REPRODUCTIONREVIEW

The histone codes for

Lina Wang1,2,*, Zhiliang Xu1,3,*, Muhammad Babar Khawar4, Chao Liu1 and Wei Li1 1State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China, 2University of Chinese Academy of Sciences, Beijing, People’s Republic of China, 3Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China and 4Department of Zoology, University of the Punjab, Lahore, Pakistan Correspondence should be addressed to W Li or C Liu; Email: [email protected] or [email protected] *(L Wang and Z Xu contributed equally to this work)

Abstract

Meiosis is a specialized process that produces haploid gametes from diploid cells by a single round of DNA replication followed by two successive cell divisions. It contains many special events, such as programmed DNA double-strand break (DSB) formation, , crossover formation and resolution. These events are associated with dynamically regulated chromosomal structures, the dynamic transcriptional regulation and remodeling are mainly modulated by histone modifications, termed ‘histone codes’. The purpose of this review is to summarize the histone codes that are required for meiosis during spermatogenesis and , involving meiosis resumption, meiotic asymmetric division and other cellular processes. We not only systematically review the functional roles of histone codes in meiosis but also discuss future trends and perspectives in this field. Reproduction (2017) 154 R65–R79

Introduction becoming shorter and thicker. During the diplotene stage, the chromosomes start to be decondensed, and the Sexual reproduction is an essential way for higher tetrad structures can be observed clearly. Chromosomes eukaryotes to produce offspring, which aims to keep continue to condense during the diakinesis stage, the genome stable and variable. It is achieved by the nucleolus disappears and chromosomes are evenly production of haploid gametes and the fusion of these distributed in the nucleus (Keeney et al. 1997, Zickler gametes. The gametes contain half of the chromosomal & Kleckner 1999, Borner et al. 2004, Strom et al. 2007, number in somatic cell because they are produced by Lange et al. 2016). a specialized cell division with a single round of DNA Different from most of the other meiotic processes, replication, followed by two successive divisions, meiotic process of the female mammals undergoes known as meiosis. Meiosis occurs in almost all sexually two of phases arrest during oocytes maturation. First, reproducing creatures and is highly conserved from the developing oocytes arrest at diplotene stage with a single-cell eukaryotes, such as yeast, to multicellular germinal vesicle (GV) in their nucleus, and this state is eukaryotes, such as insects, fish, birds and mammals maintained until meiosis resumption at puberty or even (Clancy 1998, Yamamoto et al. 1999, Hawley 2002, later. Meiosis resumption is marked by GV breakdown Smith et al. 2008). In contrast to mitosis, meiosis starts (GVBD), which indicates the completion of I from a long premeiotic DNA replication process, and the transition to meiosis I (MI). After that, the oocytes followed by prophase I. According to the chromosomal will be arrested for the second time at metaphase II (MII), configurations, meiosis prophase I can be divided into which will be activated by the fertilization with sperms leptotene, zygotene, pachytene, diplotene and diakinesis (Downs 1996). stages. At the leptotene stage, when the DNA replication Chromosomes are constituted by nucleosomes, has been completed, chromatin starts to fold and condense which are the fundamental unit of chromatin and are to form the fibrous chromosomes. When entering into the composed of DNA and various histones. Two copies zygotene stage, the chromosomes approximately line up of the four histones (H2A, H2B, H3 and H4) constitute with their homologous chromosomes, which are equal the core particle of the nucleosome by forming an in length and position. Following that is the pachytene octamer, and H1 is a linker histone for packing the stage, in which chromosomes condense further, nucleosome into higher-order structures. Histone tails

© 2017 Society for Reproduction and Fertility DOI: 10.1530/REP-17-0153 ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access

10.1530/REP-17-0153 R66 L Wang, Z Xu and others

can be substituted by their variants or modified by Table 1 Histone modifications and variants in the meiosis. various modifiers, such as acetylation, ubiquitination, Modifications/variants Function Species/cells types Reference methylation and phosphorylation, to regulate the Acetylation chromatin status (Thompson et al. 2013, Watanabe et al. H3K9ac, H3K18ac, H3K23ac Meiotic cell division Mouse/primary spermatocytes Song et al. (2011) 2013, Hiragami-Hamada et al. 2016, Kadoch et al. H3K9ac Gene transcription Primary spermatocytes Li et al. (2017) 2016), which plays essential roles in DNA replication H3K9ac Meiotic recombination Yeast Yamada et al. (2013) (Franke et al. 2016), gene transcription (Yang et al. 2016), hotspots homologous recombination (Yoon et al. 2016), DSB H3K9ac, H3K14ac, H4K5ac, Open chromatin for the Mouse/oocytes Akiyama et al. (2004), Sarmento et al. repair (Lange et al. 2016) and chromatin stability. These H4K8ac, H4K12ac, gene expression in GV (2004), Kageyama et al. (2007), H4K16ac oocytes Nagashima et al. (2007), Gu et al. (2010) histone variants and post-translational modifications H4K5ac, H4K8ac, H4K12ac Transcription in leptotene Mouse/spermatocytes Shirakata et al. (2014) (PTMs) constitute the so-called ‘histone codes’ (Fig.1; and zygotene Tang et al. 2015, Wolfe et al. 2008). More than 50 H4K8ac Chromatin remodeling in Mouse/oocytes Kim et al. (2003) histone PTMs have been identified, and this number meiosis of oocytes is growing rapidly (Grunstein 1997, Jenuwein & Allis H4K16ac Meiotic recombination Yeast Cavero et al. (2016) checkpoint 2001, Eberharter & Becker 2002, Iizuka & Smith 2003, H4K44ac Facilitates homologous Yeast Hu et al. (2015) Zentner & Henikoff 2013). Histone codes have been recombination found to participate and play important roles in meiosis Methylation (Fig. 2, Table 1 and Box 1, 2, 3 and 4) (Maleki & Keeney H3K4me1/me2/me3 Chromatin reorganization Mouse/spermatocytes Godmann et al. (2007) 2004, Hu et al. 2015, Luense et al. 2016, Xu et al. H3K4me3 Programmed DSB hotspots Yeast/mouse spermatocyte/oocytes Borde et al. (2009), Buard et al. (2009), and meiotic Brick et al. (2012), Acquaviva et al. 2016), and the functions of these histone codes during recombination initiation (2013), Sommermeyer et al. (2013) meiosis will be summarized in the following sections of H3K9me1/me2 Synapsis and genes Mouse/spermatocyte/oocytes Tachibana et al. (2007) this review. transcription H3K9me3 Synapsis and pericentric Mouse/spermatocyte Peters et al. (2001), Kato et al. (2015) The histone codes of H1in meiosis heterochromatin modification Histone H1 is the linker histone in nucleosomes, and H3K27me3 Transcriptional silencing Mouse/spermatocyte Song et al. (2011) it is important for higher-order chromatin formation H3K27me3 Transcription activation Mouse/oocytes Xu et al. (2016) Figure 1 The histone codes. The genetic information is stored in the and compaction (Izzo & Schneider 2016). The H3K36me3 Meiotic recombination Mouse/spermatocytes Powers et al. (2016) nucleus and is divided into different chromosomes. The basic units of H3K79me2 Transcriptional reactivation Mouse/spermatocytes Ontoso et al. (2014) the chromosomes are nucleosomes, which contain double-stranded histone H1 family is heterogeneous, and multiple H3K79me3 Transcription repression on Mouse/spermatocytes Ontoso et al. (2014) DNA and a histone octamer connected by a linker histone H1. H1 variants are expressed in different organisms. For centromeric regions and

Various histone modifications are located at histone –NH2 or –COOH example, in humans, the histone H1 family includes the sex body tails and some histone variants form the complex histone codes. 11 different members with 7 somatic subtypes, Ubiquitination H2Aub Transcriptional silencing Mouse/spermatocyte/oocytes Baarends et al. (2005), An et al. (2010), Hasegawa et al. (2015), Luo et al. (2015) H2BK123ub Meiosis initiation and DSB Yeast Robzyk et al. (2000), Yamashita et al. (2004) formation H2BK120ub Chromatin relaxation and Mouse/spermatocyte Xu et al. (2016) homologous recombination Phosphorylation H2AT119ph Disassembly of the Drosophila/oocyte Ivanovska et al. (2005) synaptonemal complex H2AXS139ph (γH2AX) Marker of programmed Mouse/spermatocyte/oocyte Turner et al. (2005), Blanco-Rodriguez DSB and promote DSB (2012) repair in meiosis H2AS121 ph Shugoshin localization and Yeast Watanabe et al. (2012) centromeric cohesion protection in meiosis I H3S10ph Chromosome condensation Worm/tetrahymena/yeast/mammals Wei et al. (1998, 1999), Song et al. (2011) and cell division Variants H1T, TH2B, H2AX, H2AZ, Meiotic divisions and Mouse/spermatocyte Govin and Khochbin (2013), H3.3 global histone removal Montellier et al. (2013) Figure 2 Various histone modifications H3T Spermatogonial differentia- Mouse/spermatocyte Ueda et al. (2017) involved in meiosis. The major histone tion and meiosis modifications include acetylation (Ac), initiation methylation (Me), ubiquitination (Ub) and phosphorylation (P), which play important roles in meiosis. The histone sequences are derived from mammals.

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R67

Table 1 Histone modifications and variants in the meiosis.

Modifications/variants Function Species/cells types Reference Acetylation H3K9ac, H3K18ac, H3K23ac Meiotic cell division Mouse/primary spermatocytes Song et al. (2011) H3K9ac Gene transcription Primary spermatocytes Li et al. (2017) H3K9ac Meiotic recombination Yeast Yamada et al. (2013) hotspots H3K9ac, H3K14ac, H4K5ac, Open chromatin for the Mouse/oocytes Akiyama et al. (2004), Sarmento et al. H4K8ac, H4K12ac, gene expression in GV (2004), Kageyama et al. (2007), H4K16ac oocytes Nagashima et al. (2007), Gu et al. (2010) H4K5ac, H4K8ac, H4K12ac Transcription in leptotene Mouse/spermatocytes Shirakata et al. (2014) and zygotene H4K8ac Chromatin remodeling in Mouse/oocytes Kim et al. (2003) meiosis of oocytes H4K16ac Meiotic recombination Yeast Cavero et al. (2016) checkpoint H4K44ac Facilitates homologous Yeast Hu et al. (2015) recombination Methylation H3K4me1/me2/me3 Chromatin reorganization Mouse/spermatocytes Godmann et al. (2007) H3K4me3 Programmed DSB hotspots Yeast/mouse spermatocyte/oocytes Borde et al. (2009), Buard et al. (2009), and meiotic Brick et al. (2012), Acquaviva et al. recombination initiation (2013), Sommermeyer et al. (2013) H3K9me1/me2 Synapsis and genes Mouse/spermatocyte/oocytes Tachibana et al. (2007) transcription H3K9me3 Synapsis and pericentric Mouse/spermatocyte Peters et al. (2001), Kato et al. (2015) heterochromatin modification H3K27me3 Transcriptional silencing Mouse/spermatocyte Song et al. (2011) H3K27me3 Transcription activation Mouse/oocytes Xu et al. (2016) H3K36me3 Meiotic recombination Mouse/spermatocytes Powers et al. (2016) H3K79me2 Transcriptional reactivation Mouse/spermatocytes Ontoso et al. (2014) H3K79me3 Transcription repression on Mouse/spermatocytes Ontoso et al. (2014) centromeric regions and the sex body Ubiquitination H2Aub Transcriptional silencing Mouse/spermatocyte/oocytes Baarends et al. (2005), An et al. (2010), Hasegawa et al. (2015), Luo et al. (2015) H2BK123ub Meiosis initiation and DSB Yeast Robzyk et al. (2000), Yamashita et al. (2004) formation H2BK120ub Chromatin relaxation and Mouse/spermatocyte Xu et al. (2016) homologous recombination Phosphorylation H2AT119ph Disassembly of the Drosophila/oocyte Ivanovska et al. (2005) synaptonemal complex H2AXS139ph (γH2AX) Marker of programmed Mouse/spermatocyte/oocyte Turner et al. (2005), Blanco-Rodriguez DSB and promote DSB (2012) repair in meiosis H2AS121 ph Shugoshin localization and Yeast Watanabe et al. (2012) centromeric cohesion protection in meiosis I H3S10ph Chromosome condensation Worm/tetrahymena/yeast/mammals Wei et al. (1998, 1999), Song et al. (2011) and cell division Variants H1T, TH2B, H2AX, H2AZ, Meiotic divisions and Mouse/spermatocyte Govin and Khochbin (2013), H3.3 global histone removal Montellier et al. (2013) H3T Spermatogonial differentia- Mouse/spermatocyte Ueda et al. (2017) tion and meiosis initiation

www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access R68 L Wang, Z Xu and others

