperones acting on histones after their synthesis i ii

Hsc70

iii

iv

H3

tNASP

sNASP Importin-4

Hsp90

HAT1 H3–H4

Hsp90

ASF1

ASF1 p48

Hsc70 H3–H4

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

H4

b

H3K9me1

H4K5K12ac

Histone chaperones in storage, buffering; protection of H3–H4 by NASP New H3–H4

NASP

Hsc70

Hsp90

Storage

NASP

NASP

NASP

Direct use

NASP ASF1

Soluble H3–H4 reservoir

Degradation (autophagy)

ASF1

Deposition

Figure 3 Histone chaperones acting on histones in transit. (a) Histone subcomplexes in transit following their synthesis. (i ) H3 with Hsc70. H3 is monomethylated at Lys9; H4 is with Hsc70 and Hsp90. (ii ) An H3–H4 dimer with Hsp90 and testicular nuclear autoantigenic sperm protein (tNASP). H3 is still monomethylated; a small fraction of H4 is acetylated at Lys12. (iii ) An H3–H4 dimer with somatic NASP (sNASP), histone acetyltransferase 1 (HAT1), antisilencing function 1 (ASF1), and RbAp48. H4 is acetylated at Lys5 and Lys12, but H3K9me1 has decreased. (iv) An H3–H4 dimer with Importin-4 and ASF1; H4 displays the Lys5 and Lys12 diacetylation pattern commonly associated with newly synthesized H4 (203). Note that these subcomplexes are dynamic and may not be unidirectional. (b) NASP maintains a soluble H3–H4 pool by protecting them from degradation by chaperone-mediated autophagy involving Hsc70 and Hsp90. Dispensable for normal proliferation, NASP is important for S-phase progression when H3–H4 provision is impaired, deals with an overload of new H3–H4, and deals with H3–H4 that suddenly accumulates during replication stress. The fine-tuning of the soluble reservoir of H3–H4 by NASP therefore integrates contingency into the histone supply network to cope with unanticipated changes in demand or provision. Modified from Reference 52.

identity between NASP and the H3–H4 chaperones N1/N2 in X. laevis, implicated in histone storage in oocytes (21), may provide a clue about its function in other species. Depletion

or overexpression of NASP in human cells does not yield a strong change to the cell cycle or perturb histone deposition. Only during acute replication stress in human cells, when new www.annualreviews.org • Histone Chaperones

497

ARI

30 April 2014

11:12

soluble histones cannot be deposited onto DNA, does NASP become critical through a storage function (52). Indeed, the storage of histones via NASP and their transfer to a multichaperone complex containing NASP, ASF1, and RbAp48 (8) allow deposition to restart once replication resumes. This reservoir of H3–H4 (51, 52), although dispensable during normal proliferative conditions, is needed to cope with unanticipated changes in demand or impaired histone production (52). NASP fine-tunes the capacity of this reservoir through a mechanism that involves a dynamic balance between histone protection and degradation by chaperonemediated autophagy (Figure 3b) (52). Intriguingly, this degradation pathway in mammals differs from that in yeast, in which histones in excess of cellular requirements are degraded in a Rad53- and proteasome-dependent pathway (94, 95). Thus, although some general principles may broadly apply in various species, the details of the mechanisms can differ and depend on the variant.

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

Directing Histone Traffic: ASF1 ASF1 (also known as CIA-1), originally identified as a transcriptional derepressor in a yeast genetic screen (96), was later described as an H3–H4 histone chaperone (61, 97, 98). ASF1 is the most conserved eukaryotic H3–H4 chaperone. In yeast, ASF1 exists as a single protein, whereas most vertebrates have two ASF1 paralogs, termed ASF1a and ASF1b in mammals. Among its conserved functions, ASF1 synergizes with CAF-1 and HIRA to channel H3.1 and H3.3 histones into distinct deposition pathways. Even though a high sequence identity and a conserved binding domain for histones and CAF-1/HIRA suggest common properties, ASF1a and ASF1b are not functionally equivalent, as evidenced by different gene-expression patterns (73, 99, 100), different protein–protein interactions (73, 101), and the finding that the human paralogs rescue different defects in yeast depleted of yASF1 (102). Comparative genomics approaches provided interesting clues about how the human paralogs evolved distinct functions. The phylogeny suggested that ASF1 498

Gurard-Levin

·

Quivy

·

Almouzni

experienced subfunctionalization, where ancestral functions are distributed between the two gene duplicates. Furthermore, the different genomic contexts allowed optimization of distinct functions (73). In support of subfunctionalization, biochemical approaches revealed that human ASF1a interacts selectively with HIRA in vitro and in vivo (65, 73, 101, 103) and that ASF1b has a preferential interaction for CAF-1 p60 in vivo (73). The productive combination of multiple approaches to elucidate the function of this histone chaperone could apply to various chaperones and even other protein families. As mentioned above, replication is one instance in which ASF1 is in the middle of heavy histone traffic, as we discuss below in more detail in the section titled Histone Chaperones’ Role During Replication: Handling Histones at the Fork. Importantly, by interfacing with the replication machinery (104) and associating with newly synthesized and parental histones (105), ASF1 may coordinate de novo histone deposition and histone recycling. These actions make ASF1 a critical factor in the regulation of chromatin inheritance, which can affect genome function and cell fate. This observation emphasizes the importance of histone chaperones acting on histones in transit.

HISTONE CHAPERONES INVOLVED IN DISTINCT HISTONE DEPOSITION PATHWAYS Several histone chaperones play a role in nucleosome dynamics at or near chromatin. In this section, we consider each histone chaperone individually and discuss the role of each in different deposition pathways for distinct H3 variants (Figure 4).

CAF-1 De Novo Deposition of Replicative H3.1–H4 in a DNA Synthesis–Coupled Pathway CAF-1 is a key factor that deposits histones in a manner coupled to DNA replication (28) and in DNA repair (106). CAF-1 is a complex comprising three subunits, termed p150, p60, and

BI83CH20-Almouzni

ARI

30 April 2014

a

11:12

b

c

H3.1–H4

H3.3–H4

HIRA

p150 p48

HIRA and DAXX complexes

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

DSC

DNA repair

HJURP ATRX

UBN1

CAF-1 complex

DNA replication

DAXX CABIN1

p60

CENP-A–H4

? DSI

DSI

Transient nucleosomefree regions

HJURP complex

Genes and regulatory elements

Telomeres

DSI

Centromeres

Figure 4 Histone chaperones involved in histone deposition pathways. (a) The chromatin assembly factor 1 (CAF-1) complex deposits the replicative H3.1 variant in a DNA synthesis–coupled (DSC) pathway during replication and repair. (b) The histone regulator A (HIRA) complex deposits the replacement variant H3.3 in a DNA synthesis–independent (DSI) pathway, particularly at transient nucleosome-free regions and at transcribed genes and regulatory elements. Death domain–associated protein (DAXX) may participate in the deposition of H3.3 at telomeres. (c) Holliday junction recognition protein (HJURP) deposits centromere protein A (CENP-A) in a DSI pathway. Abbreviations: ATRX, α-thalassemia/mental retardation X-linked; UBN1, ubinuclein 1.

RbAp48 (or simply p48) in human cells (66). The large subunit, p150, is crucial for coupling to DNA synthesis through an interaction with the proliferating cell nuclear antigen (PCNA) protein (106–109). This interaction is promoted by the phosphorylation of CAF1 p150 by Cdc7/Dbf4 (110). The p60 subunit is also a substrate for phosphorylation by Cdk in vitro, which may represent another mechanism by which CAF-1 deposition activity is regulated (111). Interestingly, chromatin assembly coupled to DNA synthesis is more efficient than assembly independent of DNA synthesis (33– 35), indicating that an advantage is conferred by this coupling. This finding accords with the fact that CAF-1 prominently deposits the replicative variant H3.1–H4 histones onto DNA at DNA synthesis sites; this deposition has been observed through the colocalization of CAF-1 with new H3.1, as detected with the SNAP assay (112). CAF-1 receives histone dimers provided by ASF1, as discussed above. A possible way for CAF-1 to then promote tetramer formation may be its capacity to dimerize through the p150 subunit, as determined in both human

and X. laevis CAF-1 orthologs (30). Interestingly, yeast CAF-1 can bind two H3–H4 dimers or a cross-linked (H3–H4)2 tetramer. This finding suggests that yeast CAF-1 may even assemble (H3–H4)2 tetramers prior to deposition (114). Finally, although CAF-1 function is conserved across several species, including Drosophila (115–117), Xenopus (30), Arabidopsis (118), and yeast (119, 120), its sequence has diverged, and the exact mechanisms of de novo deposition and/or their interfaces with various partners may differ accordingly and should be considered species specific. This general principle also applies to other histone deposition pathways.

Histone Chaperone Deposition of Replacement H3.3–H4 in a DNA Synthesis–Independent Pathway HIRA is a H3.3 chaperone that deposits this variant throughout interphase, independently of DNA synthesis (37, 45, 112). Although HIRA-mediated H3.3 deposition may potentially address all the needs for dynamics outside www.annualreviews.org • Histone Chaperones

499

ARI

30 April 2014

11:12

S phase, during S phase it can be considered a backup plan for faulty H3.1 deposition under CAF-1 guidance. In yeast, HIRA is a complex composed of Hir1p, Hir2p, Asf1p, Hir3p, and Hpc2p (103, 121–124). Interestingly, the sequence of human HIRA resembles that of Hir1p and Hir2p, suggesting that orthologs for the other components may also exist in human cells. Bioinformatics approaches have been critical for the identification of ubinuclein 1 (UBN1) (125) and calcineurin-binding protein 1 (CABIN1) (126, 127), the human orthologs of Hpc2p and Hir3p. Notably, UBN1 and CABIN1 had previously been identified in the soluble H3.3 complex by mass spectrometry (45), but their placement as members of the HIRA complex was not known. Thus, this strategy underscores the ability of bioinformatics and phylogenetic techniques to gain insights into histone chaperone function. Bioinformatics has also been critical in analyses of genome-wide data from chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) experiments (128). This technique provides an enrichment profile of a particular chromatin-bound factor from a mixed population of cells, and it was used to map the H3.3 enrichment profile in the presence of and following depletion of HIRA (112, 129). The HIRA binding sites were further confirmed by a HIRA ChIP-seq experiment (130). Notably, for proper enrichment profiles to be detected via these methods, the populations of cells being analyzed should display homogeneous behavior. Single cell–level analyses enable the detection of events that may escape genome-wide analyses. Thus, the combination of genome-wide approaches with individual cell analyses is essential. Moreover, enrichment data provide an end point and do not address the time frame of in vivo deposition assays, such as SNAP technology (112), that distinguish new and old histones and can provide further insight into histone chaperone function. ChIP-seq experiments have also generated a DAXX-dependent H3.3 enrichment profile that is distinct from HIRA (129, 131). Interestingly, when the SNAP assay is used, DAXX

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

500

Gurard-Levin

·

Quivy

·

Almouzni

depletion does not affect global H3.3 deposition in human HeLa cells (112). Whether DAXX-dependent H3.3 deposition plays a role at particular genomic loci in other cell types or is beyond the detection limits of the assay remains to be considered. Indeed, following neuronal activation, DAXX mediates H3.3 loading at regulatory regions of select genes in which calcium- and calcineurindependent phosphorylation dynamics regulate DAXX chaperone activity (132). DAXX can associate with the α-thalassemia/mental retardation X-linked (ATRX) protein, which is also found in the soluble H3.3 complex (45). A member of the Swi2/Snf2 family of chromatin remodelers (133), ATRX may recruit DAXX activity to specific genomic loci (129, 131). Together, HIRA and DAXX are representative chaperones that bind the same histone variant but demonstrate distinct cellular roles by interfacing with different cellular machinery.

