Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 1.
Published in final edited form as: Dev Biol. 2021 Feb 18;473:105–118. doi: 10.1016/j.ydbio.2021.02.003

Epigenetic Regulation of Drosophila Germline Stem Cell Maintenance and Differentiation

Velinda Vidaurre 1, Xin Chen 1,2
PMCID: PMC7992187  NIHMSID: NIHMS1677877  PMID: 33610541

Abstract

Gametogenesis is one of the most extreme cellular differentiation processes that takes place in Drosophila male and female germlines. This process begins at the germline stem cell, which undergoes asymmetric cell division (ACD) to produce a self-renewed daughter that preserves its stemness and a differentiating daughter cell that undergoes epigenetic and genomic changes to eventually produce haploid gametes. Research in molecular genetics and cellular biology are beginning to take advantage of the continually advancing genomic tools to understand: (1) how germ cells are able to maintain their identity throughout the adult reproductive lifetime, and (2) undergo differentiation in a balanced manner. In this review, we focus on the epigenetic mechanisms that address these two questions through their regulation of germline-soma communication to ensure germline stem cell identity and activity.

Keywords: Drosophila, germline stem cells, somatic gonadal cells, signaling pathways, transcription, chromatin regulator, epigenetics

1. Introduction

Germ cells have the unique ability to differentiate into gametes that will give rise to a new organism upon fertilization. Drosophila gametogenesis is intensively studied to understand cellular differentiation in adult stem cell lineages. This is due to the linear organization of germ cells according to their differentiation status in the gonads, which is conducive for visualization (Spradling et al., 2011). The complex cellular differentiation processes that produce gametes originate from the germline stem cells (GSCs) in both sexes (Fuller and Spradling, 2007; Matunis et al., 2012; Spradling et al., 2011). GSCs must self-renew to retain their stemness and produce a differentiating daughter cell (Figure 1) (Chen and McKearin, 2003a; Davies and Fuller, 2008; Kiger et al., 2000; Yamashita and Fuller, 2005). However, both female and male GSCs are capable of symmetric cell division (SCD) to increase GSC number, albeit at low frequency under physiological conditions (Salzmann et al., 2013; Sheng and Matunis, 2011; Xie and Spradling, 2000). In addition, partially differentiated mitotic germ cells can return to the niche and become GSC-like cells under particular conditions, such as pathological and stress-induced conditions, or during aging, a process called dedifferentiation (Brawley and Matunis, 2004; Cheng et al., 2008; Herrera and Bach, 2018; Kai and Spradling, 2004; Lim et al., 2015; Liu et al., 2015; Sheng et al., 2009). The ACD of GSCs is controlled by both cell-autonomous and non-cell-autonomous mechanisms [reviewed in (Gleason et al., 2018; Kahney et al., 2019)]. The somatic gonadal cells, such as the cyst stem cells (CySCs), cyst cells, and hub cells in testes (Figure 1A), and the cap cells, escort cells and follicle cells in ovaries (Figure 1B), contribute to the control of proper germ cell maintenance and differentiation through signaling pathways in a non-cell autonomous manner (Decotto and Spradling, 2005; Gonczy and DiNardo, 1996; Hardy et al., 1979; Nystul and Spradling, 2007; Xie and Spradling, 2000).

Figure 1: Niches, early-stage germline and somatic gonadal cells in Drosophila male and female gonads.

Figure 1:

(A) The Drosophila testis is a pair to two coiled tubes, each containing a single stem cell niche at the apical tip. The stem cell niche is called the hub (dark blue), which is a cluster of 10–12 densely packed somatic cells. The germline stem cells (GSCs) (dark green) and the cyst stem cells (CySCs) (dark pink) are radially positioned around the hub. There are 10–15 GSCs arranged around the hub with two CySCs enveloping each GSC. The GSCs undergo an asymmetric division to produce a self-renewed daughter GSC and a gonialblast (GB) (light green). The GB is displaced from the hub and undergoes 4 rounds of miotic divisions with incomplete cytokinesis to create cysts of interconnected germ cells. The differentiating germline cysts continue to be encapsulated by two post mitotic cyst cells (light pink). The 16-spermatogonial cell cyst then undergoes meiosis as spermatocytes to produce 64 spermatids. (B) The Drosophila ovary consist of 16–20 tubular structures called ovarioles. An ovariole consists of a germarium at the apical tip and a linear progression of differentiating egg chambers that produce eggs, located at the base. The germarium is comprised of the GSC niche and the proliferating germ cells. The GSC niche includes a stack of 8–10 post-mitotic somatic cells called the terminal filament (dark blue) and 5–7 epithelial cells called the cap cells (light blue) that directly interact with the 2–3 GSCs (dark green). Female GSCs divide asymmetrically to produce a self-renewed GSC that stays in contact with the cap cells and a posteriorly displaced daughter cell that leaves the niche and differentiates into a cystoblast (CB) (light green). The CB then undergoes 4 rounds of synchronous mitotic cell divisions with incomplete cytokinesis to create 16 interconnected cystocytes. Interspersed between the early germline cells are 4 −6 escort cells (pink) that surrounded the GSCs and the dividing CBs. The interconnected germ cell cysts then begin to associate with somatic follicle cells. Follicle cells (orange) are derived from two follicle stem cells (FSCs) (red) that are located between the escort cell and follicle cell transition zone. Once the 16-cell germ cyst is surrounded by follicle cells, it becomes an egg chamber, buds off from the germarium, and continues to mature and undergoes meiosis to develop into an oocyte and 15 interconnected nurse cells. Adapted from Gleason et al., Genetics 2018.