Box 1: Acetylation Box 3: Methylation Histone acetylation is the first identified histone modification, Histone methylation is the most well-studied histone and it modulates gene transcription by fine-tuning chromatin modification, and it usually occurs on the lysine and arginine accessibility (Lee et al. 1993, Grunstein 1997, Kuo & Allis 1998, residues of histones H3 and H4 (Zhang & Reinberg 2001, Kurdistani et al. 2004). Histone acetyltransferases (HATs) and Kouzarides 2002, Bedford & Clarke 2009, Lan & Shi 2009). histone deacetylases (HDACs) are the main enzymes responsible Certain residues on histone H3 and H4 can be mono-, di- or for lysine acetylation and de-acetylation on histone N terminals tri-methylated; these modifications are catalyzed by histone respectively (Legube & Trouche 2003). It has been reported that methyltransferases (HMTs) (Santos-Rosa et al. 2002, Wang et al. the status of histone acetylation is very important to the meiotic 2003). HMTs utilize S-adenosylmethionine as their methyl group process during both spermatogenesis and oogenesis (Figs 3 and donor to modify arginine or lysine residues, thus regulating gene 4) (Hu et al. 2015, Getun et al. 2017). transcription, genome integrity and epigenetic inheritance (Bannister et al. 2002, Klose & Yi 2007). In mammals, arginine methylations of histones (H3R2, H3R8, H3R17, H3R26 and 3 testis-specific subtypes and 1 oocyte-specific H4R3) are catalyzed by protein arginine methyltransferases subtype (Izzo & Schneider 2016). Recently, a large (PRMTs). Methylations of lysine residues are catalyzed by other number of histone H1 subtype-specific modifications HMTs, including the SET-domain-containing protein family and the non-SET-domain proteins DOT1/DOT1L. These modifications have been identified by advanced mass spectrometric are considered markers for either activation or repression of the analysis, including phosphorylation, acetylation, related genes (Martin & Zhang 2005), among which, methylation, ubiquitination and ADP ribosylation methylations of H3K4, H3K36 and H3K79 are associated with (Wisniewski et al. 2007). These modifications of gene activation, whereas methylations of H3K9, H3K27 and H1 are involved in the regulation of all aspects H4K20 are responsible for gene repression (Martin & Zhang of linker histone functions. The functions of H1 2005, Klose & Yi 2007). In fact, histone methylation plays a vital phosphorylation have been extensively studied, and role in meiosis by modulating meiotic-related genes expression (Figs 3 and 4, see below). they include transcription, chromatin compaction, DNA damage and apoptosis, cell differentiation, aging and cancer (Wisniewski et al. 2007). H1 Although diverse H1 variants and modifications ubiquitination has also been reported to be essential have been investigated, their functional roles during for the initiation and amplification of ubiquitin meiosis have seldom been studied. In yeast, histone signaling after DNA damage. In this process, RNF8 is H1 is dispensable for proper meiotic recombination first recruited to DNA damage sites, and it assembles (Brush 2015), whereas in mice, H1t (Fig. 2), which a K63-linked ubiquitin chain on H1; then, RNF168 is specifically expressed in testis, is not essential for can recognize ubiquitinated H1 and catalyze the spermatogenesis because H1t knockout has no effect ubiquitination of H2A to recruit related DNA repair on the development and maturation of sperm. This proteins (Thorslund et al. 2015). phenomenon might be due to the redundancy of H1 variants, because they found that once H1t was knocked out, the mRNA levels of the other H1 variants were all Box 2: Ubiquitination increased (Drabent et al. 2000). The H1 ubiquitination Ubiquitination is a ubiquitous PTM that participates in many E3 ligase RNF8 is also not essential for meiosis, since biological events (Hershko & Ciechanover 1998, Beck-Sickinger Rnf8-deficient male mice showed no significant & Mörl 2006). Ubiquitination is achieved by a cascade of abnormalities during spermatocytes meiosis (Lu et al. enzymatic reactions that are sequentially catalyzed by ubiquitin- 2010). These findings indicate that the physiological activating enzyme (E1), ubiquitin-conjugating enzyme (E2) and function of this H1 PTM and H1 variants during meiosis ubiquitin ligase (E3) (Callis 2014). Conversely, deubiquitinating still need further investigation. enzymes (DUBs) can remove the ubiquitin molecules (or chains) from the modified substrates Komander ( et al. 2009, Isono & Nagel 2014). Targets or substrates can be modified by one Box 4: Phosphorylation ubiquitin molecule (mono-ubiquitination) or by an ubiquitin chain (poly-ubiquitination). Different linkages inside the Phosphorylation is one of the most significant post- ubiquitin chain define its topology, which determines the final transcriptional modifications, and it plays inimitable roles in destination of the modified proteins (substrates) Hershko( & various cellular processes. The functional roles of Ciechanover 1998). For example, K48-linked ubiquitin chains phosphorylation are mainly achieved by activating or usually guide modified proteins to proteasome for degradation. inactivating its downstream factors or by recruiting related K63-linked ubiquitin chains are thought to be involved in proteins. Particularly, histone phosphorylation has diverse protein–protein interactions in a number of important signaling functions in DNA damage repair, gene transcription and pathways (Komander 2009). The major form of histone chromatin condensation (Wei et al. 1999, Ivanovska et al. 2005, ubiquitination during meiosis is mono-ubiquitination, which Bell et al. 2011, Lange et al. 2011). During meiosis, histone functions to regulate gene transcription and change or to phosphorylation can facilitate DSB formation and repair during reshape the chromatin structures to facilitate meiotic progression homologous chromosome synapsis and recombination (Lan et al. 2016, Li et al. 2017). (Perez-Cadahia et al. 2010, Bell et al. 2011).

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R69

Figure 3 Patterns of histone modifications involved in meiosis during spermatogenesis. Spermatogenesis is a well-regulated process that starts from the self-renewal of type A spermatogonia and then differentiates into type B spermatogonia. Type B spermatogonia generate preleptotene spermatocytes, and then enter meiosis. Programmed DSB formation and homologous recombination takes place in the zygotene and pachytene stages, and their repairment is finished in the diplotene/ diakinesis stage. The secondary spermatocytes that originate from the segregation of the homologous chromosomes generate the haploid round spermatids after meiosis II. Afterward, round spermatids enter the spermiogenic phase and finally transform into spermatozoon. The sketch represents the patterns of histone modifications, which are involved in meiosis and spermatogenesis.

The histone codes of H2A in meiosis are arrested at the pachytene stage, the arrest is mainly caused by defected sex chromosome segregation and Among the four core histones, H2A contains the largest impaired MLH1 foci formation (Celeste et al. 2002). number of variants, including H2A.X, H2A.Z, MacroH2A, Phosphorylation of histone H2AX on S139 ( H2AX) H2A.Lap1, TH2A and H2A-Bbd (Monteiro et al. 2014, γ is a crucial histone code for programmed DSB repair. Shinagawa et al. 2015). During meiosis, H2A.X mainly Once the programmed DSB is formed, H2A.X is rapidly participates in homologous recombination in the early phosphorylated ( H2AX) by protein kinases ATM and stage of prophase I. Further investigation found that γ ATR (Rogakou et al. 1998, Stiff et al. 2004, Lee et al. H2ax-knockout female mice are fertile, albeit litter size 2014, DeLoughery et al. 2015). H2AX signal is very was smaller than the control group, while the H2ax- γ important for the rapid activation and recruitment of knockout male mice are infertile, and the spermatocytes

Figure 4 Patterns of histone modifications involved in meiosis of oogenesis. Oogenesis is a special gametogenesis process that is characterized by the presence of two growth arrests and two asymmetric divisions. The first arrest is at GV phase, which will be resumed again after birth. Some histone modifications are involved in the meiotic resumption process. After the GVBD stage, there are two successive asymmetric nuclear divisions to generate matured oocyte. The mature oocytes then arrested at metaphase II. That is the second arrest, and will be reactivated by the sperm after fertilization. www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access R70 L Wang, Z Xu and others programmed DSB repair proteins, including MDC1, proteins to the damage site, such as proteins 53BP1 and BRCA1, 53BP1 and RAD51, to the damage sites; these BRCA1 in somatic cells (Ma et al. 2011, Mallette et al. proteins further relax the chromatin around the damage 2012). The absence of RNF168 impairs long-term sites to promote effective DSB repair (Paull et al. 2000, spermatogenesis in 12-month-old male mice, but it has Stucki et al. 2005, Botuyan et al. 2006). Although no influence on 8-week-old male mice (Bohgaki et al. phosphorylation of H2AX is highly conserved from 2011), suggesting that the first wave of spermatogenesis yeast to mammals, the specific amino acid residue that is not affected, and the H2A ubiquitination might receives this modification differs between organisms indirectly affect meiotic recombination in an age- (Harvey et al. 2005, Moore et al. 2007). dependent manner (Nakada 2016). Polycomb repressive H2A.Z has important functions in transcriptional complex 1 (PRC1) is one of the polycomb group (PcG) regulation and is enriched on transcription start sites complexes in mammals, and it can catalyze mono- (TSS). During meiosis, the expression of H2A.Z is ubiquitination of H2A at lysine 119 (H2AK119ub) to increased in the pachytene stage, when meiotic sex suppress the expression of large numbers of genes during chromosome inactivation (MSCI) occurs, peaking development (Bohgaki et al. 2011). Sex comb on midleg- at the round spermatid stage, indicating that H2A.Z like 2 (SCML2) is a germline-specific subunit of PRC1, participates in the process of silencing sex chromosomes and it can promote RNF2-dependent ubiquitination of (Greaves et al. 2006, Guillemette & Gaudreau 2006). In H2A to mark somatic/progenitor genes on autosomes Arabidopsis thaliana, H2A.Z is associated with meiotic for repression. SCML2 can also recruit USP7, a recombination hot spots, and the mutation of ARP6, deubiquitinase, to the XY body in spermatocytes, thereby which is required for H2A.Z deposition, reduces the preventing RNF2-dependent ubiquitination of H2A on number of crossover, DMC1 or RAD51 foci, suggesting sex chromosomes during meiosis and enabling unique that H2A.Z promotes the formation or processing of epigenetic programming of sex chromosomes for male meiotic DNA DSBs (Choi et al. 2013). H2A.Z is also reproduction (Nakagawa et al. 2008, Hasegawa et al. essential for pericentric heterochromatin integrity and 2015, Luo et al. 2015). UBR2, an N-end rule proteolytic sister chromatid cohesion formation, and the deletion pathway ubiquitin ligase, can also catalyze H2A of H2A.Z results in defective chromosomal segregation ubiquitination by promoting the HR6B (E2)–H2A (Rangasamy et al. 2004). In female mice, H2A.Z is interaction and the HR6B-to-H2A transfer of ubiquitin. highly expressed during GV, GVBD, MII oocytes and UBR2-deficient spermatocytes are profoundly impaired the blastula stage. However, the precise roles of H2A.Z in chromosome-wide transcriptional silencing of genes in oocyte maturation and differentiation are largely linked to unsynapsed axes of the X and Y chromosomes unknown (Wu et al. 2014). (An et al. 2010). Some other H2A variants have also been identified In addition, the phosphorylation of H2A Thr119 has in meiosis. MacroH2A, is enriched on inactive X been reported to be required for proper chromosomal chromosomes and is capable of mediating X chromosome architecture establishment in Drosophila oocytes, which inactivation in both males and females by setting up a is mediated by nucleosomal histone kinase-1 (NHK-1), repressive chromatin environment (Angelov et al. 2003, and H2AT119 phosphorylation is also essential for Mietton et al. 2009). A novel H2A histone variant, H2A. synaptonemal complex disassembly and chromosome Lap1, has also been identified, and it is dynamically condensation during meiosis (Ivanovska et al. 2005). loaded onto the inactive X chromosome and directly H2AS121 can be phosphorylated in yeast during opens the chromatin structure of the TSS, enabling the meiosis, and this phosphorylation is mediated by BUB1 transcriptional activation of previously repressed genes (Kawashima et al. 2010). H2AS121 phosphorylation during spermatogenesis (Soboleva et al. 2012). TH2A might be important for shugoshin localization, and it is and TH2B are highly expressed in testis. Disruption required for centromeric cohesion protection in female of the Th2a and Th2b genes results in sterility due to meiosis I (Watanabe 2012). The Bub1-H2Aph-Sgo1 incomplete release of cohesin at interkinesis after pathway is highly conserved from yeast to mammals, meiosis I and the failure of histone replacement during albeit the phosphorylation site is T120 in humans spermiogenesis (Shinagawa et al. 2015). (Kawashima et al. 2010). As the first identified ubiquitination substrate, the physiological function of H2A ubiquitination remained unknown for a long time. In the DSB response signal The PTMs of histone H2B during meiosis pathway, H2A ubiquitination plays a key role in the transduction of the damage signals and recruitment of Both ubiquitination and phosphorylation of histone repair proteins. This H2A ubiquitination is mediated H2B have been identified to play important functional by RNF168, which can add K63-linked ubiquitin roles in meiosis (Fig. 2). Mono-ubiquitination of H2B on chains on H2A or H2AX and is assisted by UBC13 (E2) K120 has been found to peak in spermatocytes, and the (Mattiroli et al. 2012). H2A ubiquitination is crucial for RNF20/RNF40 complex is the E3 ligase responsible for recruiting downstream DNA damage response (DDR) the mono-ubiquitination of H2BK120 (Zhang & Yu 2011,