HJURP Deposits CENP-A in a Cell Cycle–Dependent Manner HJURP represents a prototype of a histone chaperone that can define a chromatin landmark by depositing a particular histone variant at a specific locus (e.g., the centromere). Indeed, the centromere marks the site of kinetochore formation, which drives chromosome segregation. Except in budding yeast, in which the centromere is dictated by the underlying DNA sequence (134), the centromere is defined epigenetically by the presence of the H3 centromeric variant CENP-A in human and mouse (135). HJURP, initially characterized as a protein that binds to Holliday junctions (recombination intermediates) in vitro (136), was later described as a histone chaperone for human CENP-A after being purified from soluble CENP-A complexes (48, 49). Using the SNAP-tag approach, Jansen et al. (55) observed that newly synthesized CENP-A is deposited during late telophase and early G1 in human cells. This deposition timing coincided with the localization of HJURP at centromeres (48). Thus, during replication, new CENP-A

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

is not deposited and the concentration of CENP-A is halved in each daughter cell, giving rise to questions about the composition of the centromeric particle post-replication. Experiments using chromatin fibers to locate the presence of H3.3 during replication at centromeric regions suggested that H3.3 may act as a placeholder and is later replaced by CENP-A in an HJURP-dependent manner (137). Interestingly, a recent report showed that HJURP dimerization is required for CENP-A deposition (138); this property may ensure that centromeric nucleosome particles contain two copies of CENP-A (homotypic nucleosome), thereby contributing to the maintenance of this chromatin landmark during the dilution that eventually occurs during replication. A related HJURP in X. laevis (139) deposits CENP-A during early interphase. Finally, dedicated centromeric histone chaperones are also found in more distant species, including yeast (140). In Drosophila, an evolutionarily distinct protein, CAL1, has been proposed to be a functional homolog (141). Together, these data suggest that HJURP and its functional homologs are key factors for CENP-A deposition and the maintenance of centromere identity.

Histone Chaperones Participate in Cross Talk In S. cerevisiae, which has a single H3 that bears the strongest relationship to H3.3, deletion of either the CAF-1 or HIRA ortholog causes minor defects, whereas the double mutant is severely compromised (142, 143). These findings suggest that CAF-1 or HIRA may compensate for the loss of the other. When double mutants are robust, triple-mutant analyses provide a model system enabling the identification of cross talk. Indeed, this approach determined that the Swi/Snf component RdH54 can compensate for the loss of Cac1 (CAF-1 p150 ortholog) and Asf1 (144). In mammals, the H3.1 and H3.3 deposition pathways described above act in concert in vivo, and cross-talk mechanisms have been reported in different species. In mouse NIH3T3 cells depleted of CAF-1

subunits p150 and p60, nucleosomes are still detected, indicating that there are in vivo compensation mechanisms (113). Indeed, the SNAP assay revealed possible cross talk between the HIRA and CAF-1 deposition pathways in human cells. CAF-1 p60 depletion allows HIRA to place H3.3 at replication foci where it does not normally go (112). However, the converse is not true, and CAF-1 does not compensate for the depletion of HIRA (112). Interestingly, despite the common property to associate with H3.3, no evidence has demonstrated cross-talk mechanisms for HIRA and DAXX. Thus, when considering possible crosstalk mechanisms, it is important to question (a) how relaxed selectivity for histones can permit cross talk, particularly during stress conditions or following a change in histone provision, and (b) whether cross talk can cause pathological situations due to improper histone placement. The latter issue stresses that although histone chaperones may compensate for histone binding, they do not demonstrate perfect redundancy because they also interface with distinct partners. To further illustrate this point, in the following section we discuss how histone chaperones operate as an escort network in different DNA metabolic pathways.

HISTONE CHAPERONES OPERATE AS AN ESCORT NETWORK In this section, we discuss how histone chaperones act in concert in vivo. We begin by observing that histone chaperones exploit adapter domains that interface with several partners to regulate the handling of histones. We then consider these actions in the context of DNA replication, in response to DNA damage, and in the setting of DNA transcription.

Histone Chaperones’ Interface with Various DNA Processes at the Cellular Level: Adaptor Domains The presence of so-called adapter domains can help recruit or localize histone chaperones to www.annualreviews.org • Histone Chaperones

501

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Transcriptio n

Nu

RNA Pol II

ng filli ap eg om os cle

His ton ep roc es sin

TAFII250

g Tlk-1/2

Codanin-1 HIRA

NASP

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

DNA

CABIN1 UBN 1

DAXX

Telomeres

ASF1 a/b

ATRX p60

HJURP p48

?

?

p150

is hes ynt s A DN

CENP-A

PCNA

HP1

Ce n t rom ere s

MCM2-7

SetDB1

rmation/maintenan ce atin fo rom och ter He H3.1–H4

H3.3–H4

CENP-A–H4

Direct interaction

Interaction through histones

Figure 5 Histone chaperones interface with several partners to link histone metabolism to specific genomic regions or particular functions. Representative H3–H4 histone chaperones and several other binding partners broadly link the chaperones to various cellular processes. Note that antisilencing function 1a (ASF1a) specifically interacts with histone regulator A (HIRA), and ASF1b preferentially binds chromatin assembly factor 1 (CAF-1) p60. Abbreviations: ATRX, α-thalassemia/mental retardation X-linked; CENP-A, centromere protein A; HJURP, Holliday junction recognition protein; HP1, heterochromatin protein 1; NASP, nuclear autoantigenic sperm protein; MCM2-7, minichromosome maintenance protein 2-7; PCNA, proliferating cell nuclear antigen; Pol II, polymerase II; SetDB1, a histone lysine N-methyltransferase; Tlk-1/2, tousled-like kinases 1/2; UBN1, ubinuclein 1.

particular genomic loci or regulate chaperone activity for specific biological functions (Figure 5). A prime example is the coupling of CAF-1 to sites of DNA synthesis, mediated 502

Gurard-Levin

·

Quivy

·

Almouzni

by the interaction between p150 and PCNA (107, 108). Additionally, p150 interacts with heterochromatin protein 1 alpha (HP1α) (145) and the histone lysine N-methyltransferase

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

SetDB1 (146). Here, CAF-1 recruits these partners to defined chromosomal regions. The ability of HIRA to bind naked DNA (112) suggests that HIRA may deposit H3.3 at transiently nucleosome-depleted regions, possibly to maintain genome integrity. Also, the presence of HIRA in a complex with RNA polymerase II (Pol II) supports its recruitment to transcription sites, a finding supported by ChIP-seq data showing a correlation between HIRA-dependent H3.3 accumulation and RNA Pol II at transcript sites and specific regulatory elements (112). DAXX, through its interaction with ATRX, is implicated in the enrichment of H3.3 at telomeres, in ribosomal DNA, and in embryonic stem cells (129, 147). ATRX also recruits DAXX to pericentric heterochromatin in mouse embryonic fibroblasts (MEFs) (131). These examples demonstrate that adapter domains can yield histone chaperones that bind the same histone variant but carry out different functions. The use of adaptor domains can also regulate histone handling away from chromatin. ASF1 interacts with Codanin-1 through the same region that interacts with the B domain of HIRA and CAF-1. Codanin-1 is proposed to sequester ASF1 with histones in the cytoplasm, which would prevent ASF1 from donating histones to CAF-1 and HIRA for subsequent deposition (148). Furthermore, ASF1 interacts with several other partners, either directly or through histones, revealing a complex network with various functional outcomes that may differ between organisms. Thus, the use of adapter domains expands the interaction network and plays an integral role in dictating histone chaperone function.

Histone Chaperones’ Role During Replication: Handling Histones at the Fork Histone chaperones play a critical role in the transient disruption of chromatin organization during replication and its restoration after fork passage (Figure 6) (22, 149, 150). Ahead of the replication fork, a tetramer of (H3–H4)2

and two H2A–H2B dimers are evicted. Here, ASF1 may act as a histone acceptor, potentially disrupting the (H3–H4)2 tetramer (68, 70). Analyses of bulk histones demonstrated that an entire parental (H3–H4)2 tetramer can be recycled (151–157). SILAC (stable isotope labeling of amino acids in cell culture)-based mass spectrometry experiments designed to visualize whether endogenous and tagged histone dimers mix after replication demonstrated that (H3.1–H4)2 tetramers rarely mix, whereas approximately 10–20% of (H3.3–H4)2 tetramers do mix (158, 159). Importantly, this finding does not rule out the possibility that the parental tetramers experience a transient dimeric state and reassociate prior to deposition. Mixing requires splitting, but splitting does not necessarily lead to mixing. In addition to recycling parental histones, newly synthesized histones are deposited during replication, which accounts for the doubling of nucleosomes required to package the duplicated genome (Figure 6). Recycling and de novo deposition must be coordinated because the inheritance of chromatin marks can dictate cell fate (reviewed in Reference 160). ASF1, in escorting both parental and newly synthesized histones, may help coordinate these processes (for a review, see Reference 150). During de novo assembly, newly synthesized H3.1–H4 histones associated with ASF1 can be handed off to CAF-1 for subsequent deposition (39, 117). For recycling, ASF1 complexed with the MCM2-7 helicase may handle evicted parental histones and direct their distribution onto the daughter strands. Once the (H3–H4)2 tetramer is deposited onto DNA, H2A–H2B dimers rapidly associate to form the complete nucleosome. Although the dynamic nature of H2A–H2B has limited our understanding of this process, the FACT complex is possibly involved (161, 162), FACT binds several components of the replisome (163–166), it is required for replication in several organisms, and DT40 chicken cells lacking the FACT subunit structure-specific recognition protein 1 (SSRP1) experience reduced fork progression speed (167). Thus, replication represents a www.annualreviews.org • Histone Chaperones

503

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Nap1

FACT

Recycling

H2A–H2B

?

(H3–H4)2

?

FACT

?

?