In recent years, broad epigenetic mechanisms that alter gene expression due to heritable chromatin structure change, but no primary DNA sequence change, have been shown to regulate germ cell maintenance and differentiation (Buszczak et al., 2009; Casper et al., 2011; Davies et al., 2013; Maines et al., 2007; Xi and Xie, 2005). Such epigenetic mechanisms include the modification of DNA-associated proteins or RNAs which can create chromatin structural changes or recruit transcription regulators. These actions subsequently allow gene expression changes, such as repression of non-lineage genes or activation of germline differentiation genes.

The basic structure of chromatin is comprised of DNA and histones. A single nucleosome consists of 147 bp of DNA wrapping around a canonical histone octamer, composed of an H3-H4 tetramer and two H2B and H2A dimers. The main epigenetic mechanisms known to control germline fate and regulate germ cell differentiation are (1) DNA methylation, (2) the incorporation of histone and (3) histone variants, (4) the post-translational modifications of histones by different histone modifying enzymes, (5) the repositioning of nucleosomes by chromatin remodelers, and (6) the regulation of non-coding RNA-mediated chromatin landscape by piRNAs and miRNAs. This review focuses on recent discoveries of epigenetic mechanisms in maintaining GSCs and controlling differentiation of early-stage germline cells using Drosophila as the model organism.

2. Epigenetic Mechanisms Regulating GSC Maintenance and Self-Renewal

2.1. DNA modifications

DNA methylation is an epigenetic mechanism conserved from plants to mammals. It functions through the covalent modification of cytosine and adenine residues (Luo et al., 2015). Methylation at the fifth position of the cytosine base (5mC) is established and maintained by a conserved enzyme family called DNA methyltransferases (DNMTs). 5mC modifications normally at the promoter region silence the transcriptional activity of target genes, as well as repressing transcription at transposable elements and other repetitive sequences (Suzuki and Bird, 2008; Wu and Zhang, 2014).

There are two main DNMT families, normally classified by their distinct functions to either regulate de novo methylation (DNMT3) or maintain existing methylation (DNMT1) (Law and Jacobsen, 2010). Although DNMT2 shows structural similarities to DNMT1 and DNMT3 family proteins, its function and substrate remain unclear. For example, DNMT2 family proteins show transposon silencing in the Drosophila early embryo cells and weak methyltransferase activity to methylate ribonucleic acids (tRNAs) (Goll et al., 2006; Phalke et al., 2009; Schaefer et al., 2010). Drosophila belongs to the “DNMT2 only” species by retaining the DNMT2 homolog Mt2 but loses both DNMT1 and DNMT3 (Raddatz et al., 2013). In Drosophila, the level of 5mC has been shown to be exceptionally low compared to other organisms, with only 0.034% 5mC out of total cytosines in the adult fly genome. Additionally, this methylation appears to be independent of DNMT2 activity, further indicating that DNMT2 may not act as a DNA methylase in Drosophila (Capuano et al., 2014; Raddatz et al., 2013; Takayama et al., 2014). DNMT2 is involved in non-random sister chromatid inheritance during ACD of Drosophila male GSCs (Yadlapalli and Yamashita, 2013). Therefore, the scarcity of 5mC in the Drosophila genome and the unclear molecular function of DNMT2 suggest that other epigenetic mechanisms may play more important roles in the Drosophila germline.