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R71

Xu et al. 2016). Germ cell-specific knockout of Rnf20 and zygotene stages, acetylated H3 lysine 9(K9) and results in the complete blockage of spermatogenesis. lysine 18(K18) are abundant in chromatin. In fission Therefore, RNF20-mediated H2BK120 mono- yeast, H3K9ac is enriched at meiotic recombination ubiquitination is indispensable for spermatogenesis. hotspots, and the absence of H3K9ac reduces the Further investigations reveal that this modification levels of the DSB-inducing protein Rec12 and DSB at mainly participates in meiotic recombination by hotspots, indicating that H3K9ac may be involved in promoting chromatin relaxation in the pachytene stage, DSB formation by enhancing the interaction between and a similar regulation mechanism is highly conserved Rec12 and hotspots (Hansen et al. 2011, Yamada et al. in yeast. Furthermore, H2B ubiquitination has been 2013). When spermatocytes enter the pachytene stage, reported to be involved in the initiation of programmed the meiotic genes are inactivated by a decreased DSBs during meiosis in yeast (Fig. 3) (Yamashita et al. bonding of H3K9ac and H3K18ac, suggesting that these 2004); while, this function may not apply to meiosis in modifications might also be involved in the meiotic genes mammals. transcription regulation (Khalil et al. 2004, Song et al. Histone H2BS10 phosphorylation in yeast occurs 2011, Shirakata et al. 2014). The level of H3K18ac in a cell cycle-dependent manner and peaks in the has been found to increase again in the diplotene and pachytene stage. This modification has been reported diakinesis stages (Khalil et al. 2004, Song et al. 2011), so to affect chromosomal condensation (Ahn et al. 2005). it is speculated that H3K18ac may also be required in However, this type of potential function in mammals the following meiotic events. still needs to be investigated. In females, in addition to the leptotene stage, there is large-scale gene transcription at the diplotene stage of oogenesis (Edwards 1965). A large amount of mRNAs The histone codes of H3 during meiosis for essential proteins in oocyte maturation and zygote The histone codes of histone H3 have been well studied, development after fertilization are synthesized during the including acetylation, methylation, phosphorylation and GV phase (De La Fuente 2006, Virant-Klun et al. 2013). some of its variants (Fig. 2), and their functional roles H3K9ac and H3K14ac exist in these active transcription will be introduced one-by-one in the following sections. regions to create a microenvironment for recruiting transcriptional factors and RNA polymerase II at the GV stage in oocytes (Kim et al. 2003, De La Fuente et al. 2004, Gu et al. 2010, Ding et al. 2012). After meiosis resumes Histone H3 acetylation in meiosis at the GVBD stage, these acetylations are removed by Many of the lysine residues presented on H3 can be histone deacetylases (HDACs) to further condense the acetylated. The acetylations of K9, K14, K18 and K56 chromatin for meiotic division (Akiyama et al. 2004, have been reported to be involved in various aspects Ma & Schultz 2016). of meiosis. Acetylated H3 lysine 56 (H3K56ac) can be detected in the premeiotic S phase and persists during Histone H3 methylation during meiosis meiotic recombination in budding yeast. A histone chaperone, ASF1, is necessary for H3K56ac, as the Histone H3 is the major target of methyltransferase. disruption of Asf1 causes the disappearance of this Lysines 4, 9, 27, 36 and 79 of H3 can be modified by modification. In C. elegans, the mutants of two C. elegans different histone methyltransferases (HMTases), and they Asf1 homologs, ASFL-1 and UNC-85, influence DNA mainly exert their functions in the pachytene stage. replication in the germline (Grigsby et al. 2009). In The methylations of H3K4, H3K9 and H3K36 have addition, the absence of ASF1B in mice delays the been detected on meiotic chromatin from leptonema meiosis initiation both in males and female, while the (Hochwagen & Marais 2010, Grey et al. 2011, subsequent gonad development is affected more severely Powers et al. 2016). Recent studies also reveal that H3K4, in Asf1b-null female mice than that in male mice, thus H3K9 and H3K36 could be methylated by PR domain- resulting in the reproductive capacity were severely containing 9 (PRDM9, also known as Meisetz) and that reduced in females (Messiaen et al. 2016). H3K56 they participate in meiotic recombination initiation in acetylation is also necessary for genome stability and spermatocytes (Powers et al. 2016). PRDM9 is a hotspot chromosome assembly in response to DNA damage in marker protein of meiotic recombination initiation yeast (Recht et al. 2006, Simoneau et al. 2015). In mice, (Berg et al. 2010, Parvanov et al. 2010, Grey et al. H3K56ac, together with H3K9ac, has been reported to 2011, Brick et al. 2012, Wu et al. 2013, Pratto et al. participate in DSB repair possibly by relaxing chromatin 2014, Sun et al. 2015, Patel et al. 2016). Direct binding structures on the damage sites (Masumoto et al. 2005, of PRDM9 to the 13-mer hotspot DNA motif mainly Tjeertes et al. 2009, Battu et al. 2011, Guo et al. 2011, depends on its C2H2 Zinc finger domain (Hayashi et al. Gong & Miller 2013, Zhong et al. 2016), suggesting a 2005, Myers et al. 2008), while the PR/SET domain is similar mechanism might exist during programmed DSB responsible for methylating many histone lysine residues. initiation and meiotic recombination. At the preleptotene H3K9 can also be methylated by G9a and Suv39h. www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access R72 L Wang, Z Xu and others

G9a is a major mammalian H3K9 methyltransferase, Histone H3 phosphorylation during meiosis G9a depletion impairs H3K9 (me1/3) methylation and During oocyte maturation, HASPIN-mediated H3T3 results in abnormal meiotic homologous recombination, phosphorylation is required for meiotic resumption and which leads to the failure of spermatogenesis anaphase onset (Nguyen et al. 2014, Wang et al. 2016). (Tachibana et al. 2001, 2007, Takada et al. 2011). In addition, phosphorylation of histone H3 at S10 is Suv39h has 2 isoforms, and Suv39h2 is highly expressed another important modification for oocyte maturation in adult testis (O’Carroll et al. 2000). Suv39h catalyzes that ensures proper chromosomal condensation the methylation of H3K9 at a specific stage in which and subsequent segregation. The absence of H3S10 the spermatogonia transform into spermatocytes, and phosphorylation inhibits meiosis and causes defects in Suv39h is mainly responsible for pericentric H3K9 meiotic chromosome condensation and segregation in methylation. Loss of Suv39h results in nonhomologous both Tetrahymena and worms (Wei et al. 1998, 1999, interactions and impaired synapsis, particularly for Hsu et al. 2000). Furthermore, H3S10 phosphorylation the sex chromosomes. The depletion of Suv39h also was detected in yeast and mammals during meiosis impedes chromosome segregation and specialized (Song et al. 2011), so we speculate that H3S10 chromatin structure formation (Peters et al. 2001). In phosphorylation might be essential for meiotic division addition, the cooperation of H3K4 methyltransferase during meiosis in an evolutionary conserved manner. Set1C/COMPASS and the Mer2/Mei4/Rec114 complex have been found to promote meiotic homologous recombination (Nottke et al. 2011, Sommermeyer et al. Histone H3 variants in meiosis 2013). In yeast, histone methyltransferase Dot1 works together with H3K79me to participate in a meiotic The variants of histone H3 play essential roles in every recombination checkpoint. If meiotic DSBs were stage of spermatogenesis. H3.3 is an evolutionarily unrepaired and/or interhomolog interactions are faulty, conserved variant involved in transcriptional regulation Dot1-dependent H3K79me can exclude Pch2 from the (Sakai et al. 2009). Histone variant H3.3 is encoded by chromosomes and promote Mek1 autophosphorylation, two genes in the mouse, H3f3a and H3f3b, but they have thereby driving Hop1 localization along chromosome different functions during gametogenesis, as knockouts axes and activating the pachytene checkpoint to trigger of either of these genes cause different phenotypes. Adult downstream responses (Ontoso et al. 2013). Dot1L- and female H3f3a mutants are fertile, while male H3f3a Dot1L-mediated H3K79methylation are essential for mutants are subfertile with abnormal sperm, which is oogenesis, as any impairment of them results in oocyte mainly caused by impaired transformation from round to arrest at metaphase of meiosis I (Wang et al. 2014). elongated spermatids. Loss of H3f3b is lethal (Tang et al. The ESET/SETDB1 complex is a methyltransferase for 2015). A decreased level of H3.3b results in remarkable H3K9me2 in oocytes, and it has irreplaceable functions enrichment of H3K9me3 in spermatogonia and in pre- in oocyte maturation. Deficiency ofSetdb1 greatly leptotene, leptotene, zygotene and early (stage I–V) impairs oocyte maturation, meiosis resumption, spindle pachytene spermatocytes during spermatogenesis, and formation and distribution, and also chromosome it not only disrupts the transcription of related genes but dynamics (Eymery et al. 2016, Kim et al. 2016). also changes chromosome structures, thereby inhibiting Many histone H3 methylation have been found to be histone replacement (Yuen et al. 2014). In addition, the involved in MSCI. For instance, H3K4me3 disappears functional roles of H3.3 in nucleosome replacement from the XY body, while H3K9me3, H3K27me3 after MSCI or meiotic silencing of unsynapsed chromatin and H3K36me3 are accumulated on it, and all of might be evolutionary conserved from fly to mammals them can indicate the transcriptional silencing state (van der Heijden et al. 2007, Sakai et al. 2009). H3.3 of spermatocytes (Berletch et al. 2010, Boggs et al. is also essential for oogenesis. Conditional ablation of 2002). H3 lysine79 methylation (H3K79me1, me2 and H3f3b at the beginning of the folliculogenesis stage me3) may also be involved in MSCI maintenance. The results in zygote cleavage failure, while simultaneous distribution of all three types of H3K79me is diverse ablation of H3f3a and H3f3b in the folliculogenesis during spermatogenesis. Lower levels of H3K79me1 are stage leads to primary oocyte death (Tang et al. 2015). found in the prophase spermatocytes, while H3K79me2 Recently, a new H3 variant, H3t, was found in mice. and H3K79me3 levels start to increase at the pachytene H3t has a counterpart H3.4 (H3T) in humans, and both stage. H3K79me3 is enriched on the heterochromatic of them are specifically expressed in testis, especially centromeric regions and XY body, while H3K79me2 in B-type spermatogonia and early-stage spermatocytes. is detected on all chromatins except the XY body. During spermatogonia differentiation, H3t replaces Therefore, H3K79me3 and H3K79me2 may have canonical H3 proteins, thus forming a more open antagonistic functions, H3K79me3 may be involved in chromatin structure. H3t (H3T) containing nucleosomes MSCI and centromeric region repression and H3K79me2 are required for spermatogonial differentiation, and is related to transcriptional reactivation on autosomes ensure entry into meiosis, as H3t gene deficiency (Ontoso et al. 2014). leads to male infertility due to the failure of germ cell