HIRA

ASF1

? Old H3–H4 New H3.1–H4

Disruption

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

MCM2-7

New H3.3–H4 H2A–H2B

ASF1 as histone donor/acceptor

PCNA New PTM CAF-1 Old PTM ASF1b

De novo assembly Figure 6 Histone chaperones during replication: escorting histones at the fork. Nucleosomes are disrupted ahead of the replication fork, and parental histones are escorted by antisilencing function 1 (ASF1) (for H3–H4) and possibly FACT (facilitates chromatin transcription) (for H2A–H2B). Evicted (H3–H4)2 tetramers may proceed through a transient dimeric state. During recycling, ASF1 complexes with the minichromosome maintenance protein 2-7 (MCM2-7) helicase, an interaction mediated by histones. During de novo assembly, ASF1 donates newly synthesized H3.1–H4 dimers to chromatin assembly factor 1 (CAF-1), which is coupled to synthesis sites through an interaction with proliferating cell nuclear antigen (PCNA). After (H3–H4)2 tetramers are loaded onto DNA, H2A–H2B dimers rapidly associate, potentially aided by the FACT or Nap1 chaperones. In case of faulty CAF-1 deposition of H3.1–H4 at the fork (right), histone regulator A (HIRA) may deposit H3.3, away from the replication fork, in transiently nucleosome-depleted regions as a way to maintain genome integrity. Abbreviation: PTM, posttranslational modification.

critical time window in which the histone chaperone network is paramount, ensuring that the overall chromatin organization is properly duplicated to maintain genome integrity. Notably, histone chaperones are also vital in stress conditions, such as problems encountered during replication. As described above, ASF1 buffers histones that accumulate upon replication stress (104). Interestingly, if there are problems in the deposition of histones at the fork, HIRA may act away from the fork to fill in gaps (Figure 6), exploiting its DNA binding ability to deposit H3.3 at these transiently nucleosome-free regions (112). In the next section, we consider the role of chaperones in the 504

Gurard-Levin

·

Quivy

·

Almouzni

response to DNA damage, another type of cellular stress.

Histone Chaperones’ Place During DNA Damage Response Histone chaperones play key roles in responding to various types of DNA damage (31, 168– 172). In this section, we describe the role of histone chaperones in the response to DNA damage, with a particular focus on UV-C damage. The DNA damage response may be divided into three nonexclusive events: detection, processing, and restoration (Figure 7). During

BI83CH20-Almouzni

ARI

30 April 2014

11:12

DNA damage repair steps

Detection

UV-C damage

INO80 ? Arp8

γH2A.X

FACT

Chaperones involved

Priming HIRA Nucleosome sliding

Transcription Off

Displaced parental histones

Buffering chaperone?

HIRA

Processing Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

FACT INO80/Arp8 ?

Handling DNA synthesis

HIRA Others?

Recy ? cling

CAF-1

w

Ne

FACT

SWR1/ TIP60

Restoration

Assembly HIRA CAF-1 (p48, p60, p150)

INO80 H2A.Z Transcription On

H2A Restored nucleosomal organization

FACT

Figure 7 Histone chaperones in response to UV-C DNA damage. During the detection and priming steps, H2A.X phosphorylation initiates the signaling cascade and inhibits the H2A.X/H2A exchange mediated by FACT (facilitates chromatin transcription) (in mammals). Additionally, Arp8 recruits the INO80 complex to damage sites (218). Histone regulator A (HIRA) is recruited early, perhaps in a ubiquitin-dependent manner. Other histone modifying enzymes are important in the response to DNA damage, and histone chaperones may be present in these complexes. During the processing and handling steps, parental histones are displaced from the damage site to provide access and may be buffered by antisilencing function 1 (ASF1). HIRA then promotes the deposition of H3.3 and exits. During restoration and assembly, the chromatin assembly factor 1 (CAF-1) complex and FACT are involved in the deposition of newly synthesized H3.1–H4 and H2A–H2B, respectively. Finally, nucleosome restoration and γH2A.X exchange, aided by the INO80 complex, contribute to turn off DNA damage signaling. Modified from Reference 168.

detection, the presence of phosphorylated H2A.X, termed γH2A.X in mammals (173), is an early marker for DNA damage (174). To provide access for the DNA repair machinery, the parental histones flanking the DNA damage site are transiently displaced and thus escorted by histone chaperones during the processing event. ASF1 may handle evicted H3–H4 dimers, similarly to the way in which it buffers histones that accumulate in response to

a DNA replication fork block (104). The HIRA complex is recruited very early to UV-C damage regions at the detection step, potentially through a ubiquitin-dependent mechanism in which ubiquitylated proteins, including histones, may be displaced and transient access to DNA is provided. HIRA uses its DNA binding property to promote H3.3 deposition at damage sites and then leaves. This transient action of HIRA, which leads to new H3.3 deposition, www.annualreviews.org • Histone Chaperones

505

ARI

30 April 2014

11:12

acts as a marking system, licensing transcription restart following DNA damage repair (175). During the later steps of this process, the CAF1 complex is recruited to the DNA synthesis repair site for the restoration event. CAF-1 recruitment can occur through the interaction with PCNA (107), and de novo H3.1–H4 histones are deposited (40). CAF-1-mediated H3.1–H4 deposition participates to reassemble nucleosomal organization and may provide a signal to terminate the DNA damage response (168). For H2A–H2B, the FACT complex facilitates accelerated exchange of H2A–H2B that is also important for transcription restart (176). The link between the DNA damage response to UV-C and transcription demonstrates that DNA metabolic processes are interdependent and that the cellular response to one process can affect the other and, therefore, cell fate.

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

Histone Chaperones’ Participation During Transcription During transcription, changes to the chromatin structure accompany the progression of the RNA Pol II machinery (177–179). The recruitment of several classes of chromatin regulators, including histone chaperones, to transcription sites can facilitate a local rearrangement of the chromatin structure to permit the passage of RNA Pol II (for reviews, see References 31 and 180). Additionally, these factors can help regulate histone exchange to maintain chromatin integrity during transcription elongation (181, 182). One mechanism by which this process occurs is that histone chaperones can handle and buffer histones displaced ahead of the polymerase, thereby functioning as a so-called histone sink (183). Indeed, several histone chaperones have been implicated in accepting H2A–H2B dimers to facilitate transcription factor binding; these include yeast Nap1 and nucleoplasmin (184, 185), human FACT (58), and nucleolin (26). Following the passage of RNA Pol II, the reassembly of nucleosomes restores the chromatin structure, preventing cryptic transcription; ASF1, FACT, and Spt6 may facilitate this process (58, 186, 187). 506

Gurard-Levin

·

Quivy

·

Almouzni

The targeting of histone chaperones to transcription sites may occur through interactions with the transcriptional machinery or transcription-associated chromatin marks by the use of the adapter domains described above. For example, human Spt6 and HIRA have been found in complex with RNA Pol II (112, 188), and ASF1 associates with TAFII250, the largest subunit of the TFIID transcription factor (189). Finally, interactions between histone chaperones and chromatin remodelers may recruit histone chaperones to transcription sites, as demonstrated by FACT and the CHD1 chromatin remodeler (190). Thus, similarly to replication and repair, transcription represents another transient disruption to the chromatin organization and another window of opportunity to either maintain or alter the chromatin landscape. Here we consider the role of histone chaperones in facilitating these processes, including the reassembly of nucleosomes via a combination of evicted parental and newly synthesized histones to restore the chromatin organization. This balance, if perturbed, may slow down processes that reestablish chromatin marks at transcription sites that dictate genome function. This possibility suggests that regulation of histone chaperone function, to achieve timely and efficient handling of histones, in conjunction with histone-modifying enzymes may help control and maintain the timely execution of the specific expression profile that dictates cell identity.

DISCUSSION AND PERSPECTIVES This review addresses the definition of histone chaperones and their selectivity for cognate histones. Along with structural data and evolutionary studies, the use of complementary in vitro and in vivo assays enabled the elucidation of several functional aspects of histone chaperones, including their roles in histone metabolism, namely storage, buffering, nuclear import, deposition, and degradation. By interfacing with additional partners, adapter

BI83CH20-Almouzni

ARI

30 April 2014

11:12

domains can regulate histone chaperone activity through recruitment to particular genomic loci or during specific biological processes. Together, these observations at the molecular and cellular levels enabled the placement of various histone chaperones in an escort network that shuttles histones throughout their cellular life and affects all aspects of histone dynamics.

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Histone Chaperone Integration at the Organism Level During Development and Reprogramming The role of histone chaperones in the disruption of the chromatin organization intimately links these proteins with development and reprogramming (for a review, see Reference 42). The role in development is illustrated by evidence that knockout mice for several histone chaperones, including HIRA, DAXX, ASF1a, and CAF-1 p150, are embryonic lethal (Table 2) (191–195; for ASF1b, see http://www.informatics.jax.org). This effect may arise from improper placement of particular histone variants over space and time, at either a defined developmental stage or a specific genomic location. Indeed, in Drosophila and in mice, after fertilization but before the first round of replication, H3.3 is deposited globally in the paternal chromatin (196), a process that requires HIRA (197). Furthermore, in the early development of X. laevis, H3.3 expression peaks at gastrulation and HIRA promotes its deposition. Interfering with morpholinos to titrate out either HIRA or H3.3 impairs normal development by impeding progression through late gastrulation and affecting mesoderm induction. Interestingly, the morpholino titration of all H3 causes defects at a very early gastrulation stage (198). The most dramatic effect is observed with a dominant-negative strategy targeting CAF-1. This process causes early developmental arrest at the midblastula transition (30). Additionally, the reprogramming of somatic cell nuclei by oocytes illustrates that HIRA-dependent H3.3 deposition is necessary during global changes in transcription that accompany changes in cell fate (199). Thus,

Table 2 Histone chaperone knockout studies in mouse and corresponding phenotype Histone chaperone

Phenotype

Reference

ASF1a

Embryonic lethal

ASF1b

Viable

195

p150 CAF-1

Embryonic lethal at 8–16-cell stage; abnormal PCH

191

HIRA

Embryonic lethal at E11; abnormal gastrulation

192

http://www.informatics. jax.org

DAXX

Embryonic lethal at E9.5

194

ATRX

Embryonic lethal at E9.5 pc; abnormal trophoblast development

193

Abbreviations: ASF1, antisilencing function 1; ATRX, α-thalassemia/mental retardation X-linked; CAF-1, chromatin assembly factor 1; DAXX, death domain–associated protein; E, embryonic day; HIRA, histone regulator A; PCH, pericentric heterochromatin; pc, postcoitus.

histone chaperones not only help maintain chromatin organization during normal lineage propagation, but also help facilitate changes in the chromatin landscape that enable different expression profiles to regulate developmental programs.

Histone Chaperone Misfunction and Disease Several studies recently identified a connection between histone chaperones and disease (for a review, see Reference 82). Evidence for this finding is that mutations in DAXX and ATRX appear in 44% of pediatric glioblastoma tumor samples (200). These mutations may cause errors in the timing and placement of particular histone variants. Also, the expression level of histone chaperones can affect cell fate, as illustrated by research showing that human ASF1b, but not ASF1a, has a major role in proliferation and is a marker for breast cancer (100), having prognostic value for metastasis (201). Thus, histone chaperones could represent new opportunities in drug discovery, both in prognosis and as therapeutic targets. www.annualreviews.org • Histone Chaperones

507

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Questions About Histone Chaperones for Future Perspectives

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Based on the current knowledge as discussed in this review, new interesting questions have arisen. How are histone chaperones regulated in terms of their expression and availability during the cell cycle? In this respect, the role of histone chaperones in their own disposal remains to be explored, and how this may link with histones that are marked for disposal or those that are to be protected. How does HJURP specifically target centromeric chromatin, and what is the role of dimerization of various histone chaperones in depositing histones? In these areas,

structural analyses with additional (full-length) partners are needed to better understand the molecular basis for these interactions and to establish larger complexes, perhaps in combination with mass spectrometry approaches to identify interactions between large complexes at the single-residue level (202). The link between histone chaperones, development, and pathology is an area that will continue to receive much attention, will shed more light on the role of histone chaperones in the maintenance of genome integrity and function, and will find applications in drug discovery for disease treatment.