2.2. Canonical histones

Due to the lack of DNA methylation in the fly genome, histones are likely the main source of epigenetic information in Drosophila. Histones closely interact with DNA and carry extensive post-translational modifications (PTMs), which can recruit factors, such as histone modifying enzymes and chromatin remodelers, to either activate or repress local gene expression (Allfrey et al., 1964; Bannister and Kouzarides, 2011; Boros, 2012; Dawson et al., 2009). In the Drosophila male germline, pre-existing (old) histone H3 is found to segregate to the GSC, while the newly synthesized (new) histone H3 is preferentially inherited by the differentiating daughter cell (Tran et al., 2012). This asymmetric inheritance pattern is specific to histones H3 and H4, which have the most known PTMs that regulate gene expression and whose incorporation is DNA replication-dependent. Histone variants such as histone H3.3, which are incorporated independently of replication, do not show this asymmetric inheritance pattern (Tran et al., 2012). Therefore, it was hypothesized and later shown that this old versus new H3 and H4 asymmetry between sister chromatids is established during DNA replication (Wooten et al., 2019). Additionally, to achieve the asymmetric segregation pattern, the mitotic machinery ensures that old histone H3-enriched sister chromatids are segregated to the GSC while new histone H3-enriched sister chromatids are segregated to differentiating daughter cell. Differential phosphorylation at Threonine 3 of H3 (H3T3ph) is shown in the mitotic GSCs, where old H3 is more enriched with H3T3ph than new H3. Mis-regulation of this phosphorylation using either a dominant negative H3T3A mutant or a phosphomimetic H3T3D mutant leads to randomized segregation of old- versus new-histone enriched sister chromatids, as well as cellular defects such as stem cell loss and progenitor germline tumor (Xie et al., 2015). Recently, a GSC-specific ‘mitotic drive’ phenomenon has been identified. Sister chromatids have different amounts of the histone variant that specifies the centromere region, the CENP-A homolog Centromere IDentifier (CID). This asymmetric distribution of CID between sister centromeres coordinates with the dynamic activities of the spindle microtubules and temporal breakdown of the nuclear envelop, first at the proximal side toward the stem cell niche followed by the distal side. Together, the spatiotemporally regulated events ensure preferential recognition and attachment of the asymmetric sister centromeres by dynamic microtubes. Loss of either the asymmetric CID distribution between sister chromatids, by compromising the activity of the CID-specific histone chaperone CAL1, or temporally asymmetric microtubule activities using microtubule depolymerization drug Nocodazole, causes randomized sister chromatid segregation (Ranjan et al., 2019). Together, spatially asymmetric CID and H3T3ph between sister chromatids and the temporally asymmetric microtubule activity provide the cellular basis for asymmetric histone inheritance during ACD of Drosophila male GSCs. Therefore, in light of the phenotypes caused by disrupting asymmetric histone inheritance, this process likely maintains GSC identity by retaining old histone while allow differentiation of the daughter germ cell by resetting its chromatin structure. However, it remains unclear how histones are inherited during SCD of GSCs or during spermatogonial dedifferentiation. As SCD normally occurs at a low frequency, it is likely that old and new H3/H4 as well as the H3 variant CID display more symmetric inheritance patterns during SCD, which are also detectable at a relatively low frequency under normal conditions (Ranjan et al., 2019; Tran et al., 2012; Wooten et al., 2019). In order to understand histone inheritance during dedifferentiation, certain technical caveats need to be solved. For example, spermatogonial dedifferentiation under physiological conditions during aging often results in GSC-like cells with misoriented centrosome (Cheng et al., 2008), which tend to be arrested and fail to proceed with active mitosis. Even though pathological conditions or genetic depletion of bona fide GSCs could induce more efficient dedifferentiation, whether and how dedifferentiated GSC-like cells undergo a balanced self-renewal versus differentiation is unclear. Combination of these treatments with precise lineage tracing to locate these GSC-like cells at single cell resolution, along with histone labeling strategies and live cell imaging tools, will illuminate these intriguing questions.

Canonical histone H1, the linker histone, regulates proper female GSC self-renewal. In the Drosophila ovary, the Bone Morphogenic Protein (BMP) ligands Dpp and Gbb are expressed from the niche and regulate GSC identity and activity. These ligands activate BMP signaling mediated by phosphorylated Mothers against Dpp (pMad), which subsequently activates Daughters against Dpp (Dad) expression in order to repress the transcription of the main differentiation factor called Bag of marbles (Bam) in GSCs (Figure 2B) (Casanueva and Ferguson, 2004; Chen and McKearin, 2003a; Chen and McKearin, 2003b; Eliazer and Buszczak, 2011; Kawase et al., 2004; McKearin and Ohlstein, 1995; McKearin and Spradling, 1990; Song et al., 2004; Xie and Spradling, 1998). Knockdown of histone H1 in the early-stage germline results in the loss of GSCs and premature differentiation due to precocious Bam expression. Interestingly, loss of H1 also results in an increase of transcription activating histone modification H4K16ac in the GSCs. This is similar to the effect caused by overexpression of MOF, the acetyltransferase for H4K16 (Akhtar and Becker, 2000). Collectively, these results suggest that H1 and MOF act antagonistically to maintain H4K16ac at a level that prevents ectopic expression of differentiation genes in Drosophila female GSCs (Sun et al., 2015).

Figure 2: Signaling pathways regulating GSC self-renewal and differentiation at niches in Drosophila male and female gonads.

Figure 2:

(A) In the Drosophila testis, Hub cells secrete the ligand Unpaired (Upd), which activates Janus Kinase and Signal Transducer and Activator of Transcription (JAK-STAT) signaling in the GSCs and CySCs. In the CySCs, JAK-STAT activation is sufficient for stem cell self-renewal, while the GSCs are maintained by signaling from both the hub cells and CySCs. Activated pStat92E in the CySCs activates expression of zn finger homeodomain 1 (zfh1) and chronologically inappropriate morphogenesis (chinmo), which are important for CySC self-renewal and identity. Activation of Zfh1 turns on expression of the BMP ligands glass bottom boat (gbb) and decapentaplegic (dpp) in the CySCs, which activate BMP signaling in the GSCs, important for GSC self-renewal and maintenance. Activation of BMP signaling in the GSCs by the hub cells and CySCs also inhibits expression of bag of marbles (bam) to prevent differentiation. Activation of pStat92E in the GSCs promotes GSC adhesion to the hub. Activation of pStat92E in the CySCs also activates expression of suppressor of cytokine signaling at 36E (socs36E), a negative regulator of the JAK-STAT pathway. In addition, EGF from the GSCs to the CySCs regulates their abilities to encapsulate germ cells and GSC divisions. (B) In the Drosophila ovary, JAK-STAT acts in the cap cells to activate BMP ligand, dpp. Dpp/Gbb activates BMP signaling in the GSCs, resulting in phosphorylation of Mothers against dpp (pMad) and activated transcription of dads against dpp (dad). pMad and its partner Medea translocate to nucleus and repress transcription of bam, inhibiting GSC differentiation (Hudson et al., 1998). This repression is relieved once the GSC daughter cell leaves the niche. Brain tumor (Brat) reduces BMP responsiveness in the CB. EGF from the CB activates EGF signaling in the neighboring escort cells, where it represses the transcription of dally (Liu et al., 2010). This ensures that BMP ligands Dpp/Gbb are restricted to the cap cells and BMP activation only occurs in the GSCs. Adapted from Matunis et al., Spermatogenesis 2012.