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R73 differentiation from spermatogonia to spermatocytes in the diplotene and diakinesis stages (Khalil et al. 2004, (Ueda et al. 2017). Song et al. 2011), indicating that H4K16ac may also be CENPA is another histone H3 variant that is involved in the following events during meiosis. specifically localized at the centromeres and modulates In females, high levels of H4K5ac, H4K8ac, their behavior (Niikura et al. 2015). Recently, CENPA has H4K12ac and H4K16ac can be detected on meiotic been reported to participate in meiotic recombination chromosomes (Kim et al. 2003, De La Fuente et al. by recruiting RAD51 to the nuclear periphery in a 2004, Kageyama et al. 2007, Gu et al. 2010, Ding et al. SAC- and a DDR-dependent manner in C. elegans germ 2012). They are necessary for the resumption of meiosis cells (Lawrence et al. 2015). In humans, a novel H3 at the GVBD stage. In addition, H4 deacetylation, variant, H3.5, has been found to accumulate around including H4K8, H4K12, H4K14 and H4K16, is then the TSS region in human testis, forming an unstable accompanied by two asymmetric meiotic divisions, and nucleosome. This variant might be involved in regulating inadequate meiotic histone deacetylation may result in chromatin dynamics during human spermatogenesis aneuploidy and embryo abnormality (Akiyama et al. (Urahama et al. 2016). 2006, Ma & Schultz 2013). In Hdac2-knockout mouse oocytes, the hyperacetylation of H4K16 impairs kinetochore function, with unstable microtubule The PTMs of histone H4 during meiosis attachments and less CENPA located at the centromeres, The PTMs of histone H4 play important roles in almost thereby causing defective chromosome condensation all stages of meiosis (Fig. 2). During the proleptotene and segregation (Ma & Schultz 2013). During human and the zygotene stages, H4 lysine 5, 8, 12 and 16 are oocyte maturation, abundant H4K12ac is closely linked acetylated and are concentrated on chromatin. They to chromosome misalignment (van den Berg et al. were reported to participate in gene transcription during 2011), further indicating that normal deacetylation is many cellular processes. For example, H4K5ac has been important for normal oocyte development. H4K20me3 reported to promote the expression of cognitive-related is also required for proper chromosome alignment genes (Park et al. 2013). Therefore, we speculate that they and polar body extrusion during meiosis I, because might be involved in male meiotic gene transcription knockdown of Suv4-20h, an H4K20 methyltransferase, (Khalil et al. 2004, Song et al. 2011, Shirakata et al. remarkably decreases H4K20me3, resulting in a high 2014). In yeast, H4K44ac is predominantly enriched proportion of abnormal meiotic metaphase I oocytes at meiotic recombination hotspots during meiosis. The (Xiong et al. 2013). absence of H4K44ac results in increased nucleosomal occupancy around DSB hotspots, indicating that H4K44ac is essential for normal DSB formation and Cross-talk among the histone codes meiotic recombination by facilitating chromatin Phosphorylation, acetylation, methylation and accessibility (Hu et al. 2015). Recently, it was revealed ubiquitination are the most important PTMs of histones that several histone H3 and H4 acetylations harbored during meiosis, many modifications often occur on open and active chromatin regions promote the simultaneously in the same events, or even at the same recruitment of PRDM9 and Spo11, thereby facilitating sites of histones, and they interact and influence each programmed DSBs formation at the leptotene stage and other to ultimately determine the meiotic process. crossover resolution at the pachytene stage (Getun et al. The PRC1 and PRC2 complexes can regulate gene 2016) (Fig. 3). transcription during meiosis, and their functions are During the zygotene stage, H4K5ac, H4K12ac and mainly modulated by cross-talk between H2AK119 H4K16ac maintain relatively higher levels. H4K16ac has ubiquitination and H3K27me3. First, the PRC2 been reported to participate in DSB repair by relaxing complex is loaded onto the chromatin that catalyzes chromatin around the DNA damage sites (Tjeertes et al. H3K27 trimethylation, which, in turn, recruits PRC1 to 2009, Battu et al. 2011, Guo et al. 2011, Gong & Miller ubiquitinate H2AK119; this ubiquitination is important for 2013, Zhong et al. 2016). polycomb group (PcG)-mediated silencing (Margueron & During the pachytene stage, transcriptional Reinberg 2011, Cooper et al. 2016). Furthermore, NHK- repression first initiates on the XY body. The methylation 1-mediated H2AT119 phosphorylation is a prerequisite and acetylation of H4 might be involved in MSCI. for H3K14 and H4K5 acetylation, and the disruption H4K20me3, H4K12ac and H4K16ac are excluded of NHK-1 results in the loss of H3K14ac and H4K5ac from the X and Y chromosomes, while H4K5ac and (Ivanovska et al. 2005). Recently, we also found that H4K8ac persist on them, suggesting that H4K5ac and H2BK120 ubiquitination affects H3K14ac (marker H4K8ac might be involved in MSCI formation during of loosened chromatin) and H3K4me2 (marker of meiosis; however, their real functions still need further condensed chromatin) on autosomes (Xu et al. 2016), investigation (Vaskova et al. 2010, Lawrence et al. indicating an interplay of these modifications during 2015). It is noteworthy that H4K16ac is increased again meiosis. As increased studies focus on this aspect, more www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access R74 L Wang, Z Xu and others

(6) With the development of chromosome conformation capture techniques (3C) to 4C, 5C and Hi-C, can these regulations be investigated in a three- dimensional manner in the near future? The abovementioned questions definitely need further investigation, and the answers to these questions will help us to understand their relationships and the exact regulating mechanisms of meiosis. Histone codes are used to write an elegant program to direct complex meiotic process, and the dissection of these codes will shed light on various aspects of meiosis.

Declaration of interest The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding This work was supported by the National Natural Science Foundation of China (grants 91519317, 91649202 and 31471277) and National Key R&D Program of China (grant 2016YFA0500901).

Acknowledgement The authors apologize to the colleagues whose work could not be cited due to space limitation.

Figure 5 Summary of the functions of histone modifications during male (A) and female (B) meiosis. (A) Profile summary of histone modifications in some events of spermatogenesis. (B) Summary of histone modifications in some events of oogenesis. References Acquaviva L, Szekvolgyi L, Dichtl B, Dichtl BS, Saint Andre CD, Nicolas A and more cross-talk among these histone codes will be The following open questions in the field still need an & Geli V 2013 The COMPASS Subunit Spp1 Links Histone Methylation recognized in the future (Fig. 5). in-depth investigation. to Initiation of Meiotic Recombination. Science 339 215–218. (doi:10.1126/science.1225739) (1) Besides the above mentioned PTMs, are there other Ahn SH, Henderson KA, Keeney S & Allis CD 2005 H2B (Ser10) phosphorylation is induced during apoptosis and meiosis in S-cerevisiae. Future perspectives on histone modifications novel histone PTMs involved in meiosis? How can we identify them? Cell Cycle 4 780–783. (doi:10.4161/cc.4.6.1745) during meiosis Akiyama T, Kim JM, Nagata M & Aoki F 2004 Regulation of histone (2) How do we establish the epigenetic modulation acetylation during meiotic maturation in mouse oocytes. Molecular A wide variety of histone modifications form the highly network during meiosis? For example, once the Reproduction and Development 69 222–227. (doi:10.1002/mrd.20121) complicated, but well organized, regulatory network programmed DSBs have been formed during Akiyama T, Nagata M, & Aoki F 2006 Inadequate histone deacetylation during oocyte meiosis causes aneuploidy and embryo death in mice. during meiosis, ensuring that this process sequentially meiosis, which type of histone code is initially used? PNAS 103 7339–7344. (doi:10.1073/pnas.0510946103) progresses according to a well-designed program and (3) How do we restrict the histone modifications in a An JY, Kim EA, Jiang YH, Zakrzewska A, Kim DE, Lee MJ, Mook-Jung I, ultimately produces haploid gametes. The dramatic specific region of the meiotic chromosome? What Zhang Y & Kwon YT 2010 UBR2 mediates transcriptional silencing during spermatogenesis via histone ubiquitination. PNAS 107 renovation of chromatin structures during meiosis mechanism ensures that histone modifications only 1912–1917. (doi:10.1073/pnas.0910267107) creates a tremendous demand for PTMs, variants and occur at particular positions? Angelov D, Molla A, Perche PY, Hans F, Cote J, Khochbin S, Bouvet P the replacement of histones. Thus, the histone codes (4) How do we control the range of chromatin & Dimitrov S 2003 The histone variant macroH2A interferes with inevitably become the main players in meiosis. Although remodeling? During meiotic recombination, when transcription factor binding and SWI/SNF nucleosome remodeling. Molecular Cell 11 1033–1041. (doi:10.1016/S1097-2765(03)00100-X) numerous studies have been performed to decipher the the chromatin needs to be loosened, will the whole Baarends WM, Wassenaar E, van der Laan R, Hoogerbrugge J, Sleddens- histone codes during meiosis, most of them are still chromatin or only the regions near the DSB sites be Linkels E, Hoeijmakers JHJ, de Boer P & Grootegoed JA 2005 Silencing descriptive and correlative, and direct manipulation of relaxed? If the answer is the latter, is there a type of of unpaired chromatin and histone H2A ubiquitination in mammalian antagonistic PTM to limit it? meiosis. Molecular and Cellular Biology 25 1041–1053. (doi:10.1128/ the histone modification sites is urgently needed to study MCB.25.3.1041-1053.2005) their bona fide functions. With the rapid development (5) What is the relationship among these PTMs? What Bannister AJ, Schneider R & Kouzarides T 2002 Histone methylation: of gene editing tools, such as the CRISPR/Cas9 system, is the mechanism underlying the cross-talk among dynamic or static? Cell 109 801–806. (doi:10.1016/S0092- these problems may be solved in the near future. these PTMs? 8674(02)00798-5)