SUMMARY POINTS 1. Histone chaperones are involved in histone transfer but are not necessarily part of the final product. 2. Histone chaperones escort histones throughout their cellular life in all aspects of histone metabolism, including regulation of their provision, their placement within chromatin, eviction, and degradation. 3. Histone chaperone binding to histones and variant selectivity have been illuminated by structural studies. 4. Histone chaperones provide interfaces that allow their recruitment to particular genomic loci or that link to specific biological processes. 5. Histone chaperones operate in an escort network and are implicated in DNA metabolic processes such as replication, repair, and transcription. Future studies should investigate rescue through cross talk with other chaperones. 6. The role of histone chaperones in development and disease is an area of interest.

DISCLOSURE STATEMENT The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

ACKNOWLEDGMENTS We thank Dominique Ray-Gallet and Marcus Buschbeck for critical reading of the manuscript. The authors acknowledge their affiliation with PSL, Sorbonne University. G.A.’s laboratory is supported by La Ligue contre le Cancer (Equipe Labellis´ee and postdoctoral fellowship to Z.A.G-L.), the European Commission Network of Excellence EpiGeneSys (HEALTH-F4-2010-257082), ERC Advanced Grant 2009-AdG_20090506 “Eccentric,” the European Commission largescale integrating project FP7_HEALTH-2010-259743 “MODHEP,” ANR “ChromaTin” ANR10-BLAN-1326-03, ANR-11-LABX-0044_DEEP and ANR-10-IDEX-0001-02 PSL, ANR “CHAPINHIB,” ANR-12-BSV5-0022-02, and Aviesan–ITMO cancer project “Epigenomics of Breast Cancer.” 508

Gurard-Levin

·

Quivy

·

Almouzni

BI83CH20-Almouzni

ARI

30 April 2014

11:12

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

LITERATURE CITED 1. Kornberg RD. 1974. Chromatin structure: a repeating unit of histones and DNA. Science 184:868–71 2. Oudet P, Gross-Bellard M, Chambon P. 1975. Electron microscopic and biochemical evidence that chromatin structure is a repeating unit. Cell 4:281–300 3. McGhee JD, Felsenfeld G. 1980. Nucleosome structure. Annu. Rev. Biochem. 49:1115–56 4. Luger K, M¨ader AW, Richmond RK, Sargent DF, Richmond TJ. 1997. Crystal structure of the nucleosome core particle at 2.8 A˚resolution. Nature 389:251–60 5. Kim UJ, Han M, Kayne P, Grunstein M. 1988. Effects of histone H4 depletion on the cell cycle and transcription of Saccharomyces cerevisiae. EMBO J. 7:2211–19 6. Prado F, Aguilera A. 2005. Partial depletion of histone H4 increases homologous recombination– mediated genetic instability. Mol. Cell. Biol. 25:1526–36 7. Meeks-Wagner D, Hartwell LH. 1986. Normal stoichiometry of histone dimer sets is necessary for high fidelity of mitotic chromosome transmission. Cell 44:43–52 8. Groth A, Ray Gallet D, Quivy J-P, Lukas J, Bartek J, Almouzni G. 2005. Human Asf1 regulates the flow of S phase histones during replicational stress. Mol. Cell 17:301–11 9. Laskey RA, Honda BM, Mills AD, Finch JT. 1978. Nucleosomes are assembled by an acidic protein which binds histones and transfers them to DNA. Nature 275:416–20 10. Ellis RJ. 1990. The molecular chaperone concept. Semin. Cell Biol. 1:1–9 11. Felsenfeld G. 1978. Chromatin. Nature 271:115–22 12. Nelson T, Wiegand R, Brutlag D. 1981. Ribonucleic acid and other polyanions facilitate chromatin assembly in vitro. Biochemistry 20:2594–601 13. Stein A. 1989. Reconstitution of chromatin from purified components. Methods Enzymol. 170:585–603 14. Stein A, Whitlock JP Jr, Bina M. 1979. Acidic polypeptides can assemble both histones and chromatin in vitro at physiological ionic strength. Proc. Natl. Acad. Sci. USA 76:5000–4 15. Woodland HR, Adamson ED. 1977. The synthesis and storage of histones during the oogenesis of Xenopus laevis. Dev. Biol. 57:118–35 16. Laskey RA, Mills AD, Morris NR. 1977. Assembly of SV40 chromatin in a cell-free system from Xenopus eggs. Cell 10:237–43 17. Nelson T, Hsieh TS, Brutlag D. 1979. Extracts of Drosophila embryos mediate chromatin assembly in vitro. Proc. Natl. Acad. Sci. USA 76:5510–14 18. Adamson ED, Woodland HR. 1974. Histone synthesis in early amphibian development: histone and DNA syntheses are not co-ordinated. J. Mol. Biol. 88:263–85 19. Earnshaw WC, Honda BM, Laskey RA, Thomas JO. 1980. Assembly of nucleosomes: the reaction involving X. laevis nucleoplasmin. Cell 21:373–83 20. Kleinschmidt JA, Franke WW. 1982. Soluble acidic complexes containing histones H3 and H4 in nuclei of Xenopus laevis oocytes. Cell 29:799–809 21. Kleinschmidt JA, Fortkamp E, Krohne G, Zentgraf H, Franke WW. 1985. Co-existence of two different types of soluble histone complexes in nuclei of Xenopus laevis oocytes. J. Biol. Chem. 260:1166–76 22. Kaufman DG, Almouzni G. 2006. Chromatin assembly. In DNA Replication and Human Disease, ed. ML DePamphilis, pp. 121–40. New York: Cold Spring Harb. Lab. 23. Gaume X, Monier K, Argoul F, Mongelard F, Bouvet P. 2011. In vivo study of the histone chaperone activity of nucleolin by FRAP. Biochem. Res. Int. 12:1–15 24. Cong R, Das S, Douet J, Wong J, Buschbeck M, et al. 2013. macroH2A1 histone variant represses rDNA transcription. Nucleic Acids Res. 42:181–92 25. Ratnakumar K, Duarte LF, LeRoy G, Hasson D, Smeets D, et al. 2012. ATRX-mediated chromatin association of histone variant macroH2A1 regulates α-globin expression. Genes Dev. 26:433–38 26. Angelov D, Bondarenko VA, Almagro S, Menoni H, Mong´elard F, et al. 2006. Nucleolin is a histone chaperone with FACT-like activity and assists remodeling of nucleosomes. EMBO J. 25:1669–79 27. Stillman B, Gluzman Y. 1985. Replication and supercoiling of simian virus 40 DNA in cell extracts from human cells. Mol. Cell. Biol. 5:2051–60 28. Stillman B. 1986. Chromatin assembly during SV40 DNA replication in vitro. Cell 45:555–65 www.annualreviews.org • Histone Chaperones

509

ARI

30 April 2014

11:12

29. Smith S, Stillman B. 1989. Purification and characterization of CAF-I, a human cell factor required for chromatin assembly during DNA replication in vitro. Cell 58:15–25 30. Quivy J-P, Grandi P, Almouzni G. 2001. Dimerization of the largest subunit of chromatin assembly factor 1: importance in vitro and during Xenopus early development. EMBO J. 20:2015–27 31. De Koning L, Corpet A, Haber JE, Almouzni G. 2007. Histone chaperones: an escort network regulating histone traffic. Nat. Struct. Mol. Biol. 14:997–1007 32. Dilworth SM, Black SJ, Laskey RA. 1987. Two complexes that contain histones are required for nucleosome assembly in vitro: role of nucleoplasmin and N1 in Xenopus egg extracts. Cell 51:1009–18 33. Almouzni G, Mechali M. 1988. Xenopus egg extracts: a model system for chromatin replication. Biochim. Biophys. Acta 951:443–50 34. Almouzni G, Mechali M. 1988. Assembly of spaced chromatin involvement of ATP and DNA topoisomerase activity. EMBO J. 7:4355–65 35. Almouzni G, Mechali M. 1988. Assembly of spaced chromatin promoted by DNA synthesis in extracts from Xenopus eggs. EMBO J. 7:665–72 36. Ray Gallet D, Almouzni G. 2004. DNA synthesis–dependent and –independent chromatin assembly pathways in Xenopus egg extracts. Methods Enzymol. 375:117–31 37. Ray Gallet D, Quivy J-P, Scamps C, Martini EM-D, Lipinski M, Almouzni G. 2002. HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol. Cell 9:1091–100 38. Sharp JA, Fouts ET, Krawitz DC, Kaufman PD. 2001. Yeast histone deposition protein Asf1p requires Hir proteins and PCNA for heterochromatic silencing. Curr. Biol. 11:463–73 39. Mello JA, Sillj´e HH, Roche DM, Kirschner DB, Nigg EA, Almouzni G. 2002. Human Asf1 and CAF-1 interact and synergize in a repair-coupled nucleosome assembly pathway. EMBO Rep. 3:329–34 40. Polo SE, Roche D, Almouzni G. 2006. New histone incorporation marks sites of UV repair in human cells. Cell 127:481–93 41. Szenker E, Ray Gallet D, Almouzni G. 2011. The double face of the histone variant H3.3. Cell Res. 21:421–34 42. Filipescu D, Szenker E, Almouzni G. 2013. Developmental roles of histone H3 variants and their chaperones. Trends Genet. 29:630–40 43. Boyarchuk E, Montes de Oca R, Almouzni G. 2011. Cell cycle dynamics of histone variants at the centromere, a model for chromosomal landmarks. Curr. Opin. Cell Biol. 23:266–76 44. Vardabasso C, Hasson D, Ratnakumar K, Chung CY, Duarte LF, Bernstein E. 2013. Histone variants: emerging players in cancer biology. Cell. Mol. Life Sci. 71:379–404 45. Tagami H, Ray Gallet D, Almouzni G, Nakatani Y. 2004. Histone H3.1 and H3.3 complexes mediate nucleosome assembly pathways dependent or independent of DNA synthesis. Cell 116:51–61 46. Loyola A, Bonaldi T, Roche D, Imhof A, Almouzni G. 2006. PTMs on H3 variants before chromatin assembly potentiate their final epigenetic state. Mol. Cell 24:309–16 47. Lewis PW, Els¨asser SJ, Noh K-M, Stadler SC, Allis CD. 2010. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc. Natl. Acad. Sci. USA 107:14075–80 48. Dunleavy EM, Roche D, Tagami H, Lacoste N, Ray Gallet D, et al. 2009. HJURP is a cell-cycledependent maintenance and deposition factor of CENP-A at centromeres. Cell 137:485–97 49. Foltz DR, Jansen LE, Bailey AO, Yates JR 3rd, Bassett EA, et al. 2009. Centromere-specific assembly of CENP-A nucleosomes is mediated by HJURP. Cell 137:472–84 50. Richardson RT, Batova IN, Widgren EE, Zheng LX, Whitfield M, et al. 2000. Characterization of the histone H1–binding protein, NASP, as a cell cycle–regulated somatic protein. J. Biol. Chem. 275:30378– 86 51. Campos EI, Fillingham J, Li G, Zheng H, Voigt P, et al. 2010. The program for processing newly synthesized histones H3.1 and H4. Nat. Struct. Mol. Biol. 17:1343–51 52. Cook AJL, Gurard-Levin ZA, Vassias I, Almouzni G. 2011. A specific function for the histone chaperone NASP to fine-tune a reservoir of soluble H3–H4 in the histone supply chain. Mol. Cell 44:918–27 53. Shintomi K, Iwabuchi M, Saeki H, Ura K, Kishimoto T, Ohsumi K. 2005. Nucleosome assembly protein 1 is a linker histone chaperone in Xenopus eggs. Proc. Natl. Acad. Sci. USA 102:8210–15