2.3. Histone variants

In addition to canonical histones, histone variants also regulate many cellular processes including transcriptional activity, heterochromatin formation, DNA repair and cell identity (Swaminathan et al., 2005; Talbert and Henikoff, 2017; Tanabe et al., 2008). In Drosophila, the only H2A variant is H2Av, which has the functions of both mammalian H2A variants in DNA damage repair and transcriptional regulation (Baldi and Becker, 2013; van Daal et al., 1988). In the Drosophila male germline, H2Av is required for both GSC and CySC maintenance, where knockdown of H2Av via RNAi or compromising function using H2Av mutants results in declined GSCs and CySCs over time. In the testicular niche, the hub cells secrete Unpaired (Upd), which activates JAK-STAT signaling with the downstream phosphorylation of the Stat92E transcription factor (pStat92E) in both GSCs and CySCs (Harrison et al., 1998; Kiger et al., 2001; Tulina and Matunis, 2001). In CySCs, pStat92E activates CySC and GSC self-renewing factors, Zfh1 and Chinmo (Flaherty et al., 2010; Leatherman and Dinardo, 2008). In GSCs, pStat92E ensures the expression of adhesion molecules for their attachment to the hub cells (Figure 2A) (Herrera and Bach, 2019; Leatherman and Dinardo, 2010). However, the role of H2Av in GSC self-renewal seems to be independent of the JAK-STAT signaling pathway, as the loss of H2Av does not affect pStat92E levels and mutation of the stat92E gene does not affect H2Av expression, either. Furthermore, differentiation of both progenitor germ cells and cyst cells appear to be unaffected, suggesting that H2Av regulates solely GSC self -renewal by an unknown mechanism, independent of the JAK-STAT pathway (Morillo Prado et al., 2013).

2.4. Histone-modifying enzymes

Histone modifications play profound roles in regulating chromatin landscape and function (Allfrey et al., 1964; Bannister and Kouzarides, 2011). For example, the addition of an acetyl group to the lysine side chains by acetyltransferases neutralizes the positive charge of the lysine, weakening the interaction between the histones and DNA (Hodawadekar and Marmorstein, 2007). Modifications can recruit remodeling enzymes that reposition nucleosomes to regulate many processes such as DNA replication, repair and recombination (Bartke et al., 2010; Vermeulen et al., 2010). The histone modifications are catalyzed by specific enzymes that can generate, or “write”, these covalent changes. Additional factors can recognize, or “read”, these modifications and recruit other factors to lay down additional modifications that either promote or repress transcription. Another class of enzymes can remove, or “erase”, histone modifications. The histone modifying enzymes are often encoded by a single gene in Drosophila, making it an ideal model organism to study the roles of histone modifications and their modifiers without complications caused by redundant genes (Bannister and Kouzarides, 2011; Boros, 2012; Hennig and Weyrich, 2013).

Polycomb Group (PcG) and Trithorax Group (TrxG):

Both PcG and TrxG protein complexes consist of histone modifying enzymes, as well as additional components that assist their functions through chromatin compaction, inhibition of chromatin remodeling, etc. (Kassis et al., 2017). Several PcG complexes have been characterized, including Polycomb Repressive Complex 1(PRC1) and Polycomb Repressive Complex 2 (PRC2). In Drosophila, PRC1 consists of Polycomb (Pc), Polyhomeotic (Ph), Posterior sex combs (Psc) or Suppressor of Zestes 2 [Su(z)2], and Sex combs extra (Sce/dRing) (Francis et al., 2001). Within PRC1, Pc has a chromodomain that allows it to recognize and bind to the H3K27me3 modification; Ph has a SAM domain important for the PRC1-mediated repression of target genes; Psc and Su(z) 2 have a common C-terminal region that mediates chromatin compaction or remodeling; Sce or dRing has the H2A ubiquitin ligase activity to generate the H2AK118ub modification, which promotes PRC2 activity in generating more H3K27me3 (Francis et al., 2004; Gambetta and Muller, 2014; Kalb et al., 2014; King et al., 2005; Lo et al., 2009; Messmer et al., 1992; Wang et al., 2004). For PRC2, Enhancer of Zestes [E(z)] is the core histone methyl-transferase that generates the H3K27me3 mark. Other PRC2 components include Extra Sex combs (Esc), Suppressor of Zestes 12 [Su(z) 12], and Caf1-55 (Table 1) (Czermin et al., 2002; Muller et al., 2002). The main function of Esc and Su(z)12 is to stimulate the enzymatic activity of E(z), but the biochemical role of Caf1-55 remains unclear (Anderson et al., 2011; Cao et al., 2002; Czermin et al., 2002; Kassis et al., 2017; Ketel et al., 2005; Margueron et al., 2009; Muller et al., 2002; Tie et al., 2007; Wen et al., 2012; Xu et al., 2010).