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R75

(6) With the development of chromosome conformation Battu A, Ray A & Wani AA 2011 ASF1A and ATM regulate H3K56-mediated capture techniques (3C) to 4C, 5C and Hi-C, cell-cycle checkpoint recovery in response to UV irradiation. Nucleic Acids Research 39 7931–7945. (doi:10.1093/nar/gkr523) can these regulations be investigated in a three- Beck-Sickinger AG & Mörl K 2006 Posttranslational modification of dimensional manner in the near future? proteins. Expanding nature’s inventory. By Christopher T. Walsh. Angewandte Chemie International Edition 45 1020–1020. (doi:10.1002/ The abovementioned questions definitely need anie.200585363) further investigation, and the answers to these questions Bedford MT & Clarke SG 2009 Protein arginine methylation in mammals: who, what, and why. Molecular Cell 33 1–13. (doi:10.1016/j. will help us to understand their relationships and the molcel.2008.12.013) exact regulating mechanisms of meiosis. Histone codes Bell O, Tiwari VK, Thoma NH & Schubeler D 2011 Determinants and are used to write an elegant program to direct complex dynamics of genome accessibility. Nature Reviews Genetics 12 meiotic process, and the dissection of these codes will 554–564. (doi:10.1038/nrg3017) Berg IL, Neumann R, Lam KWG, Sarbajna S, Odenthal-Hesse L, May CA shed light on various aspects of meiosis. & Jeffreys AJ 2010 PRDM9 variation strongly influences recombination hot-spot activity and meiotic instability in humans. Nature Genetics 42 859. (doi:10.1038/ng.658) Declaration of interest Berletch JB, Yang F & Disteche CM 2010 Escape from X inactivation in mice and humans. Genome Biology 11 213. (doi:10.1186/gb-2010-11- The authors declare that there is no conflict of interest that 6-213) could be perceived as prejudicing the impartiality of this Blanco-Rodriguez J 2012 Programmed phosphorylation of histone H2AX review. precedes a phase of DNA double-strand break-independent synapsis in mouse meiosis. Reproduction 144 699–712. (doi:10.1530/REP-12-0326) Boggs BA, Cheung P, Heard E, Spector DL, Chinault AC & Allis CD 2002 Differentially methylated forms of histone H3 show unique association Funding patterns with inactive human X chromosomes. Nature Genetics 30 This work was supported by the National Natural Science 73–76. (doi:10.1038/ng787) Bohgaki T, Bohgaki M, Cardoso R, Panier S, Zeegers D, Li L, Stewart GS, Foundation of China (grants 91519317, 91649202 and Sanchez O, Hande MP, Durocher D et al. 2011 Genomic instability, 31471277) and National Key R&D Program of China (grant defective spermatogenesis, immunodeficiency, and cancer in a mouse 2016YFA0500901). model of the RIDDLE syndrome. PLoS Genetics 7. (doi:10.1371/journal. pgen.100138) Borner GV, Kleckner N & Hunter N 2004 Crossover/noncrossover differentiation, synaptonemal complex formation, and regulatory Acknowledgement surveillance at the leptotene/zygotene transition of meiosis. Cell 117 The authors apologize to the colleagues whose work could not 29–45. (doi:10.1016/S0092-8674(04)00292-2) Borde V, Robine N, Lin W, Bonfils S, Geli V & Nicolas A 2009 Histone be cited due to space limitation. H3 lysine 4 trimethylation marks meiotic recombination initiation sites. EMBO Journal 28 99–111. (doi:10.1038/emboj.2008.257) Botuyan MV, Lee J, Ward IM, Kim JE, Thompson JR, Chen JJ & Mer G References 2006 Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair. Cell 127 1361–1373. Acquaviva L, Szekvolgyi L, Dichtl B, Dichtl BS, Saint Andre CD, Nicolas A (doi:10.1016/j.cell.2006.10.043) & Geli V 2013 The COMPASS Subunit Spp1 Links Histone Methylation Brick K, Smagulova F, Khil P, Camerini-Otero RD & Petukhova GV 2012 to Initiation of Meiotic Recombination. Science 339 215–218. Genetic recombination is directed away from functional genomic (doi:10.1126/science.1225739) elements in mice. Nature 485 642–645. (doi:10.1038/nature11089) Ahn SH, Henderson KA, Keeney S & Allis CD 2005 H2B (Ser10) Brush GS 2015 Evidence that histone H1 is dispensable for proper phosphorylation is induced during apoptosis and meiosis in S-cerevisiae. meiotic recombination in budding yeast. BMC Research Notes 8 275. Cell Cycle 4 780–783. (doi:10.4161/cc.4.6.1745) (doi:10.1186/s13104-015-1246-1) Akiyama T, Kim JM, Nagata M & Aoki F 2004 Regulation of histone Buard J, Barthes P, Grey C & de Massy B 2009 Distinct histone modifications acetylation during meiotic maturation in mouse oocytes. Molecular define initiation and repair of meiotic recombination in the mouse. Reproduction and Development 69 222–227. (doi:10.1002/mrd.20121) EMBO Journal 28 2616–2624. (doi:10.1038/emboj.2009.207) Akiyama T, Nagata M, & Aoki F 2006 Inadequate histone deacetylation Callis J 2014 The ubiquitination machinery of the ubiquitin system. during oocyte meiosis causes aneuploidy and embryo death in mice. Arabidopsis Book 12 e0174. (doi:10.1199/tab.0174) PNAS 103 7339–7344. (doi:10.1073/pnas.0510946103) Cavero S, Herruzo E, Ontoso D & San-Segundo P 2016 Impact of An JY, Kim EA, Jiang YH, Zakrzewska A, Kim DE, Lee MJ, Mook-Jung I, Histone H4K16 acetylation on the meiotic recombination checkpoint Zhang Y & Kwon YT 2010 UBR2 mediates transcriptional silencing in Saccaromyces cerevisiae. Microbial Cell 3 475–489. (doi:10.15698/ during spermatogenesis via histone ubiquitination. PNAS 107 mic2016.12.548) 1912–1917. (doi:10.1073/pnas.0910267107) Celeste A, Petersen S, Romanienko PJ, Fernandez-Capetillo O, Chen Angelov D, Molla A, Perche PY, Hans F, Cote J, Khochbin S, Bouvet P HT, Sedelnikova OA, Reina-San-Martin B, Coppola V, Meffre E, & Dimitrov S 2003 The histone variant macroH2A interferes with Difilippantonio MJ et al. 2002 Genomic instability in mice lacking transcription factor binding and SWI/SNF nucleosome remodeling. histone H2AX. Science 296 922–927. (doi:10.1126/science.1069398) Molecular Cell 11 1033–1041. (doi:10.1016/S1097-2765(03)00100-X) Choi KH, Zhao XH, Kelly KA, Venn O, Higgins JD, Yelina NE, Hardcastle Baarends WM, Wassenaar E, van der Laan R, Hoogerbrugge J, Sleddens- TJ, Ziolkowski PA, Copenhaver GP, Franklin FCH et al. 2013 Arabidopsis Linkels E, Hoeijmakers JHJ, de Boer P & Grootegoed JA 2005 Silencing meiotic crossover hot spots overlap with H2A. Z nucleosomes at gene of unpaired chromatin and histone H2A ubiquitination in mammalian promoters. Nature Genetics 45 1327. (doi:10.1038/ng.2766) meiosis. Molecular and Cellular Biology 25 1041–1053. (doi:10.1128/ Clancy MJ 1998 Meiosis: step-by-step through sporulation. Current Biology MCB.25.3.1041-1053.2005) 8 R461–R463. (doi:10.1016/S0960-9822(98)70293-3) Bannister AJ, Schneider R & Kouzarides T 2002 Histone methylation: Cooper S, Grijzenhout A, Underwood E, Ancelin K, Zhang TY, Nesterova dynamic or static? Cell 109 801–806. (doi:10.1016/S0092- TB, Anil-Kirmizitas B, Bassett A, Kooistra SM, Agger K et al. 2016 Jarid2 8674(02)00798-5) binds mono-ubiquitylated H2A lysine 119 to mediate crosstalk between

www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access R76 L Wang, Z Xu and others