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

510

Gurard-Levin

·

Quivy

·

Almouzni

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

54. Keppler A, Gendreizig S, Gronemeyer T, Pick H, Vogel H, Johnsson K. 2003. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat. Biotechnol. 21:86–89 55. Jansen LE, Black BE, Foltz DR, Cleveland DW. 2007. Propagation of centromeric chromatin requires exit from mitosis. J. Cell Biol. 176:795–805 56. Mousson F, Lautrette A, Thuret J-Y, Agez M, Courbeyrette R, et al. 2005. Structural basis for the interaction of Asf1 with histone H3 and its functional implications. Proc. Natl. Acad. Sci. USA 102:5975– 80 57. Park Y-J, Luger K. 2006. The structure of nucleosome assembly protein 1. Proc. Natl. Acad. Sci. USA 103:1248–53 58. Belotserkovskaya R, Oh S, Bondarenko VA, Orphanides G, Studitsky VM, Reinberg D. 2003. FACT facilitates transcription-dependent nucleosome alteration. Science 301:1090–93 59. Regnard C, Desbruy`eres E, Huet JC, Beauvallet C, Pernollet JC, Edd´e B. 2000. Polyglutamylation of nucleosome assembly proteins. J. Biol. Chem. 275:15969–76 60. Dutta S, Akey IV, Dingwall C, Hartman KL, Laue T, et al. 2001. The crystal structure of nucleoplasmin core: implications for histone binding and nucleosome assembly. Mol. Cell 8:841–53 61. Umehara T, Chimura T, Ichikawa N, Horikoshi M. 2002. Polyanionic stretch-deleted histone chaperone cia1/Asf1p is functional both in vivo and in vitro. Genes Cells Devoted Mol. Cell. Mech. 7:59–73 62. Muto S, Senda M, Akai Y, Sato L, Suzuki T, et al. 2007. Relationship between the structure of SET/TAFIβ/INHAT and its histone chaperone activity. Proc. Natl. Acad. Sci. USA 104:4285–90 63. Namboodiri VM, Dutta S, Akey IV, Head JF, Akey CW. 2003. The crystal structure of Drosophila NLP core provides insight into pentamer formation and histone binding. Structure 11:175–86 64. Park YJ, McBryant SJ, Luger K. 2008. A β-hairpin comprising the nuclear localization sequence sustains the self-associated states of nucleosome assembly protein 1. J. Mol. Biol. 375:1076–85 65. Daganzo SM, Erzberger JP, Lam WM, Skordalakes E, Zhang R, et al. 2003. Structure and function of the conserved core of histone deposition protein Asf1. Curr. Biol. 13:2148–58 66. Kaufman PD, Kobayashi R, Kessler N, Stillman B. 1995. The p150 and p60 subunits of chromatin assembly factor I: a molecular link between newly synthesized histones and DNA replication. Cell 81:1105–14 67. Verreault A, Kaufman PD, Kobayashi R, Stillman B. 1996. Nucleosome assembly by a complex of CAF-1 and acetylated histones H3/H4. Cell 87:95–104 68. Natsume R, Eitoku M, Akai Y, Sano N, Horikoshi M, Senda T. 2007. Structure and function of the histone chaperone CIA/ASF1 complexed with histones H3 and H4. Nature 446:338–41 69. Corpet A, Almouzni G. 2009. Making copies of chromatin: the challenge of nucleosomal organization and epigenetic information. Trends Cell Biol. 19:29–41 70. English CM, Adkins MW, Carson JJ, Churchill ME, Tyler JK. 2006. Structural basis for the histone chaperone activity of Asf1. Cell 127:495–508 71. Antczak AJ, Tsubota T, Kaufman PD, Berger JM. 2006. Structure of the yeast histone H3–ASF1 interaction: implications for chaperone mechanism, species-specific interactions, and epigenetics. BMC Struct. Biol. 6:26 72. Malay AD, Umehara T, Matsubara-Malay K, Padmanabhan B, Yokoyama S. 2008. Crystal structures of fission yeast histone chaperone Asf1 complexed with the Hip1 B-domain or the Cac2 C terminus. J. Biol. Chem. 283:14022–31 73. Abascal F, Corpet A, Gurard-Levin ZA, Juan D, Ochsenbein F, et al. 2013. Subfunctionalization via adaptive evolution influenced by genomic context: the case of histone chaperones ASF1a and ASF1b. Mol. Biol. Evol. 30:1853–66 74. Els¨asser SJ, Huang H, Lewis PW, Chin JW, Allis CD, Patel DJ. 2012. DAXX envelops a histone H3.3– H4 dimer for H3.3-specific recognition. Nature 491:560–65 75. Hu H, Liu Y, Wang M, Fang J, Huang H, et al. 2011. Structure of a CENP-A-histone H4 heterodimer in complex with chaperone HJURP. Genes Dev. 25:901–6 76. Zhou Z, Feng H, Zhou B-R, Ghirlando R, Hu K, et al. 2011. Structural basis for recognition of centromere histone variant CenH3 by the chaperone Scm3. Nature 472:234–37 77. Bowman A, Ward R, Wiechens N, Singh V, El-Mkami H, et al. 2011. The histone chaperones Nap1 and Vps75 bind histones H3 and H4 in a tetrameric conformation. Mol. Cell 41:398–408 www.annualreviews.org • Histone Chaperones

511

ARI

30 April 2014

11:12

78. Lacoste N, Woolfe A, Tachiwana H, Villar Garea A, Barth T, et al. 2014. Mislocalization of the centromere histone variant CenH3/CENP-A in human cells depends on the chaperone DAXX. Mol. Cell 53:631–44 79. Hondele M, Stuwe T, Hassler M, Halbach F, Bowman A, et al. 2013. Structural basis of histone H2A– H2B recognition by the essential chaperone FACT. Nature 499:111–14 80. Hondele M, Ladurner AG. 2011. The chaperone-histone partnership: for the greater good of histone traffic and chromatin plasticity. Curr. Opin. Struct. Biol. 21:698–708 81. Keck KM, Pemberton LF. 2012. Histone chaperones link histone nuclear import and chromatin assembly. Biochim. Biophys. Acta 1819:277–89 82. Burgess RJ, Zhang Z. 2013. Histone chaperones in nucleosome assembly and human disease. Nat. Struct. Mol. Biol. 20:14–22 83. Eitoku M, Sato L, Senda T, Horikoshi M. 2008. Histone chaperones: 30 years from isolation to elucidation of the mechanisms of nucleosome assembly and disassembly. Cell. Mol. Life Sci. 65:414–44 84. Alvarez F, Munoz ˜ F, Schilcher P, Imhof A, Almouzni G, Loyola A. 2011. Sequential establishment of marks on soluble histones H3 and H4. J. Biol. Chem. 286:17714–21 85. Mosammaparast N, Ewart CS, Pemberton LF. 2002. A role for nucleosome assembly protein 1 in the nuclear transport of histones H2A and H2B. EMBO J. 21:6527–38 86. Welch JE, Zimmerman LJ, Joseph DR, O’Rand MG. 1990. Characterization of a sperm-specific nuclear autoantigenic protein. I. Complete sequence and homology with the Xenopus protein, N1/N2. Biol. Reprod. 43:559–68 87. Alekseev OM, Bencic DC, Richardson RT, Widgren EE, O’Rand MG. 2003. Overexpression of the Linker histone-binding protein tNASP affects progression through the cell cycle. J. Biol. Chem. 278:8846– 52 88. Alekseev OM, Richardson RT, Pope MR, O’Rand MG. 2005. Mass spectrometry identification of NASP binding partners in HeLa cells. Proteins 61:1–5 89. Finn RM, Browne K, Hodgson KC, Ausio´ J. 2008. sNASP, a histone H1–specific eukaryotic chaperone dimer that facilitates chromatin assembly. Biophys. J. 95:1314–25 90. Wang H, Walsh STR, Parthun MR. 2008. Expanded binding specificity of the human histone chaperone NASP. Nucleic Acids Res. 36:5763–72 91. Osakabe A, Tachiwana H, Matsunaga T, Shiga T, Nozawa RS, et al. 2010. Nucleosome formation activity of human somatic nuclear autoantigenic sperm protein (sNASP). J. Biol. Chem. 285:11913–21 92. Dunleavy EM, Pidoux AL, Monet M, Bonilla C, Richardson W, et al. 2007. A NASP (N1/N2)-related protein, Sim3, binds CENP-A and is required for its deposition at fission yeast centromeres. Mol. Cell 28:1029–44 93. Ai X, Parthun MR. 2004. The nuclear Hat1p/Hat2p complex: a molecular link between type B histone acetyltransferases and chromatin assembly. Mol. Cell 14:195–205 94. Gunjan A, Verreault A. 2003. A Rad53 kinase–dependent surveillance mechanism that regulates histone protein levels in S. cerevisiae. Cell 115:537–49 95. Singh RK, Kabbaj MH, Paik J, Gunjan A. 2009. Histone levels are regulated by phosphorylation and ubiquitylation-dependent proteolysis. Nat. Cell Biol. 11:925–33 96. Le S, Davis C, Konopka JB, Sternglanz R. 1997. Two new S-phase-specific genes from Saccharomyces cerevisiae. Yeast 13:1029–42 97. Tyler JK, Adams CR, Chen SR, Kobayashi R, Kamakaka RT, Kadonaga JT. 1999. The RCAF complex mediates chromatin assembly during DNA replication and repair. Nature 402:555–60 98. Munakata T, Adachi N, Yokoyama N, Kuzuhara T, Horikoshi M. 2000. A human homologue of yeast anti-silencing factor has histone chaperone activity. Genes Cells Devoted Mol. Cell. Mech. 5:221–33 99. Umehara TT, Horikoshi MM. 2003. Transcription initiation factor IID–interactive histone chaperone CIA-II implicated in mammalian spermatogenesis. J. Biol. Chem. 278:35660–67 100. Corpet A, De Koning L, Toedling J, Savignoni A, Berger F, et al. 2011. Asf1b, the necessary Asf1 isoform for proliferation, is predictive of outcome in breast cancer. EMBO J. 30:480–93 101. Tang Y, Poustovoitov MV, Zhao K, Garfinkel M, Canutescu A, et al. 2006. Structure of a human ASF1a– HIRA complex and insights into specificity of histone chaperone complex assembly. Nat. Struct. Mol. Biol. 13:921–29