Table 1:

Histone PTMs and Histone Modifying Enzymes in the Drosophila Male and Female Gonads

PTM Writer Eraser Reader/additional components Function Germline loss-of-function phenotypes Gonad studied & cell types present Phenotype due to loss of Enzymatic activity
H3K27me3 E(z) dUTX Pc/PRC2 (Esc, Su(z)12, & Caf1-55) Present at inactive gene promoters, pericentric heterochromatin, and inactive euchromatic regions Overproliferation of early cyst cells; germ cells expressing somatic cell markers; increased hub size; defective germ cell dedifferentiation Male germline; germ cells & cyst cells N.D.
H2AK118ub Sce/dRing PR-DUB PRC1 (Pc, Ph, Psc/Su(z) 2) Promotes chromatin repression N.D. N.D. N.D.
H3K4me3 Trx, Trr, dSet1 Lid Ash2, Rbbp5, Wdr5, Dpy30 Present at promoters of actively transcribing genes GSC loss; overproliferation of GSC-like cells Male & female germline germ cells, escort cells & cap cells N.D.
H3K9m3 Su(var)3–9, Egg Kdm4B HP1 Present at constitutive heterochromatin and maintains pericentric heterochromatin GSC loss or overproliferation of GSC-like cells; escort cell death Male & female germline; germ cells & escort cells N.D.
H3K4me1/2 N. D. Su(var)3–3/Lsd1 HP1/Ova Present at promoters of actively transcribing genes Overproliferation of GSC-like cells; germ cell loss; escort cell death Female germline; germ cell & escort cells N.D.
H4K(5, 8, 12, 16)ac Tip60 N.D. Domino, Nipped-A, E(pc) Regulates gene expression, chromatin packaging and DNA repair GSC cell death; overproliferation of CySC-like cells & GSC-like cells; germ cells express somatic cell markers; spermatogonial tumors Male & female germline; germ cells & cyst cells Yes
H3K23ac Enok N.D. N.D. Promotes gene expression GSC loss Female germline; germ cells & cap cells N.D.

PRC1 components regulate male GSC self-renewal. In the Drosophila testis, compromising Psc and Su(z) 2 function by mutations or RNAi results in CySC-like tumors. Moreover, these overpopulated CySC-like cells outcompete and prevent GSCs from attaching to the hub, leading to GSC loss. This CySC overproliferation phenotype can be attributed to ectopic expression of a homeobox gene Abdominal B (Abd-B), as forced expression of Abd-B in wild-type testes results in a similar tumor phenotype (DeFalco et al., 2004). Therefore, Psc and Su(z) 2 repress Abd-B expression in the adult germline, in order to maintain CySC identity and activity in the adult testes (Morillo Prado et al., 2012). This inadvertently allows for the maintenance of GSCs in a non-cell-autonomous nature by preserving the attachment of GSCs to the hub and preventing CySC encroachment.

In addition to PRC1, PRC2 component E(z) maintains the Drosophila male germline through a cell-autonomous mechanism. In the germline, E(z) mutations cause GSC depletion over time, due to defective dedifferentiation of spermatogonial cells. Knockdown of E(z) by RNAi specifically in spermatogonial cells also induces GSC loss, confirming that dedifferentiation defect contributes to the GSC loss phenotype. In E(z) mutant germ cells, ChIP experiments show increased H3K4me3 at 39% of genes that lose H3K27me3, suggesting that a global chromatin change in the spermatogonial cells undermines their dedifferentiation potential. Therefore, E(z) maintains the global chromatin state of spermatogonial cells that is conducive for dedifferentiation to replenish lost GSCs due to aging or degeneration (Eun et al., 2017).

On the other hand, TrxG acts antagonistically to PcG by activating gene expression through a variety of mechanisms, including histone modifications, ATP-dependent chromatin remodeling, chromosome cohesion, and recruitment of RNA polymerase II to the promoters of target genes (Kassis et al., 2017). In Drosophila, three classes of Complex of Proteins Associated with SET1 (COMPASS) have been identified, each containing a distinct histone methyltransferase [Trithorax (Trx), SET domain containing 1 (Set1), or Trithorax related (Trr)], Ash2, and three additional subunits (Mohan et al., 2011). Set1 is responsible for generating the majority of H3K4me2 and H3K4me3 in Drosophila, while the role of Ash2 is to stabilize the methyltransferase activity of the COMPASS complexes (Table 1) (Ardehali et al., 2011; Dou et al., 2006; Hallson et al., 2012; Kassis et al., 2017). Recently, Set1 has been shown to act with an E3 ubiquitin ligase Bre1 to generate the majority of H3K4me3 modification and regulate GSC maintenance and differentiation in the Drosophila ovary (Bray et al., 2005). Mutation of bre1 or knockdown of set1 by RNAi cause female GSC loss and decreased expression of BMP signaling components, such as pMad or Dad. In the germaria, Dally restricts Dpp diffusion to ensure that only the GSCs receive Dpp signal (Akiyama et al., 2008; Guo and Wang, 2009). In addition, DE-cadherin ensures GSC adhesion to the niche cells (Song and Xie, 2002; Song et al., 2002) (Figure 2B). Inactivation of bre1 or set1 in the terminal filament and cap cells results in female GSC loss with reduced Dally and DE-cadherin expression, leading to precocious GSC differentiation. Moreover, compromising bre1 or set1 in the escort cells induces overproliferation of GSC-like pMad-positive cells due to increased Dpp and Dally. Therefore, Bre1 and Set1 act together to control female GSC maintenance and differentiation through multiple mechanisms (Xuan et al., 2013). It is possible that in the GSCs and the niche, the addition of H3K4me3 by Set1 at the genomic loci of BMP pathway components promotes their expression for the maintenance of GSCs. In addition, Set1 may act in the escort cells to promote the expression of a negative regulator of Dpp and Dally. However, both of these possibilities have yet to be determined by identifying direct targets of H3K4me3 and Set1 in a cell-type-specific manner. In addition, a RNAi screen study identified Set1 as a self-renewal factor in female GSCs (Yan et al., 2014).