Polycomb complexes PRC1 and PRC2. Nature Communications 7. Conference, and has undergone the Journal’s usual peer review process. (doi:10.1038/ncomms13661) Biochemistry and Cell Biology 84 528–535. (doi:10.1139/o06-077) De La Fuente R 2006 Chromatin modifications in the germinal vesicle Guo RF, Chen J, Mitchell DL & Johnson DG 2011 GCN5 and E2F1 (GV) of mammalian oocytes. Developmental Biology 292 1–12. stimulate nucleotide excision repair by promoting H3K9 acetylation at (doi:10.1016/j.ydbio.2006.01.008) sites of damage. Nucleic Acids Research 39 1390–1397. (doi:10.1093/ De La Fuente R, Viveiros MM, Burns KH, Adashi EY, Matzuk MM & Eppig nar/gkq983) JJ 2004 Major chromatin remodeling in the germinal vesicle (GV) of Hansen L, Kim NK, Marino-Ramirez L & Landsman D 2011 Analysis of mammalian oocytes is dispensable for global transcriptional silencing biological features associated with meiotic recombination hot and cold but required for centromeric heterochromatin function. Developmental spots in saccharomyces cerevisiae. PLoS ONE 6 e29711. (doi:10.1371/ Biology 275 447–458. (doi:10.1016/j.ydbio.2004.08.028) journal.pone.0029711) DeLoughery Z, Luczak MW, Ortega-Atienza S & Zhitkovich A 2015 DNA Harvey AC, Jackson SP & Downs JA 2005 Saccharomyces cerevisiae double-strand breaks by Cr(VI) are targeted to euchromatin and cause histone H2A Ser122 facilitates DNA repair. Genetics 170 543–553. ATR-dependent phosphorylation of histone H2AX and its ubiquitination. (doi:10.1534/genetics.104.038570) Toxicological Sciences 143 54–63. (doi:10.1093/toxsci/kfu207) Hasegawa K, Sin HS, Maezawa S, Broering TJ, Kartashov AV, Alavattam Ding L, Pan R, Huang X, Wang JX, Shen YT, Xu L, Zhang Y, Liu Y, He KG, Ichijima Y, Zhang F, Bacon WC, Greis KD et al. 2015 SCML2 XQ, Yang XJ et al. 2012 Changes in histone acetylation during oocyte establishes the male germline epigenome through regulation of histone meiotic maturation in the diabetic mouse. Theriogenology 78 784–792. H2A ubiquitination. Developmental Cell 32 574–588. (doi:10.1016/j. (doi:10.1016/j.theriogenology.2012.03.026) devcel.2015.01.014) Downs SM 1996 Regulation of meiotic arrest and resumption in mammalian Hawley RS 2002 Meiosis: how male flies do meiosis.Current Biology 12 oocytes. Ovary: Regulation, Dysfunction and Treatment 1106 141–148. R660–R662. (doi:10.1016/S0960-9822(02)01161-2) Drabent B, Saftig P, Bode C & Doenecke D 2000 Spermatogenesis Hayashi K, Yoshida K & Matsui Y 2005 A histone H3 methyltransferase proceeds normally in mice without linker histone H1t. Histochemistry controls epigenetic events required for meiotic prophase. Nature 438 and Cell Biology 113 433–442. (doi:10.1007/s004180000146) 374–378. (doi:10.1038/nature04112) Eberharter A & Becker PB 2002 Histone acetylation: a switch between Hershko A & Ciechanover A 1998 The ubiquitin system. Annual Review repressive and permissive chromatin – second in review series on of Biochemistry 67 425–479. (doi:10.1146/annurev.biochem.67.1.425) chromatin dynamics. EMBO Reports 3 224–229. (doi:10.1093/embo- Hiragami-Hamada K, Soeroes S, Nikolov M, Wilkins B, Kreuz S, Chen C, reports/kvf053) De-La Rosa Velázquez IA, Zenn HM, Kost N, Pohl W et al. 2016 Dynamic Edwards RG 1965 Maturation in vitro of mouse sheep cow pig rhesus monkey and flexible H3K9me3 bridging via HP1β dimerization establishes a and human ovarian oocytes. Nature 208 349. (doi:10.1038/208349a0) plastic state of condensed chromatin. Nature Communications 7 11310. Eymery A, Liu Z, Ozonov EA, Stadler MB & Peters AHFM 2016 The (doi:10.1038/ncomms11310) methyltransferaseSetdb1is essential for meiosis and mitosis in mouse Hochwagen A & Marais GAB 2010 Meiosis: a PRDM9 guide to the hotspots oocytes and early embryos. Development 143 2767–2779. (doi:10.1242/ of recombination. Current Biology 20 R271–R274. (doi:10.1016/j. dev.132746) cub.2010.01.048) Franke M, Ibrahim DM, Andrey G, Schwarzer W, Heinrich V, Schopflin R, Hu JL, Donahue G, Dorsey J, Govin J, Yuan ZF, Garcia BA, Shah PP & Kraft K, Kempfer R, Jerkovic I, Chan WL et al. 2016 Formation of new Berger SL 2015 H4K44 acetylation facilitates chromatin accessibility chromatin domains determines pathogenicity of genomic duplications. during meiosis. Cell Reports 13 1772–1780. (doi:10.1016/j. Nature 538 265. (doi:10.1038/nature19800) celrep.2015.10.070) Getun IV, Wu Z, Fallahi M, Ouizem S, Liu Q, Li WM, Costi R, Roush WR, Iizuka M & Smith MM 2003 Functional consequences of histone Cleveland JL & Bois PRJ 2017 Functional roles of acetylated histone modifications. Current Opinion in Genetics and Development 13 marks at mouse meiotic recombination hot spots. Molecular and Cellular 154–160. (doi:10.1016/S0959-437X(03)00020-0) Biology 37 e00942-15. (doi:10.1128/MCB.00942-15) Isono E & Nagel MK 2014 Deubiquitylating enzymes and their emerging Godmann M, Auger V, Ferraroni-Aguiar V, Di Sauro A, Sette C, Behr R role in plant biology. Frontiers in Plant Science 5. (doi:10.3389/ & Kimmins S 2007 Dynamic regulation of histone h3 methylation at fpls.2014.00056) lysine 4 in mammalian spermatogenesis. Biology of Reproduction 77 Ivanovska I, Khandan T, Ito T & Orr-Weaver TL 2005 A histone code in 754–764. (doi:10.1095/biolreprod.107.062265) meiosis: the histone kinase, NHK-1, is required for proper chromosomal Gong FD & Miller KM 2013 Mammalian DNA repair: HATs and HDACs architecture in Drosophila oocytes. Genes and Development 19 make their mark through histone acetylation. Mutation Research- 2571–2582. (doi:10.1101/gad.1348905) Fundamental and Molecular Mechanisms of Mutagenesis 750 23–30. Izzo A & Schneider R 2016 H1 gets the genome in shape. Genome (doi:10.1016/j.mrfmmm.2013.07.002) Biology 17 1–3. (doi:10.1186/s13059-016-0872-9) Govin J & Khochbin S 2013 Histone variants and sensing of chromatin Jenuwein T & Allis CD 2001 Translating the histone code. Science 293 functional states. Nucleus 4 438–442. (doi:10.4161/nucl.27088) 1074–1080. (doi:10.1126/science.1063127) Greaves IK, Rangasamy D, Devoy M, Graves JAM & Tremethick DJ 2006 Kadoch C, Copeland RA & Keilhack H 2016 PRC2 and SWI/SNF chromatin The X and Y chromosomes assemble into H2A.Z- containing facultative remodeling complexes in health and disease. Biochemistry 55 heterochromatin following meiosis (vol 26, pg 5394, 2006). Molecular 1600–1614. (doi:10.1021/acs.biochem.5b01191) and Cellular Biology 26 7343–7343. (doi:10.1128/MCB.01407-06) Kageyama S, Liu HL, Kaneko N, Ooga M, Nagata M & Aoki F 2007 Grey C, Barthes P, Chauveau-Le Friec G, Langa F, Baudat F & de Massy B Alterations in epigenetic modifications during oocyte growth in mice. 2011 Mouse PRDM9 DNA-binding specificity determines sites of histone Reproduction 133 85–94. (doi:10.1530/REP-06-0025) H3 lysine 4 trimethylation for initiation of meiotic recombination. PLoS Kato Y, Alavattam KG, Sin HS, Meetei AR, Pang QS, Andreassen PR Biology 9. (doi:10.1371/journal.pbio.1001176) & Namekawa SH 2015 FANCB is essential in the male germline and Grigsby IF, Rutledge EM, Morton CA & Finger FP 2009 Functional regulates H3K9 methylation on the sex chromosomes during meiosis. redundancy of two C. elegans homologs of the histone chaperone Human Molecular Genetics 24 5234–5249. (doi:10.1093/hmg/ddv244) Asf1 in germline DNA replication. Developmental Biology 329 64–79. Kawashima SA, Yamagishi Y, Honda T, Ishiguro K & Watanabe Y 2010 (doi:10.1016/j.ydbio.2009.02.015) Phosphorylation of H2A by Bub1 prevents chromosomal instability Grunstein M 1997 Histone acetylation in chromatin structure and through localizing shugoshin. Science 327 172–177. (doi:10.1126/ transcription. Nature 389 349–352. (doi:10.1038/38664) science.1180189) Gu L, Wang Q & Sun QY 2010 Histone modifications during mammalian Keeney S, Giroux CN & Kleckner N 1997 Meiosis-specific DNA oocyte maturation dynamics, regulation and functions. Cell Cycle 9 double-strand breaks are catalyzed by Spo11, a member of a widely 1942–1950. (doi:10.4161/cc.9.10.11599) conserved protein family. Cell 88 375–384. (doi:10.1016/S0092- Guillemette B & Gaudreau L 2006 Reuniting the contrasting functions of 8674(00)81876-0) H2A.ZThis paper is one of a selection of papers published in this special Khalil AM, Boyar FZ & Driscoll DJ 2004 Dynamic histone modifications issue, entitled 27th International West Coast Chromatin and Chromosome mark sex chromosome inactivation and reactivation during

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R77

mammalian spermatogenesis. PNAS 101 16583–16587. (doi:10.1073/ USP7 and counteracts histone H2A ubiquitination in the XY chromatin pnas.0406325101) during male meiosis. PLoS Genetics 11 e1004954. (doi:10.1371/journal. Kim JM, Liu HL, Tazaki M, Nagata M & Aoki F 2003 Changes in histone pgen.1004954) acetylation during mouse oocyte meiosis. Journal of Cell Biology 162 Ma PP & Schultz RM 2013 Histone deacetylase 2 (HDAC2) regulates 37–46. (doi:10.1083/jcb.200303047) chromosome segregation and kinetochore function via H4K16 Kim J, Zhao HB, Dan JM, Kim S, Hardikar S, Hollowell D, Lin K, Lu Y, deacetylation during oocyte maturation in mouse. PLoS Genetics 9 Takata Y, Shen JJ et al. 2016 Maternal Setdb1 is required for meiotic e1003377 (doi:10.1371/journal.pgen.1003377) progression and preimplantation development in mouse. PLoS Ma P & Schultz RM 2016 HDAC1 and HDAC2 in mouse oocytes and Genetics 12 e1005970 (doi:10.1371/journal.pgen.1005970) preimplantation embryos: Specificity versus compensation. Cell Death Klose RJ & Yi Z 2007 Regulation of histone methylation by demethylimination and Differentiation 23 1119–1127. (doi:10.1038/cdd.2016.31) and demethylation. Nature Reviews Molecular Cell Biology 8 307–318. Ma T, Keller JA & Yu XC 2011 RNF8-dependent histone ubiquitination (doi:10.1038/nrm2143) during DNA damage response and spermatogenesis. Acta Biochimica et Komander D 2009 The emerging complexity of protein ubiquitination. Biophysica Sinica 43 339–345. (doi:10.1093/abbs/gmr016) Biochemical Society Transactions 37 937–953. (doi:10.1042/ Maleki S & Keeney S 2004 Modifying histones and initiating meiotic BST0370937) recombination. Cell 118 404–406. (doi:10.1016/j.cell.2004.08.008) Komander D, Clague MJ & Urbe S 2009 Breaking the chains: structure and Mallette FA, Mattiroli F, Cui GF, Young LC, Hendzel MJ, Mer G, Sixma TK & function of the deubiquitinases. Nature Reviews Molecular Cell Biology Richard S 2012 RNF8-and RNF168-dependent degradation of KDM4A/ 10 550–563. (doi:10.1038/nrm2731) JMJD2A triggers 53BP1 recruitment to DNA damage sites. EMBO Journal Kouzarides T 2002 Histone methylation in transcriptional control. Current 31 1865–1878. (doi:10.1038/emboj.2012.47) Opinion in Genetics and Development 12 198–209. (doi:10.1016/ Margueron R & Reinberg D 2011 The polycomb complex PRC2 and its S0959-437X(02)00287-3) mark in life. Nature 469 343–349. (doi:10.1038/nature09784) Kuo MH & Allis CD 1998 Roles of histone acetyltransferases Martin C & Zhang Y 2005 The diverse functions of histone lysine and deacetylases in gene regulation. Bioessays 20 615–626. methylation. Nature Reviews Molecular Cell Biology 6 838–849. (doi:10.1002/(SICI)1521-1878(199808)20:8<615::AID- (doi:10.1038/nrm1761) BIES4>3.0.CO;2-H) Masumoto H, Hawke D, Kobayashi R & Verreault A 2005 A role for cell- Kurdistani SK, Tavazoie S & Grunstein M 2004 Mapping global cycle-regulated histone H3 lysine 56 acetylation in the DNA damage histone acetylation patterns to gene expression. Cell 117 721–733. response. Nature 436 294–298. (doi:10.1038/nature03714) (doi:10.1016/j.cell.2004.05.023) Mattiroli F, Vissers JHA, van Dijk WJ, Ikpa P, Citterio E, Vermeulen W, Lan F & Shi Y 2009 Epigenetic regulation: methylation of histone and non- Marteijn JA & Sixma TK 2012 RNF168 ubiquitinates K13-15 on histone proteins. Science in China Series C-Life Sciences 52 311–322. H2A/H2AX to drive DNA damage signaling. Cell 150 1182–1195. (doi:10.1007/s11427-009-0054-z) (doi:10.1016/j.cell.2012.08.005) Lan XJ, Atanassov BS, Li WQ, Zhang Y, Florens L, Mohan RD, Galardy Messiaen S, Guiard J, Aigueperse C, Fliniaux I, Tourpin S, Barroca V, PJ, Washburn MP, Workman JL & Dent SYR 2016 USP44 is an Allemand I, Fouchet P, Livera G & Vernet M 2016 Loss of the histone integral component of N-CoR that contributes to gene repression chaperone ASF1B reduces female reproductive capacity in mice. by deubiquitinating histone H2B. Cell Reports 17 2382–2393. Reproduction 151 477–489. (doi:10.1530/REP-15-0327) (doi:10.1016/j.celrep.2016.10.076) Mietton F, Sengupta AK, Molla A, Picchi G, Barral S, Heliot L, Grange T, Lange J, Pan J, Cole F, Thelen MP, Jasin M & Keeney S 2011 ATM controls Wutz A & Dimitrov S 2009 Weak but uniform enrichment of the histone meiotic double-strand-break formation. Nature 479 237–U117. variant macroH2A1 along the inactive X chromosome. Molecular and (doi:10.1038/nature10508) Cellular Biology 29 150–156. (doi:10.1128/MCB.00997-08) Lange J, Yamada S, Tischfield SE, Pan J, Kim S, Zhu X, Socci ND, Jasin Monteiro FL, Baptista T, Amado F, Vitorino R, Jeronimo C & Helguero LA M & Keeney S 2016 The landscape of mouse meiotic double-strand 2014 Expression and functionality of histone H2A variants in cancer. break formation, processing, and repair. Cell 167 695. (doi:10.1016/j. Oncotarget 5 3428–3443. (doi:10.18632/oncotarget.2007) cell.2016.09.035) Montellier E, Boussouar F, Rousseaux S, Zhang K, Buchou T, Fenaille F, Lawrence KS, Chau T & Engebrecht J 2015 DNA damage response and Shiota H, Debernardi A, Hery P, Curtet S et al. 2013 Chromatin-to- spindle assembly checkpoint function throughout the cell cycle to nucleoprotamine transition is controlled by the histone H2B variant ensure genomic integrity. PLoS Genetics 11 e1005150. (doi:10.1371/ TH2B. Genes and Development 27 1680–1692. (doi:10.1101/ journal.pgen.1005782) gad.220095.113) Lee DY, Hayes JJ, Pruss D & Wolffe AP 1993 A positive role for histone Moore JD, Yazgan O, Ataian Y & Krebs JE 2007 Diverse roles for histone acetylation in transcription factor access to nucleosomal DNA. Cell 72 H2A modifications in DNA damage response pathways in yeast. 73–84. (doi:10.1016/0092-8674(93)90051-Q) Genetics 176 15–25. (doi:10.1534/genetics.106.063792) Lee CS, Lee K, Legube G & Haber JE 2014 Dynamics of yeast histone H2A Myers S, Freeman C, Auton A, Donnelly P & McVean G 2008 A common and H2B phosphorylation in response to a double-strand break. Nature sequence motif associated with recombination hot spots and genome Structural and Molecular Biology 21 103. (doi:10.1038/nsmb.2737) instability in humans. Nature Genetics 40 1124–1129. (doi:10.1038/ Legube G & Trouche D 2003 Regulating histone acetyltransferases and ng.213) deacetylases. EMBO Reports 4 944–947. (doi:10.1038/sj.embor. Nagashima T, Maruyama T, Furuya M, Kajitani T, Uchida H, Masuda H, embor941) Ono M, Arase T, Ozato K & Yoshimura Y 2007 Histone acetylation Li JB, He QQ, Liu YT, Liu SL, Tang S, Li CM, Sun DM, Li XR, Zhou M, and subcellular localization of chromosomal protein BRD4 during Zhu P et al. 2017 Chemical synthesis of K34-ubiquitylated H2B mouse oocyte meiosis and mitosis. Molecular Human Reproduction 13 for nucleosome reconstitution and single-particle cryo-electron 141–148. (doi:10.1093/molehr/gal115) microscopy structural analysis. Chembiochem 18 176–180. Nakada S 2016 Opposing roles of RNF8/RNF168 and deubiquitinating (doi:10.1002/cbic.201600551) enzymes in ubiquitination-dependent DNA double-strand break Lu LY, Wu JX, Ye L, Gavrilina GB, Saunders TL & Yu XC 2010 RNF8- response signaling and DNA-repair pathway choice. Journal of Radiation dependent histone modifications regulate nucleosome removal during Research 57 I33–I40. (doi:10.1093/jrr/rrw027) spermatogenesis. Developmental Cell 18 371–384. (doi:10.1016/j. Nakagawa T, Kajitani T, Togo S, Masuko N, Ohdan H, Hishikawa Y, Koji devcel.2010.01.010) T, Matsuyama T, Ikura T, Muramatsu M et al. 2008 Deubiquitylation of Luense LJ, Wang XS, Schon SB, Weller AH, Shiao EL, Bryant JM, Bartolomei histone H2A activates transcriptional initiation via trans-histone cross- MS, Coutifaris C, Garcia BA & Berger SL 2016 Comprehensive analysis talk with H3K4 di- and trimethylation. Genes and Development 22 of histone post-translational modifications in mouse and human male 37–49. (doi:10.1101/gad.1609708) germ cells. Epigenetics and Chromatin 9. (doi:10.1186/s13072-016- Nguyen AL, Gentilello AS, Balboula AZ, Shrivastava V, Ohring J & Schindler 0072-6) K 2014 Phosphorylation of threonine 3 on histone H3 by haspin kinase Luo MC, Zhou J, Leu NA, Abreu CM, Wang JL, Anguera MC, de Rooij is required for meiosis I in mouse oocytes. Journal of Cell Science 127 DG, Jasin M & Wang PJ 2015 Polycomb protein SCML2 associates with 5066–5078. (doi:10.1242/jcs.158840) www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access R78 L Wang, Z Xu and others