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

512

Gurard-Levin

·

Quivy

·

Almouzni

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

102. Tamburini BA, Carson JJ, Adkins MW, Tyler JK. 2005. Functional conservation and specialization among eukaryotic anti-silencing function 1 histone chaperones. Eukaryot. Cell 4:1583–90 103. Green EM, Antczak AJ, Bailey AO, Franco AA, Wu KJ, et al. 2005. Replication-independent histone deposition by the HIR complex and Asf1. Curr. Biol. 15:2044–49 104. Groth A, Corpet A, Cook AJ, Roche D, Bartek J, et al. 2007. Regulation of replication fork progression through histone supply and demand. Science 318:1928–31 105. Jasencakova Z, Scharf AN, Ask K, Corpet A, Imhof A, et al. 2010. Replication stress interferes with histone recycling and predeposition marking of new histones. Mol. Cell 37:736–43 106. Gaillard PH, Martini EM, Kaufman PD, Stillman B, Moustacchi E, Almouzni G. 1996. Chromatin assembly coupled to DNA repair: a new role for chromatin assembly factor I. Cell 86:887–96 107. Moggs JG, Grandi P, Quivy J-P, Jonsson ZO, Hubscher U, et al. 2000. A CAF-1–PCNA–mediated ´ ¨ chromatin assembly pathway triggered by sensing DNA damage. Mol. Cell. Biol. 20:1206–18 108. Shibahara K, Stillman B. 1999. Replication-dependent marking of DNA by PCNA facilitates CAF-1coupled inheritance of chromatin. Cell 96:575–85 109. Martini E, Roche DM, Marheineke K, Verreault A, Almouzni G. 1998. Recruitment of phosphorylated chromatin assembly factor 1 to chromatin after UV irradiation of human cells. J. Cell Biol. 143:563–75 110. G´erard A, Koundrioukoff S, Ramillon V, Serg`ere J-C, Mailand N, et al. 2006. The replication kinase Cdc7–Dbf4 promotes the interaction of the p150 subunit of chromatin assembly factor 1 with proliferating cell nuclear antigen. EMBO Rep. 7:817–23 111. Keller C, Krude T. 2000. Requirement of cyclin/Cdk2 and protein phosphatase 1 activity for chromatin assembly factor 1–dependent chromatin assembly during DNA synthesis. J. Biol. Chem. 275:35512–21 112. Ray Gallet D, Woolfe A, Vassias I, Pellentz C, Lacoste N, et al. 2011. Dynamics of histone H3 deposition in vivo reveal a nucleosome gap-filling mechanism for H3.3 to maintain chromatin integrity. Mol. Cell 44:928–41 113. Quivy J-P, Gerard A, Cook AJ, Roche D, Almouzni G. 2008. The HP1-p150/CAF-1 interaction is required for pericentric heterochromatin replication and S-phase progression in mouse cells. Nat. Struct. Mol. Biol. 15:972–79 114. Winkler DD, Zhou H, Dar MA, Zhang Z, Luger K. 2012. Yeast CAF-1 assembles histone (H3–H4)2 tetramers prior to DNA deposition. Nucleic Acids Res. 40:10139–49 115. Kamakaka RT, Bulger M, Kaufman PD, Stillman B, Kadonaga JT. 1996. Postreplicative chromatin assembly by Drosophila and human chromatin assembly factor 1. Mol. Cell. Biol. 16:810–17 116. Tyler JK, Bulger M, Kamakaka RT, Kobayashi R, Kadonaga JT. 1996. The p55 subunit of Drosophila chromatin assembly factor 1 is homologous to a histone deacetylase–associated protein. Mol. Cell. Biol. 16:6149–59 117. Tyler JK, Collins KA, Prasad-Sinha J, Amiott E, Bulger M, et al. 2001. Interaction between the Drosophila CAF-1 and ASF1 chromatin assembly factors. Mol. Cell. Biol. 21:6574–84 118. Kaya H, Shibahara KI, Taoka KI, Iwabuchi M, Stillman B, Araki T. 2001. FASCIATA genes for chromatin assembly factor 1 in Arabidopsis maintain the cellular organization of apical meristems. Cell 104:131– 42 119. Enomoto S, Berman J. 1998. Chromatin assembly factor I contributes to the maintenance, but not the re-establishment, of silencing at the yeast silent mating loci. Genes Dev. 12:219–32 120. Kaufman PD, Kobayashi R, Stillman B. 1997. Ultraviolet radiation sensitivity and reduction of telomeric silencing in Saccharomyces cerevisiae cells lacking chromatin assembly factor I. Genes Dev. 11:345–57 121. Prochasson P, Florens L, Swanson SK, Washburn MP, Workman JL. 2005. The HIR corepressor complex binds to nucleosomes generating a distinct protein/DNA complex resistant to remodeling by SWI/SNF. Genes Dev. 19:2534–39 122. Sherwood PW, Tsang SV, Osley MA. 1993. Characterization of HIR1 and HIR2, two genes required for regulation of histone gene transcription in Saccharomyces cerevisiae. Mol. Cell. Biol. 13:28–38 123. Sutton A, Bucaria J, Osley MA, Sternglanz R. 2001. Yeast ASF1 protein is required for cell cycle regulation of histone gene transcription. Genetics 158:587–96 124. Xu H, Kim UJ, Schuster T, Grunstein M. 1992. Identification of a new set of cell cycle–regulatory genes that regulate S-phase transcription of histone genes in Saccharomyces cerevisiae. Mol. Cell. Biol. 12:5249–59 www.annualreviews.org • Histone Chaperones

513

ARI

30 April 2014

11:12

125. Banumathy G, Somaiah N, Zhang R, Tang Y, Hoffmann J, et al. 2009. Human UBN1 is an ortholog of yeast Hpc2p and has an essential role in the HIRA/ASF1a chromatin-remodeling pathway in senescent cells. Mol. Cell. Biol. 29:758–70 126. Balaji S, Iyer LM, Aravind L. 2009. HPC2 and ubinuclein define a novel family of histone chaperones conserved throughout eukaryotes. Mol. Biosyst. 5:269–75 127. Rai TS, Puri A, McBryan T, Hoffman J, Tang Y, et al. 2011. Human CABIN1 is a functional member of the human HIRA/UBN1/ASF1a histone H3.3 chaperone complex. Mol. Cell. Biol. 31:4107–18 128. Park PJ. 2009. ChIP-seq: advantages and challenges of a maturing technology. Nat. Rev. Genet. 10:669–80 129. Goldberg AD, Banaszynski LA, Noh K-M, Lewis PW, Els¨asser SJ, et al. 2010. Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140:678–91 130. Pchelintsev NA, McBryan T, Rai TS, van Tuyn J, Ray Gallet D, et al. 2013. Placing the HIRA histone chaperone complex in the chromatin landscape. Cell Rep. 3:1012–19 131. Dran´e P, Ouararhni K, Depaux A, Shuaib M, Hamiche A. 2010. The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev. 24:1253–65 132. Michod D, Bartesaghi S, Khelifi A, Bellodi C, Berliocchi L, et al. 2012. Calcium-dependent dephosphorylation of the histone chaperone DAXX regulates H3.3 loading and transcription upon neuronal activation. Neuron 74:122–35 133. Xue Y, Gibbons R, Yan Z, Yang D, McDowell TL, et al. 2003. The ATRX syndrome protein forms a chromatin-remodeling complex with Daxx and localizes in promyelocytic leukemia nuclear bodies. Proc. Natl. Acad. Sci. USA 100:10635–40 134. Clarke L, Carbon J. 1980. Isolation of a yeast centromere and construction of functional small circular chromosomes. Nature 287:504–9 135. Black BE, Bassett EA. 2008. The histone variant CENP-A and centromere specification. Curr. Opin. Cell Biol. 20:91–100 136. Kato T, Sato N, Hayama S, Yamabuki T, Ito T, et al. 2007. Activation of Holliday junction recognizing protein involved in the chromosomal stability and immortality of cancer cells. Cancer Res. 67:8544–53 137. Dunleavy EM, Almouzni G, Karpen GH. 2011. H3.3 is deposited at centromeres in S phase as a placeholder for newly assembled CENP-A in G1 phase. Nucleus 2:146–57 138. Zasadzinska E, Barnhart-Dailey MC, Kuich PHJL, Foltz DR. 2013. Dimerization of the CENP-A ´ assembly factor HJURP is required for centromeric nucleosome deposition. EMBO J. 32:2113–24 139. Bernad R, S´anchez P, Rivera T, Rodr´ıguez-Corsino M, Boyarchuk E, et al. 2011. Xenopus HJURP and condensin II are required for CENP-A assembly. J. Cell Biol. 192:569–82 140. Stoler S, Rogers K, Weitze S, Morey L, Fitzgerald-Hayes M, Baker RE. 2007. Scm3, an essential Saccharomyces cerevisiae centromere protein required for G2 /M progression and Cse4 localization. Proc. Natl. Acad. Sci. USA 104:10571–76 141. Erhardt S, Mellone BG, Betts CM, Zhang W, Karpen GH, Straight AF. 2008. Genome-wide analysis reveals a cell cycle–dependent mechanism controlling centromere propagation. J. Cell Biol. 183:805–18 142. Kaufman PD, Cohen JL, Osley MA. 1998. Hir proteins are required for position-dependent gene silencing in Saccharomyces cerevisiae in the absence of chromatin assembly factor I. Mol. Cell. Biol. 18:4793–806 143. Sharp JA, Franco AA, Osley MA, Kaufman PD. 2002. Chromatin assembly factor I and Hir proteins contribute to building functional kinetochores in S. cerevisiae. Genes Dev. 16:85–100 144. Haber JE, Braberg H, Wu Q, Alexander R, Haase J, et al. 2013. Systematic triple-mutant analysis uncovers functional connectivity between pathways involved in chromosome regulation. Cell Rep. 3:2168–78 145. Murzina N, Verreault A, Laue E, Stillman B. 1999. Heterochromatin dynamics in mouse cells: interaction between chromatin assembly factor 1 and HP1 proteins. Mol. Cell 4:529–40 146. Loyola A, Tagami H, Bonaldi T, Roche D, Quivy J-P, et al. 2009. The HP1α–CAF1–SetDB1–containing complex provides H3K9me1 for Suv39-mediated K9me3 in pericentric heterochromatin. EMBO Rep. 10:769–75 147. Wong LH, Ren H, Williams E, McGhie J, Ahn S, et al. 2009. Histone H3.3 incorporation provides a unique and functionally essential telomeric chromatin in embryonic stem cells. Genome Res. 19:404–14 148. Ask K, Jasencakova Z, Menard P, Feng Y, Almouzni G, Groth A. 2012. Codanin-1, mutated in the anaemic disease CDAI, regulates Asf1 function in S-phase histone supply. EMBO J. 31:2013–23