Su(var)3-9 and Heterochromatin Protein 1 (HP1):

Telomeric and pericentric chromosomal regions are enriched with transcriptionally repressed heterochromatin, whose formation is regulated by H3K9me2/3 “writer” [Su(var)3-9] and H3K9me2/3 “reader” Heterochromatin Protein 1 (HP1) (Table 1) (Eissenberg et al., 1990; James et al., 1989; James and Elgin, 1986; Schotta et al., 2002; Schotta et al., 2003). The recognition of H3K9me3 by HP1 facilitates heterochromatin formation by recruiting more Su(var)3-9 (Bannister et al., 2001; Grewal and Jia, 2007; Lachner et al., 2001). HP1 has a variety of functions in addition to repressing gene expression, such as telomere capping and maintaining telomere length (Fanti et al., 1998; Perrini et al., 2004; Vermaak and Malik, 2009). In Drosophila, HP1 regulates both male and female GSC maintenance. In the male germline, knockdown of HP1 or Su(var)3-9 induces ectopic Bam expression in the GSCs and GBs, resulting in GSC loss. These phenotypes in the early-stage germline are concomitant with the loss of two germ cell markers, Vasa and Escargot (Esg) (Streit et al., 2002; Voog et al., 2014). On the other hand, overexpression of HP1 or Su(var)3-9 in the early-stage germline results in the opposite phenotypes, shown as loss of Bam expression and overexpression of Vasa and Esg. Overexpression of HP1 also partially rescues the GSC loss phenotype in hopscotch (hop) mutant testes. Since Hop is the Drosophila Receptor Tyrosine Kinase (RTK), mutations in hop result in the loss of proper JAK-STAT signaling in male GSCs and GSC loss (Herrera and Bach, 2019; Kiger et al., 2001; Tulina and Matunis, 2001). Based on these results, HP1 seems to enhance GSC self-renewal by promoting the JAK-STAT signaling (Xing and Li, 2015). Interestingly, in GSC-like cells in the Upd-overexpressing testes, overexpression of HP1 induces differentiation by regaining Bam expression. Therefore, HP1 reverts the GSC tumor phenotype upon hyperactivating the JAK-STAT signaling pathway, suggesting a negative regulation of STAT signaling by HP1 (Loza-Coll et al., 2019). However, it is unclear whether these seemingly contradictory results are due to different genetic backgrounds carrying distinct JAK-STAT signaling activities. For example, HP1 could act as a “rheostat” to upregulate the activity of JAK-STAT signaling when it is low, but downregulate it when it is too high. The molecular mechanisms will await more investigations.

In the female germline, HP1 is also important for GSC maintenance and differentiation. Knockdown of HP1 results in germaria containing only GSC-like cells or only differentiated cells, suggesting an imbalance between GSC self-renewal and differentiation at the cellular level. At the molecular level, both differentiation gene bam and genes promoting stemness as well as germline identity, such as nanos, cup, p-element induced wimpy testis (piwi), and vasa are all downregulated in HP1 mutant ovaries (Forbes and Lehmann, 1998; Gonzalez et al., 2015; Wang and Lin, 2004). CLIP-PCR and ChIP experiments demonstrate that HP1 protein binds to nanos, cup and piwi mRNAs. Therefore, HP1 regulates female GSC maintenance and differentiation though post-transcriptional stabilization of stemness and differentiation gene mRNA by forming an hnRNP nuclear complex (Casale et al., 2019). However, how HP1 balances between these two group of mRNAs for proper GSC maintenance and differentiation remains to be addressed.

Eggless/ dSETDB1:

Eggless/dSETDB1 (Egg) is another histone lysine methyltransferase that catalyzes tri-methylation of H3K9 and regulates Drosophila oogenesis (Table 1) (Clough et al., 2007). Mutations of egg gene causes two main phenotypes in the ovary: accumulation of GSC-like tumor cells and loss of differentiated germ cells (Clough et al., 2014; Wang et al., 2011). Dad expression is lost in the egg mutant GSCs; however, both Bam and Benign gonial cell neoplasm (Bgcn), another germline differentiation factor, are found to be normally expressed, suggesting that Egg maintains GSCs in a Bam-Bgcn independent manner (Clough et al., 2014; Li et al., 2009; Wang et al., 2011). It is possible that Egg represses differentiation genes in GSC, but it is unknown whether this repression is through direct regulation of Dad expression.