Niikura Y, Kitagawa R, Ogi H, Abdulle R, Pagala V & Kitagawa K 2015 Sarmento OF, Digilio LC, Wang YM, Perlin J, Herr JC, Allis CD & CENP-A K124 ubiquitylation is required for CENP-A deposition at Coonrod SA 2004 Dynamic alterations of specific histone modifications the centromere. Developmental Cell 32 589–603. (doi:10.1016/j. during murine development. Journal of Cell Science 117 4449–4459. devcel.2015.01.024) (doi:10.1242/jcs.01328) Nottke AC, Beese-Sims SE, Pantalena LF, Reinke V, Shi Y & Colaiacovo Shinagawa T, Huynh LM, Takagi T, Tsukamoto D, Tomaru C, Kwak HG, MP 2011 SPR-5 is a histone H3K4 demethylase with a role in meiotic Dohmae N, Noguchi J & Ishii S 2015 Disruption of Th2a and Th2b double-strand break repair. PNAS 108 12805–12810. (doi:10.1073/ genes causes defects in spermatogenesis. Development 142 1287–1292. pnas.1102298108) (doi:10.1242/dev.121830) O’Carroll D, Scherthan H, Peters AHFM, Opravil S, Haynes AR, Shirakata Y, Hiradate Y, Inoue H, Sato E & Tanemura K 2014 Histone H4 Laible G, Rea S, Schmid M, Lebersorger A, Jerratsch M et al. modification during mouse spermatogenesis. Journal of Reproduction 2000 Isolation and characterization of Suv39h2, a second histone and Development 60 383–387. (doi:10.1262/jrd.2014-018) H3 methyltransferase gene that displays testis-specific expression. Simoneau A, Delgoshaie N, Celic I, Dai JB, Abshiru N, Costantino Molecular and Cellular Biology 20 9423–9433. (doi:10.1128/ S, Thibault P, Boeke JD, Verreault A & Wurtele H 2015 Interplay mcb.20.24.9423-9433.2000) between histone H3 lysine 56 deacetylation and chromatin modifiers Ontoso D, Acosta I, van Leeuwen F, Freire R & San-Segundo PA 2013 Dot1- in response to DNA damage. Genetics 200 185–U382. (doi:10.1534/ dependent histone H3K79 methylation promotes activation of the Mek1 genetics.115.175919) meiotic checkpoint effector kinase by regulating the Hop1 adaptor. PLoS Smith CA, Roeszler KN, Bowles J, Koopman P & Sinclair AH 2008 Onset of Genetics 9 e1003262. (doi:10.1371/journal.pgen.1003262) meiosis in the chicken embryo; evidence of a role for retinoic acid. BMC Ontoso D, Kauppi L, Keeney S & San-Segundo PA 2014 Dynamics of Developmental Biology 8 1–19. (doi:10.1186/1471-213X-8-85) DOT1L localization and H3K79 methylation during meiotic prophase Soboleva TA, Nekrasov M, Pahwa A, Williams R, Huttley GA & Tremethick I in mouse spermatocytes. Chromosoma 123 147–164. (doi:10.1007/ DJ 2012 A unique H2A histone variant occupies the transcriptional s00412-013-0438-5) start site of active genes. Nature Structural and Molecular Biology 19 Park CS, Rehrauer H & Mansuy IM 2013 Genome-wide analysis of H4K5 U25–U37. (doi:10.1038/nsmb.2161) acetylation associated with fear memory in mice. BMC Genomics 14. Sommermeyer V, Beneut C, Chaplais E, Serrentino ME & Borde V (doi:10.1186/1471-2164-14-539) 2013 Spp1, a member of the Set1 complex, promotes meiotic DSB Parvanov ED, Petkov PM & Paigen K 2010 Prdm9 controls activation formation in promoters by tethering histone H3K4 methylation sites of mammalian recombination hotspots. Science 327 835–835. to chromosome axes. Molecular Cell 49 43–54. (doi:10.1016/j. (doi:10.1126/science.1181495) molcel.2012.11.008) Patel A, Horton JR, Wilson GG, Zhang X & Cheng XD 2016 Structural Song N, Liu J, An SC, Nishino T, Hishikawa Y & Koji T 2011 basis for human PRDM9 action at recombination hot spots. Genes and Immunohistochemical analysis of histone H3 modifications in germ Development 30 257–265. (doi:10.1101/gad.274928.115) cells during mouse spermatogenesis. Acta Histochemica et Cytochemica Paull TT, Rogakou EP, Yamazaki V, Kirchgessner CU, Gellert M & Bonner 44 183–190. (doi:10.1267/ahc.11027) WM 2000 A critical role for histone H2AX in recruitment of repair Stiff T, O’Driscoll M, Rief N, Iwabuchi K, Lobrich M & Jeggo PA 2004 factors to nuclear foci after DNA damage. Current Biology 10 886–895. ATM and DNA-PK function redundantly to phosphorylate H2AX (doi:10.1016/S0960-9822(00)00610-2) after exposure to ionizing radiation. Cancer Research 64 2390–2396. Perez-Cadahia B, Drobic B, Khan P, Shivashankar CC & Davie JR 2010 (doi:10.1158/0008-5472.CAN-03-3207) Current understanding and importance of histone phosphorylation in Strom L, Karlsson C, Lindroos HB, Wedahl S, Katou Y, Shirahige K & regulating chromatin biology. Current Opinion in Drug Discovery and Sjogren C 2007 Postreplicative formation of cohesion is required for Development 13 613–622. (doi:10.4161/epi.21975) repair and induced by a single DNA break. Science 317 242–245. Peters AHFM, O’Carroll D, Scherthan H, Mechtler K, Sauer S, Schofer (doi:10.1126/science.1140649) C, Weipoltshammer K, Pagani M, Lachner M, Kohlmaier A et al. 2001 Stucki M, Clapperton JA, Mohammad D, Yaffe MB, Smerdon SJ & Jackson Loss of the Suv39h histone methyltransferases impairs mammalian SP 2005 MDC1 directly binds phosphorylated histone H2AX to regulate heterochromatin and genome stability. Cell 107 323–337. (doi:10.1016/ cellular responses to DNA double-strand breaks. Cell 123 1213–1226. S0092-8674(01)00542-6) (doi:10.1016/j.cell.2005.09.038) Powers NR, Parvanov ED, Baker CL, Walker M, Petkov PM & Paigen K Sun F, Fujiwara Y, Reinholdt LG, Hu JJ, Saxl RL, Baker CL, Petkov PM, 2016 The meiotic recombination activator PRDM9 trimethylates both Paigen K & Handel MA 2015 Nuclear localization of PRDM9 and its H3K36 and H3K4 at recombination hotspots in vivo. PLoS Genetics 12. role in meiotic chromatin modifications and homologous synapsis. Pratto F, Brick K, Khil P, Smagulova F, Petukhova GV & RD Camerini-Otero Chromosoma 124 397–415. (doi:10.1007/s00412-015-0511-3) 2014 Recombination initiation maps of individual human genomes. Tachibana M, Sugimoto K, Fukushima T & Shinkai Y 2001 SET domain- Science 346 826. containing protein, G9a, is a novel lysine-preferring mammalian Rangasamy D, Greaves I & Tremethick DJ 2004 RNA interference histone methyltransferase with hyperactivity and specific selectivity to demonstrates a novel role for H2A.Z in chromosome segregation. Nature lysines 9 and 27 of histone H3. Journal of Biological Chemistry 276 Structural and Molecular Biology 11 650–655. (doi:10.1038/nsmb786) 25309–25317. (doi:10.1074/jbc.M101914200) Recht J, Tsubota T, Tansy JC, Diaz RL, Berger JM, Zhang X, Garcia BA, Tachibana M, Nozaki M, Takeda N & Shinkai Y 2007 Functional dynamics Shabanowitzn J, Burlingame AL, Hunt DF et al. 2006 Histone chaperone of H3K9 methylation during meiotic prophase progression. EMBO Asf1 is required for histone H3 lysine 56 acetylation, a modification Journal 26 3346–3359. (doi:10.1038/sj.emboj.7601767) associated with S phase in mitosis and meiosis. PNAS 103 6988–6993. Takada Y, Naruse C, Costa Y, Shirakawa T, Tachibana M, Sharif J, Kezuka- (doi:10.1073/pnas.0601676103) Shiotani F, Kakiuchi D, Masumoto H, Shinkai Y et al. 2011 HP1 Robzyk K, Recht L & Osley MA 2000 Rad6-dependent ubiquitination gamma links histone methylation marks to meiotic synapsis in mice. of histone H2B in yeast. Science 287 501–504. (doi:10.1126/ Development 138 4207–4217. (doi:10.1242/dev.064444) science.287.5452.501) Tang MCW, Jacobs SA, Mattiske DM, Soh YM, Graham AN, Tran A, Lim SL, Rogakou EP, Pilch DR, Orr AH, Ivanova VS & Bonner WM 1998 DNA Hudson DF, Kalitsis P, O’Bryan MK et al. 2015 Contribution of the two double-stranded breaks induce histone H2AX phosphorylation on serine genes encoding histone variant H3.3 to viability and fertility in mice. 139. Journal of Biological Chemistry 273 5858–5868. (doi:10.1074/ PLoS Genetics 11. (doi:10.1371/journal.pgen.1005754) jbc.273.10.5858) Thompson LL, Guppy BJ, Sawchuk L, Davie JR & McManus KJ 2013 Sakai A, Schwartz BE, Goldstein S & Ahmad K 2009 Transcriptional and Regulation of chromatin structure via histone post-translational developmental functions of the H3.3 histone variant in drosophila. modification and the link to carcinogenesis. Cancer and Metastasis Current Biology 19 1816–1820. (doi:10.1016/j.cub.2009.09.021) Reviews 32 363–376. (doi:10.1007/s10555-013-9434-8) Santos-Rosa H, Schneider R, Bannister AJ, Sherriff J, Bernstein BE, Emre Thorslund T, Ripplinger A, Hoffmann S, Wild T, Uckelmann M, Villumsen NCT, Schreiber SL, Mellor J & Kouzarides T 2002 Active genes are tri- B, Narita T, Sixma TK, Choudhary C, Bekker-Jensen S et al. 2015 Histone methylated at K4 of histone H3. Nature 419 407–411. (doi:10.1038/ H1 couples initiation and amplification of ubiquitin signalling after DNA nature01080) damage. Nature 527 389. (doi:10.1038/nature15401)