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

514

Gurard-Levin

·

Quivy

·

Almouzni

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

149. Kaufman PD, Almouzni G. 2000. DNA replication, nucleotide excision repair, and nucleosome assembly. In Chromatin Structure and Gene Expression, ed. SCR Elgin, JL Workman, pp. 24–48. Oxford, UK: Oxford Univ. Press 150. Alabert C, Groth A. 2012. Chromatin replication and epigenome maintenance. Nat. Rev. Mol. Cell Biol. 13:153–67 151. Prior CP, Cantor CR, Johnson EM, Allfrey VG. 1980. Incorporation of exogenous pyrene-labeled histone into Physarum chromatin: a system for studying changes in nucleosomes assembled in vivo. Cell 20:597–608 152. Jackson DA, Cook PR. 1987. The nucleoskeleton: active site of transcription and replication. Haematol. Blood Transfus. 31:299–303 153. Jackson V. 1988. Deposition of newly synthesized histones: Hybrid nucleosomes are not tandemly arranged on daughter DNA strands. Biochemistry 27:2109–20 154. Annunziato AT. 1989. Inhibitors of topoisomerases I and II arrest DNA replication, but do not prevent nucleosome assembly in vivo. J. Cell Sci. 93(Part 4):593–603 155. Yamasu K, Senshu T. 1990. Conservative segregation of tetrameric units of H3 and H4 histones during nucleosome replication. J. Biochem. 107:15–20 156. Kass SU, Wolffe AP. 1998. DNA methylation, nucleosomes and the inheritance of chromatin structure and function. Novartis Found. Symp. 214:22–50 157. McKittrick E, Gafken PR, Ahmad K, Henikoff S. 2004. Histone H3.3 is enriched in covalent modifications associated with active chromatin. Proc. Natl. Acad. Sci. USA 101:1525–30 158. Xu M, Long C, Chen X, Huang C, Chen S, Zhu B. 2010. Partitioning of histone H3–H4 tetramers during DNA replication–dependent chromatin assembly. Science 328:94–98 159. Huang C, Zhang Z, Xu M, Li Y, Li Z, et al. 2013. H3.3–H4 tetramer splitting events feature cell-type specific enhancers. PLoS Genet. 9:e1003558 160. Nakatani Y, Ray Gallet D, Quivy J-P, Tagami H, Almouzni G. 2004. Two distinct nucleosome assembly pathways: dependent or independent of DNA synthesis promoted by histone H3.1 and H3.3 complexes. Cold Spring Harb. Symp. Quant. Biol. 69:273–80 161. Formosa T. 2012. The role of FACT in making and breaking nucleosomes. Biochim. Biophys. Acta 1819:247–55 162. Winkler DD, Muthurajan UM, Hieb AR, Luger K. 2011. Histone chaperone FACT coordinates nucleosome interaction through multiple synergistic binding events. J. Biol. Chem. 286:41883–92 163. Gambus A, Jones RC, Sanchez-Diaz A, Kanemaki M, van Deursen F, et al. 2006. GINS maintains association of Cdc45 with MCM in replisome progression complexes at eukaryotic DNA replication forks. Nat. Cell Biol. 8:358–66 164. Tan BC, Chien CT, Hirose S, Lee SC. 2006. Functional cooperation between FACT and MCM helicase facilitates initiation of chromatin DNA replication. EMBO J. 25:3975–85 165. VanDemark AP, Blanksma M, Ferris E, Heroux A, Hill CP, Formosa T. 2006. The structure of the yFACT Pob3-M domain, its interaction with the DNA replication factor RPA, and a potential role in nucleosome deposition. Mol. Cell 22:363–74 166. Wittmeyer J, Formosa T. 1997. The Saccharomyces cerevisiae DNA polymerase α catalytic subunit interacts with Cdc68/Spt16 and with Pob3, a protein similar to an HMG1-like protein. Mol. Cell. Biol. 17:4178–90 167. Abe T, Sugimura K, Hosono Y, Takami Y, Akita M, et al. 2011. The histone chaperone facilitates chromatin transcription (FACT) protein maintains normal replication fork rates. J. Biol. Chem. 286:30504–12 168. Soria G, Polo SE, Almouzni G. 2012. Prime, repair, restore: the active role of chromatin in the DNA damage response. Mol. Cell 46:722–34 169. Adam S, Polo SE. 2012. Chromatin dynamics during nucleotide excision repair: histones on the move. Int. J. Mol. Sci. 13:11895–911 170. Smerdon MJ. 1991. DNA repair and the role of chromatin structure. Curr. Opin. Cell Biol. 3:422–28 171. Baldeyron C, Soria G, Roche D, Cook AJ, Almouzni G. 2011. HP1α recruitment to DNA damage by p150CAF-1 promotes homologous recombination repair. J. Cell Biol. 193:81–95 172. Soria G, Almouzni G. 2013. Differential contribution of HP1 proteins to DNA end resection and homology-directed repair. Cell Cycle 12:422–29 www.annualreviews.org • Histone Chaperones

515

ARI

30 April 2014

11:12

173. Rogakou EP, Boon C, Redon C, Bonner WM. 1999. Megabase chromatin domains involved in DNA double-strand breaks in vivo. J. Cell Biol. 146:905–16 174. Fillingham J, Keogh MC, Krogan NJ. 2006. γH2AX and its role in DNA double-strand break repair. Biochem. Cell Biol. 84:568–77 175. Adam S, Polo SE, Almouzni G. 2013. Transcription recovery after DNA damage requires chromatin priming by the H3.3 histone chaperone HIRA. Cell 155:94–106 176. Dinant C, Ampatziadis-Michailidis G, Lans H, Tresini M, Lagarou A, et al. 2013. Enhanced chromatin dynamics by FACT promotes transcriptional restart after UV-induced DNA damage. Mol. Cell 51:469– 79 177. Boeger H, Griesenbeck J, Kornberg RD. 2008. Nucleosome retention and the stochastic nature of promoter chromatin remodeling for transcription. Cell 133:716–26 178. Boeger H, Griesenbeck J, Strattan JS, Kornberg RD. 2004. Removal of promoter nucleosomes by disassembly rather than sliding in vivo. Mol. Cell 14:667–73 179. Kornberg RD, Lorch Y. 2002. Chromatin and transcription: Where do we go from here? Curr. Opin. Genet. Dev. 12:249–51 180. Kulaeva OI, Gaykalova DA, Studitsky VM. 2007. Transcription through chromatin by RNA polymerase II: histone displacement and exchange. Mutat. Res. 618:116–29 181. Smolle M, Venkatesh S, Gogol MM, Li H, Zhang Y, et al. 2012. Chromatin remodelers Isw1 and Chd1 maintain chromatin structure during transcription by preventing histone exchange. Nat. Struct. Mol. Biol. 19:884–92 182. Venkatesh S, Smolle M, Li H, Gogol MM, Saint M, et al. 2012. Set2 methylation of histone H3 lysine 36 suppresses histone exchange on transcribed genes. Nature 489:452–55 183. Owen-Hughes T, Workman JL. 1996. Remodeling the chromatin structure of a nucleosome array by transcription factor–targeted trans-displacement of histones. EMBO J. 15:4702–12 184. Chen H, Li B, Workman JL. 1994. A histone-binding protein, nucleoplasmin, stimulates transcription factor binding to nucleosomes and factor-induced nucleosome disassembly. EMBO J. 13:380–90 185. Walter PP, Owen-Hughes TA, Cot´ ˆ e J, Workman JL. 1995. Stimulation of transcription factor binding and histone displacement by nucleosome assembly protein 1 and nucleoplasmin requires disruption of the histone octamer. Mol. Cell. Biol. 15:6178–87 186. Schwabish MA, Struhl K. 2006. Asf1 mediates histone eviction and deposition during elongation by RNA polymerase II. Mol. Cell 22:415–22 187. Kaplan CD, Laprade L, Winston F. 2003. Transcription elongation factors repress transcription initiation from cryptic sites. Science 301:1096–99 188. Endoh M, Zhu W, Hasegawa J, Watanabe H, Kim DK, et al. 2004. Human Spt6 stimulates transcription elongation by RNA polymerase II in vitro. Mol. Cell. Biol. 24:3324–36 189. Chimura T, Kuzuhara T, Horikoshi M. 2002. Identification and characterization of CIA/ASF1 as an interactor of bromodomains associated with TFIID. Proc. Natl. Acad. Sci. USA 99:9334–39 190. Simic R, Lindstrom DL, Tran HG, Roinick KL, Costa PJ, et al. 2003. Chromatin remodeling protein Chd1 interacts with transcription elongation factors and localizes to transcribed genes. EMBO J. 22:1846– 56 191. Houlard M, Berlivet S, Probst AV, Quivy J-P, H´ery P, et al. 2006. CAF-1 is essential for heterochromatin organization in pluripotent embryonic cells. PLoS Genet. 2:e181 192. Roberts C, Sutherland HF, Farmer H, Kimber W, Halford S, et al. 2002. Targeted mutagenesis of the Hira gene results in gastrulation defects and patterning abnormalities of mesoendodermal derivatives prior to early embryonic lethality. Mol. Cell. Biol. 22:2318–28 193. Garrick D, Sharpe JA, Arkell R, Dobbie L, Smith AJ, et al. 2006. Loss of Atrx affects trophoblast development and the pattern of X-inactivation in extraembryonic tissues. PLoS Genet. 2:e58 194. Michaelson JS, Bader D, Kuo F, Kozak C, Leder P. 1999. Loss of Daxx, a promiscuously interacting protein, results in extensive apoptosis in early mouse development. Genes Dev. 13:1918–23 195. Hartford SA, Luo Y, Southard TL, Min IM, Lis JT, Schimenti JC. 2011. Minichromosome maintenance helicase paralog MCM9 is dispensible for DNA replication but functions in germ-line stem cells and tumor suppression. Proc. Natl. Acad. Sci. USA 108:17702–7