Enoki mushroom (Enok):

Enok is a Monocyticleukaemia zinc finger, Ybf2/ Sas3, Sas2 and Tip60 (MYST) family histone acetyltransferase that preferentially acetylates Lys23 on histone H3 tail and promotes transcription (Table 1) (Huang et al., 2014). Compromised Enok in female GSCs causes rapid GSC loss through premature differentiation due to ectopic expression of Bruno, an RNA Recognition Motif (RRM)-containing RNA binding protein that targets Sxl for translational repression, in order to promote CB differentiation and female germline cyst formation (Wang and Lin, 2007). Additionally, loss of Enok in the cap cells results in impaired BMP and Notch signaling, leading to GSC loss due to premature differentiation and niche size reduction, respectively. Therefore, Enok maintains Drosophila female GSCs through both cell-autonomous and non-cell-autonomous mechanisms. This indicates that Enok positively regulates BMP and Notch signaling in the cap cells but negatively regulates Bruno in the female GSCs (Xin et al., 2013).

Ubiquitously transcribed tetratricopeptide repeat, X chromosome histone demethylase (dUTX):

The Drosophila UTX (dUTX) is a H3K27me2/me3-specific demethylase that can form a complex with an acetyltransferase CREB-binding protein (CBP), a chromatin remodeler and TrxG component Brahma (Brm), in order to antagonize PcG-mediated gene silencing (Table 1) (Holowatyj et al., 2015; Smith et al., 2008; Tie et al., 2012). In the male germline, dUTX maintains active transcription of Suppressor of cytokine signaling at 36E (Socs36E) gene, which encodes a negative regulator of the JAK-STAT pathway, by ensuring the transcription start site is deprived of the repressive H3K27me3 mark. Socs36E is predominantly detected in the hub cells and CySCs to maintain proper JAK-STAT signaling activity and the 1:2 ratio of GSCs: CySCs (Figure 2A) (Herrera and Bach, 2019; Issigonis et al., 2009; Terry et al., 2006). Compromising dUTX in the CySCs induces increased Zfh1-positive cells inside and around the hub area as well as ectopic pStat92E in the more differentiated germ cells. In addition, knockdown of dUTX in the germline results in enhanced DE-cadherin expression, leading to increased hub size and hub cell number. These defects can be suppressed by either overexpressing Socs36E or halving Stat92E. Together, dUTX controls JAK-STAT signaling activity and regulates the feedback from stem cells (i.e. GSCs and CySCs) to the niche, in order to maintain niche morphology and regulate proper gene expression non-cell-autonomously (Tarayrah et al., 2013).

Little imaginal Disc (Lid):

Lid is a H3K4me3-specific demethylase that maintains male GSC self-renewal and prevents premature differentiation (Eissenberg et al., 2007; Holowatyj et al., 2015; Lee et al., 2007; Tarayrah et al., 2015). Inactivation of Lid via RNAi knockdown or mutations in testes results in GSC loss and the presence of more differentiated spermatogonial cysts next to the hub, in place of GSCs. In addition, lid mutant testes have ectopic Bam and reduced Stat92E expression in the GSCs. Furthermore, enhanced expression of Stat92E or DE-Cadherin can partially rescue the GSC loss phenotype in lid mutant testes. Therefore, Lid acts cell-autonomously to regulate JAK-STAT signaling in the germline to maintain proper GSC activity and proliferation potential (Tarayrah et al., 2015).

2.5. Chromatin remodeling factors

For biological processes such as DNA replication, recombination, repair, and transcription, chromatin remodeling enzymes change the packaging state of the chromatin (Narlikar et al., 2013). Chromatin remodeling factors use energy from ATP hydrolysis to move, eject or restructure nucleosomes to allow certain factors to either gain or lose access to specific genomic regions. There are four families of chromatin remodeling enzymes: Switch Defective/Sucrose Nonfermenting (SWI/SNF), Imitation Switch (ISWI), Chromodomain-Helicase-DNA-binding protein (CHD), and Inositol Requiring 80 (INO80). All four families have a conserved ATPase subunit that belongs to the helicase superfamily 2 (SNF2), but each has unique flanking domains that specifically regulate its corresponding functions and activities (Eisen et al., 1995). For example, the SWI/SNF family ATPase contains a C-terminal bromodomain that assists in sliding and ejecting nucleosomes (Hassan et al., 2002). The ISWI family ATPase contains a SANT domain that optimizes nucleosomal spacing for chromatin assembly and transcriptional repression activities (Corona et al., 2002; Corona and Tamkun, 2004; Deuring et al., 2000). The CHD family ATPase contains a chromodomain that recognizes modified nucleosomes to be ejected or moved, allowing for transcriptional activity changes (Marfella and Imbalzano, 2007; Woodage et al., 1997). Lastly, the INO80 family has a split ATPase domain, which is responsible for promoting transcriptional activation and DNA repair (Bao and Shen, 2007; Clapier and Cairns, 2009; Narlikar et al., 2013; Shen et al., 2000).