Reproduction (2017) 154 R65–R79 www.reproduction-online.org

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access The histone codes for meiosis R79

Tjeertes JV, Miller KM & Jackson SP 2009 Screen for DNA-damage- of the H3K4 methyltransferase activity of PRDM9. Cell Reports 5 13–20. responsive histone modifications identifies H3K9Ac and H3K56Ac (doi:10.1016/j.celrep.2013.08.035) in human cells. EMBO Journal 28 1878–1889. (doi:10.1038/ Wu BJ, Dong FL, Ma XS, Wang XG, Lin F & Liu HL 2014 Localization emboj.2009.119) and expression of histone H2A variants during mouse oogenesis and Turner JMA, Mahadevaiah SK, Fernandez-Capetillo O, Nussenzweig A, preimplantation embryo development. Genetics and Molecular Research Xu XL, Deng CX & Burgoyne PS 2005 Silencing of unsynapsed meiotic 13 5929–5939. (doi:10.4238/2014.August.7.8) chromosomes in the mouse. Nature Genetics 37 41–47. (doi:10.1038/ Xiong K, Wu W, Wang XG, Ma XS, Chen J & Liu HL 2013 Mouse oocyte ng1484) meiosis is disturbed by knockdown of Suv4-20h. Reproduction Fertility Ueda J, Harada A, Urahama T, Machida S, Maehara K, Hada M, Makino and Development 25 503–510. (doi:10.1071/RD12116) Y, Nogami J, Horikoshi N, Osakabe A et al. 2017 Testis-specific histone Xu Z, Song Z, Li G, Tu H, Liu W, Liu Y, Wang P, Wang Y, Cui X, Liu C variant H3t gene is essential for entry into spermatogenesis. Cell Reports et al. 2016 H2B ubiquitination regulates meiotic recombination by 18 593–600. (doi:10.1016/j.celrep.2016.12.065) promoting chromatin relaxation. Nucleic Acids Research 44 9681–9697. Urahama T, Harada A, Maehara K, Horikoshi N, Sato K, Sato Y, Shiraishi K, (doi:10.1093/nar/gkw652) Sugino N, Osakabe A, Tachiwana H et al. 2016 Histone H3.5 forms an Yamada S, Ohta K & Yamada T 2013 Acetylated Histone H3K9 is associated unstable nucleosome and accumulates around transcription start sites in with meiotic recombination hotspots, and plays a role in recombination human testis. Epigenetics and Chromatin 9 1–16. (doi:10.1186/s13072- redundantly with other factors including the H3K4 methylase Set1 in 016-0051-y) fission yeast. Nucleic Acids Research 41 3504–3517. (doi:10.1093/nar/ van den Berg IM, Eleveld C, van der Hoeven M, Birnie E, Steegers EAP, gkt049) Galjaard RJ, Laven JSE & van Doorninck JH 2011 Defective deacetylation Yamamoto A, West RR, McIntosh JR & Hiraoka Y 1999 A cytoplasmic of histone 4 K12 in human oocytes is associated with advanced dynein heavy chain is required for oscillatory nuclear movement of maternal age and chromosome misalignment. Human Reproduction 26 meiotic prophase and efficient meiotic recombination in fission yeast. 1181–1190. (doi:10.1093/humrep/der030) Journal of Cell Biology 145 1233–1249. (doi:10.1083/jcb.145.6.1233) van der Heijden GW, Derijck AAHA, Posfai E, Giele M, Pelczar P, Ramos Yamashita K, Shinohara M & Shinohara A 2004 Rad6-Bre1-mediated L, Wansink DG, van der Vlag J, Peters AHFM & de Boer P 2007 histone H2B ubiquitylation modulates the formation of double- Chromosome-wide nucleosome replacement and H3.3 incorporation strand breaks during meiosis. PNAS 101 11380–11385. (doi:10.1073/ during mammalian meiotic sex chromosome inactivation. Nature pnas.0400078101) Genetics 39 251–258. (doi:10.1038/ng1949) Yang Y, Yamada T, Hill KK, Hemberg M, Reddy NC, Cho HY, Guthrie AN, Vaskova EA, Pavlova SV, Shevchenko AI & Zakian SM 2010 Meiotic Oldenborg A, Heiney SA, Ohmae S et al. 2016 Chromatin remodeling inactivation of sex chromosomes in mammals. Russian Journal of inactivates activity genes and regulates neural coding. Science 353 Genetics 46 385–393. (doi:10.1134/S1022795410040010) 300–305. (doi:10.1126/science.aad4225) Virant-Klun I, Knez K, Tomazevic T & Skutella T 2013 Gene expression Yoon SW, Lee MS, Xaver M, Zhang LR, Hong SG, Kong YJ, Cho HR, profiling of human oocytes developed and matured in vivo or in vitro. Kleckner N & Kim KP 2016 Meiotic prophase roles of Rec8 in crossover Biomed Research International 1–21. (doi:10.1155/2013/879489) recombination and chromosome structure. Nucleic Acids Research 44 Wang XG, Gao WL, Ma XS, Wang XN, Song CL, Huang XJ & Liu HL 2014 9296–9314. (doi:10.1093/nar/gkw682) Dot1L mediated histone H3 lysine79 methylation is essential to meiosis Yuen BTK, Bush KM, Barrilleaux BL, Cotterman R & Knoepfler PS 2014 progression in mouse oocytes. Neuroendocrinology Letters 35 523–530. Histone H3.3 regulates dynamic chromatin states during spermatogenesis. Wang Q, Wei HJ, Du J, Cao Y, Zhang NN, Liu XY, Liu XY, Chen DD & Ma Development 141 3483–3494. (doi:10.1242/dev.106450) W 2016 H3 Thr3 phosphorylation is crucial for meiotic resumption and Zentner GE & Henikoff S 2013 Regulation of nucleosome dynamics by anaphase onset in oocyte meiosis. Cell Cycle 15 213–224. (doi:10.1080/ histone modifications.Nature Structural and Molecular Biology 20 15384101.2015.1121330) 259–266. (doi:10.1038/nsmb.2470) Watanabe Y 2012 Geometry and force behind kinetochore orientation: Zhang Y & Reinberg D 2001 Transcription regulation by histone lessons from meiosis. Nature Reviews Molecular Cell Biology 13 methylation: interplay between different covalent modifications of the 370–382. (doi:10.1038/nrm3349) core histone tails. Genes and Development 15 2343–2360. (doi:10.1101/ Watanabe S, Radman-Livaja M, Rando OJ & Peterson CL 2013 A histone gad.927301) acetylation switch regulates H2A.Z deposition by the SWR-C remodeling Zhang F & Yu X 2011 WAC, a functional partner of RNF20/40, regulates enzyme. Science 340 195–199. (doi:10.1126/science.1229758) histone H2B ubiquitination and gene transcription. Molecular Cell 41 Wei Y, Mizzen CA, Cook RG, Gorovsky MA & Allis CD 1998 Phosphorylation 384–397. (doi:10.1016/j.molcel.2011.01.024) of histone H3 at serine 10 is correlated with chromosome condensation Zhong J, Li X, Cai W, Wang Y, Dong S, Yang J, Zhang JA, Wu N, Li Y, Mao during mitosis and meiosis in Tetrahymena. PNAS 95 7480–7484. F et al. 2016 TET1 modulates H4K16 acetylation by controlling auto- (doi:10.1073/pnas.95.13.7480) acetylation of hMOF to affect gene regulation and DNA repair function. Wei Y, Yu LL, Bowen J, Gorovsky MA & Allis CD 1999 Phosphorylation Nucleic Acids Research 45 672–684. (doi:10.1093/nar/gkw919). of histone H3 is required for proper chromosome condensation and Zickler D & Kleckner N 1999 Meiotic chromosomes: Integrating structure segregation. Cell 97 99–109. (doi:10.1016/S0092-8674(00)80718-7) and function. Annual Review of Genetics 33 603–754. (doi:10.1146/ Wisniewski JR, Zougman A, Kruger S & Mann M 2007 Mass spectrometric annurev.genet.33.1.603) mapping of linker histone H1 variants reveals multiple acetylations, methylations, and phosphorylation as well as differences between cell culture and tissue. Molecular and Cellular Proteomics 6 72–87. (doi:10.1074/mcp.M600255-MCP200) Wolfe SA, vanWert JM & Grimes SR 2008 Transcription factor RFX4 binding to the testis-specific histone H1t promoter in spermatocytes may be important for regulation of H1t gene transcription during Received 12 March 2017 spermatogenesis. Journal of Cellular Biochemistry 105 61–69. First decision 29 March 2017 (doi:10.1002/jcb.21793) Wu H, Mathioudakis N, Diagouraga B, Dong AP, Dombrovski L, Baudat F, Revised manuscript received 10 June 2017 Cusack S, de Massy B & Kadlec J 2013 Molecular basis for the regulation Accepted 19 June 2017

www.reproduction-online.org Reproduction (2017) 154 R65–R79

Downloaded from Bioscientifica.com at 10/03/2021 12:59:21AM via free access