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

516

Gurard-Levin

·

Quivy

·

Almouzni

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

BI83CH20-Almouzni

ARI

30 April 2014

11:12

196. Loppin B, Bonnefoy E, Anselme C, Laurenc¸on A, Karr TL, Couble P. 2005. The histone H3.3 chaperone HIRA is essential for chromatin assembly in the male pronucleus. Nature 437:1386–90 197. Bonnefoy E, Orsi GA, Couble P, Loppin B. 2007. The essential role of Drosophila HIRA for de novo assembly of paternal chromatin at fertilization. PLoS Genet. 3:1991–2006 198. Szenker E, Lacoste N, Almouzni G. 2012. A developmental requirement for HIRA-dependent H3.3 deposition revealed at gastrulation in Xenopus. Cell Rep. 1:730–40 199. Jullien J, Astrand C, Szenker E, Garrett N, Almouzni G, Gurdon JB. 2012. HIRA dependent H3.3 deposition is required for transcriptional reprogramming following nuclear transfer to Xenopus oocytes. Epigenet. Chromatin 5:17 200. Lewis PW, Muller MM, Koletsky MS, Cordero F, Lin S, et al. 2013. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340:857–61 201. Almouzni G, Corpet A. 2011. ASF1b as a prognosis marker and therapeutic target in human cancer. US patent no. 20130149320 202. Robinson PJ, Bushnell DA, Trnka MJ, Burlingame AL, Kornberg RD. 2012. Structure of the mediator head module bound to the carboxy-terminal domain of RNA polymerase II. Proc. Natl. Acad. Sci. USA 109:17931–35 203. Sobel RE, Cook RG, Perry CA, Annunziato AT, Allis CD. 1995. Conservation of deposition-related acetylation sites in newly synthesized histones H3 and H4. Proc. Natl. Acad. Sci. USA 92:1237–41 204. Huang S, Zhou H, Katzmann D, Hochstrasser M, Atanasova E, Zhang Z. 2005. Rtt106p is a histone chaperone involved in heterochromatin-mediated silencing. Proc. Natl. Acad. Sci. USA 38:13410–15 205. Bortvin A, Winston F. 1996. Evidence that Spt6p controls chromatin structure by a direct interaction with histones. Science 272:1473–76 206. Luk E, Vu N, Patteson K, Mizuguchi G, Wu W, et al. 2007. Chz1, a nuclear chaperone for histone H2AZ. Mol. Cell 5:357–68 207. Harata M, Oma Y, Mizuno S, Jiang YW, Sillman DJ, et al. 1999. The nuclear actin–related protein of Saccharomyces cerevisiae, Act3p/Arp4, interacts with core histones. Mol. Biol. Cell 10:2595–605 208. Shen X, Ranallo R, Choi E, Wu C. 2003. Involvement of actin-related proteins in ATP-dependent chromatin remodeling. Mol. Cell 12:147–55 209. Okuwaki M, Matsumoto K, Tsujimoto M, Nagata K. 2001. Function of nucleophosmin/B23, a nuclear acidic protein, as a histone chaperone. FEBS Lett. 506:272–76 210. Rouguelle C, Avner P. 1996. Cloning and characterization of a murine brain specific gene Bpx and its human homologue lying within the Xic region. Hum. Mol. Genet. 5:41–49 211. Okuwaki M, Nagata K. 1998. Template activating factor 1 remodels the chromatin structure and stimulates transcription from the chromatin template. J. Biol. Chem. 273:34511–18 212. Wang GS, Hong CJ, Yen TY, Huang HY, Ou Y, et al. 2004. Transcriptional modification by a CASKinteracting nucleosome assembly protein. Neuron 42:113–28 213. Selth L, Svejstrup JQ. 2007. Vps75, a new yeast member of the NAP histone chaperone family. J. Biol. Chem. 282:12358–62 214. Loyola A, LeRoy G, Wang YH, Reinberg D. 2001. Reconstitution of recombinant chromatin establishes a requirement for histone-tail modifications during chromatin assembly and transcription. Genes Dev. 15:2837–51 215. Peterson CL, Zhao Y, Chait BT. 1998. Subunits of the yeast SWI/SNF complex are members of the actin-related protein (ARP) family. J. Biol. Chem. 273:23641–44 216. Ito T, Bulger M, Pazin MJ, Kobayashi R, Kadonaga JT. 1997. ACF, an ISWI-containing and ATPutilizing chromatin assembly and remodeling factor. Cell 90:145–55 217. Sawatsubashi S, Murata T, Lim J, Fujiki R, Ito S, et al. 2010. A histone chaperone, DEK, transcriptionally coactivates a nuclear receptor. Genes Dev. 24:159–70 218. Kashiwaba S, Kitahashi K, Watanabe T, Onoda F, Ohtsu M, Murakami Y. 2010. The mammalian INO80 complex is recruited to DNA damage sites in an ARP8 dependent manner. Biochem. Biophys. Res. Commun. 402:619–25

www.annualreviews.org • Histone Chaperones

517

BI83-Front-Matter

ARI

30 April 2014

10:15

Contents

Annual Review of Biochemistry Volume 83, 2014

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Journeys in Science: Glycobiology and Other Paths Raymond A. Dwek p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1 Lipids and Extracellular Materials William Dowhan p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p45 Topological Regulation of Lipid Balance in Cells Guillaume Drin p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p51 Lipidomics: Analysis of the Lipid Composition of Cells and Subcellular Organelles by Electrospray Ionization Mass Spectrometry Britta Brugger ¨ p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p79 Biosynthesis and Export of Bacterial Lipopolysaccharides Chris Whitfield and M. Stephen Trent p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p99 Demystifying Heparan Sulfate–Protein Interactions Ding Xu and Jeffrey D. Esko p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 129 Dynamics and Timekeeping in Biological Systems Christopher M. Dobson p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 159 Metabolic and Nontranscriptional Circadian Clocks: Eukaryotes Akhilesh B. Reddy and Guillaume Rey p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 165 Interactive Features of Proteins Composing Eukaryotic Circadian Clocks Brian R. Crane and Michael W. Young p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 191 Metabolic Compensation and Circadian Resilience in Prokaryotic Cyanobacteria Carl Hirschie Johnson and Martin Egli p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 221 Activity-Based Profiling of Proteases Laura E. Sanman and Matthew Bogyo p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 249 Asymmetry of Single Cells and Where That Leads Mark S. Bretscher p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 275 Bringing Dynamic Molecular Machines into Focus by Methyl-TROSY NMR Rina Rosenzweig and Lewis E. Kay p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 291

v

BI83-Front-Matter

ARI

30 April 2014

10:15

Chlorophyll Modifications and Their Spectral Extension in Oxygenic Photosynthesis Min Chen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 317 Enzyme Inhibitor Discovery by Activity-Based Protein Profiling Micah J. Niphakis and Benjamin F. Cravatt p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 341 Expanding and Reprogramming the Genetic Code of Cells and Animals Jason W. Chin p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 379 Genome Engineering with Targetable Nucleases Dana Carroll p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 409 Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Hierarchy of RNA Functional Dynamics Anthony M. Mustoe, Charles L. Brooks, and Hashim M. Al-Hashimi p p p p p p p p p p p p p p p p p p 441 High-Resolution Structure of the Eukaryotic 80S Ribosome Gulnara Yusupova and Marat Yusupov p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 467 Histone Chaperones: Assisting Histone Traffic and Nucleosome Dynamics Zachary A. Gurard-Levin, Jean-Pierre Quivy, and Genevi`eve Almouzni p p p p p p p p p p p p p p 487 Human RecQ Helicases in DNA Repair, Recombination, and Replication Deborah L. Croteau, Venkateswarlu Popuri, Patricia L. Opresko, and Vilhelm A. Bohr p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 519 Intrinsically Disordered Proteins and Intrinsically Disordered Protein Regions Christopher J. Oldfield and A. Keith Dunker p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 553 Mechanism and Function of Oxidative Reversal of DNA and RNA Methylation Li Shen, Chun-Xiao Song, Chuan He, and Yi Zhang p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 585 Progress Toward Synthetic Cells J. Craig Blain and Jack W. Szostak p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 615 PTEN Carolyn A. Worby and Jack E. Dixon p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 641 Regulating the Chromatin Landscape: Structural and Mechanistic Perspectives Blaine Bartholomew p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 671 RNA Helicase Proteins as Chaperones and Remodelers Inga Jarmoskaite and Rick Russell p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 697

vi

Contents

BI83-Front-Matter

ARI

30 April 2014

10:15

Selection-Based Discovery of Druglike Macrocyclic Peptides Toby Passioura, Takayuki Katoh, Yuki Goto, and Hiroaki Suga p p p p p p p p p p p p p p p p p p p p p p p p p 727 Small Proteins Can No Longer Be Ignored Gisela Storz, Yuri I. Wolf, and Kumaran S. Ramamurthi p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 753 The Scanning Mechanism of Eukaryotic Translation Initiation Alan G. Hinnebusch p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 779 Understanding Nucleic Acid–Ion Interactions Jan Lipfert, Sebastian Doniach, Rhiju Das, and Daniel Herschlag p p p p p p p p p p p p p p p p p p p p p p 813

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Indexes Cumulative Index of Contributing Authors, Volumes 79–83 p p p p p p p p p p p p p p p p p p p p p p p p p p p 843 Cumulative Index of Article Titles, Volumes 79–83 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 847 Errata An online log of corrections to Annual Review of Biochemistry articles may be found at http://www.annualreviews.org/errata/biochem

Contents

vii

Annual Reviews It’s about time. Your time. It’s time well spent.

New From Annual Reviews:

Annual Review of Statistics and Its Application Volume 1 • Online January 2014 • http://statistics.annualreviews.org

Editor: Stephen E. Fienberg, Carnegie Mellon University

Annu. Rev. Biochem. 2014.83:487-517. Downloaded from www.annualreviews.org by Seattle University on 06/08/14. For personal use only.

Associate Editors: Nancy Reid, University of Toronto Stephen M. Stigler, University of Chicago The Annual Review of Statistics and Its Application aims to inform statisticians and quantitative methodologists, as well as all scientists and users of statistics about major methodological advances and the computational tools that allow for their implementation. It will include developments in the field of statistics, including theoretical statistical underpinnings of new methodology, as well as developments in specific application domains such as biostatistics and bioinformatics, economics, machine learning, psychology, sociology, and aspects of the physical sciences.

Complimentary online access to the first volume will be available until January 2015. table of contents:

• What Is Statistics? Stephen E. Fienberg • A Systematic Statistical Approach to Evaluating Evidence from Observational Studies, David Madigan, Paul E. Stang, Jesse A. Berlin, Martijn Schuemie, J. Marc Overhage, Marc A. Suchard, Bill Dumouchel, Abraham G. Hartzema, Patrick B. Ryan

• High-Dimensional Statistics with a View Toward Applications in Biology, Peter Bühlmann, Markus Kalisch, Lukas Meier • Next-Generation Statistical Genetics: Modeling, Penalization, and Optimization in High-Dimensional Data, Kenneth Lange, Jeanette C. Papp, Janet S. Sinsheimer, Eric M. Sobel

• The Role of Statistics in the Discovery of a Higgs Boson, David A. van Dyk

• Breaking Bad: Two Decades of Life-Course Data Analysis in Criminology, Developmental Psychology, and Beyond, Elena A. Erosheva, Ross L. Matsueda, Donatello Telesca

• Brain Imaging Analysis, F. DuBois Bowman

• Event History Analysis, Niels Keiding

• Statistics and Climate, Peter Guttorp

• Statistical Evaluation of Forensic DNA Profile Evidence, Christopher D. Steele, David J. Balding

• Climate Simulators and Climate Projections, Jonathan Rougier, Michael Goldstein • Probabilistic Forecasting, Tilmann Gneiting, Matthias Katzfuss • Bayesian Computational Tools, Christian P. Robert • Bayesian Computation Via Markov Chain Monte Carlo, Radu V. Craiu, Jeffrey S. Rosenthal • Build, Compute, Critique, Repeat: Data Analysis with Latent Variable Models, David M. Blei • Structured Regularizers for High-Dimensional Problems: Statistical and Computational Issues, Martin J. Wainwright

• Using League Table Rankings in Public Policy Formation: Statistical Issues, Harvey Goldstein • Statistical Ecology, Ruth King • Estimating the Number of Species in Microbial Diversity Studies, John Bunge, Amy Willis, Fiona Walsh • Dynamic Treatment Regimes, Bibhas Chakraborty, Susan A. Murphy • Statistics and Related Topics in Single-Molecule Biophysics, Hong Qian, S.C. Kou • Statistics and Quantitative Risk Management for Banking and Insurance, Paul Embrechts, Marius Hofert

Access this and all other Annual Reviews journals via your institution at www.annualreviews.org.

Annual Reviews | Connect With Our Experts Tel: 800.523.8635 (us/can) | Tel: 650.493.4400 | Fax: 650.424.0910 | Email: [email protected]

Histone chaperones: assisting histone traffic and nucleosome dynamics.

The functional organization of eukaryotic DNA into chromatin uses histones as components of its building block, the nucleosome. Histone chaperones, wh...
2MB Sizes 1 Downloads 3 Views