Nucleosome remodeling factor (NURF):

In the ISWI family, NURF is an ATP-dependent complex that plays important roles in both Drosophila male and female early-stage germline (Table 2) (Badenhorst et al., 2002; Xi and Xie, 2005). In the male, the NURF complex components ISWI, Nurf55 and Nurf301 are expressed in the testicular niche, including hub cells, GSCs, and CySCs (Figure 1A). Nurf301, a specific NURF complex component, is required cell-autonomously for both GSC and CySC maintenance. Compromising Nurf301 results in the loss of both GSCs and CySC over time, due to premature differentiation caused by ectopic Bam expression and the loss of Stat92E expression. Increasing Stat92E levels partially rescues the CySC loss phenotype in nurf301 mutant testes. In addition, downregulation of Nurf301 results in partial rescue of the GSC loss phenotype in socs36E mutant. These results indicate that NURF interacts with JAK-STAT pathway components to ensure proper stem cell maintenance in testes, by promoting Stat92E expression and preventing precocious Bam expression (Cherry and Matunis, 2010).

Table 2:

Chromatin Remodelers in the Drosophila Male and Female Gonads

Family Complex ATPase other subunits Function Germline loss of function phenotype Gonad studied & cell types present
SWI/SNF PBAP BRM/Brahma Polybromo / BAP180, BAP170 Slides and ejects nucleosomes for diverse processes including DNA damage repair and recruitment and initiation of transcription GSC loss Female germline; cap cells & germ cells
ISWI NURF ISWI Nurf301, Nurf55, Nurf38 Optimize nucleosome spacing for chromatin assembly; promote H1 loading for chromatin compaction; regulates Dosage compensation; randomizes spacing for RNA Pol II activation; transcriptional repression GSC and CySC loss Male and female germline; hub cells, germ cells & cyst cells
CHD Mi-2/NuRD dMi-2 dMBD2/3, dMTA, dRPD3, p55, p66/68 Slides or ejects nucleosomes to promote transcription elongation N.D. Male germline; germ cells
INO80 Tip60 Domino dTip60, E(Pc) INO80 complexes promote transcriptional activation and DNA repair; promotes H2Av incorporation GSC and CySC loss; cystoblast cell differentiation defects Male and female germline; germ cells, cyst cells, escort cells, cap cells, & follicle cells

In addition, the NURF complex interacts with the Ecdysone (Ecd) steroid hormone pathway in female GSCs and male CySCs to maintain stem cell self-renewal. Binding of Ecd to the Ecdysone Receptor (EcR) results in the dimerization between EcR and Ultraspecle (Usp), which initiates a transcriptional cascade including E74, E75, and Broad (Br) (Schwedes and Carney, 2012). In the ovary, loss of Usp, E74, Ecd, or EcR results in female GSC loss. Additionally, Usp or E74 mutant female GSCs display decreased ISWI expression and compromised BMP signaling. Therefore, Ecdysone signaling acts through the NURF chromatin remodeling complex to promote female GSC maintenance by regulating BMP signaling (Ables and Drummond-Barbosa, 2010). In the testes, Ecdysone signaling components are expressed in hub cells and the CySC lineage. Inactivation of Ecd in the CySCs results in both GSC and CySC loss. Additionally, EcR has been shown to interact with Nurf301 to maintain both stem cell populations. Together, these results show that EcR and Nurf301 interact to regulate the transcription of stemness factors to promote the self-renewal of both GSCs and CySCs in the testis (Li et al., 2014).

Polybromo-containing Bap (PBAP):

In Drosophila, Brm is the only SWI/SNF chromatin remodeling ATPase and is present in both SWI/SNF complexes: Brm-associated protein (BAP) complex and PBAP complex (Table 2) (Mohrmann et al., 2004; Moshkin et al., 2007). In the Drosophila ovary, loss of PBAP components Bap180 and Brm in the germline or niche has been shown to result in GSC loss over time, suggesting that PBAP complex is required for female GSC maintenance (He et al., 2014).

Domino (Dom):

In the INO80 family, Dom is a SWR1-like ATPase that is linked to the chromatin incorporation of the histone variant H2Av in both male and female germline (Table 2) (Kobor et al., 2004; Mizuguchi et al., 2004; Ruhf et al., 2001). In the male, clonal induction using dom mutations results in both GSC and CySC loss over time, suggesting that Dom is required for GSC and CySC maintenance cell-autonomously. Additionally, reduced H2Av incorporation is detected in dom mutant GSCs. Therefore, both Dom and H2Av are required to maintain GSCs and CySCs, likely by repressing differentiation genes and/or maintaining active expression of self-renewal genes (Morillo Prado et al., 2013). In the female, Dom is identified as a GSC self-renewal factor (Yan et al., 2014)

Nucleosome Remodeling and Deacetylase (NuRD):

In the CHD family, NuRD is the ATPase complex that functions antagonistically to the DNA replication-related element factor (DREF), in order to regulate male GSC maintenance (Table 2) (Hirose et al., 2002; Hirose et al., 1996). Compromising DREF leads to precocious GSC differentiation and GSC loss over time. GSCs without DREF display impaired Dad expression and ectopic Bam expression due to compromised BMP signaling. Genetic interaction experiments show that mutations in NuRD complex components Mi-2 and Caf1 suppress the GSC loss phenotype in the DREF mutants. Therefore, NuRD antagonizes DREF while DREF allows GSCs to self-renew, likely as a transcription factor to promote expression of BMP pathway genes to inhibit differentiation (Angulo et al., 2019).

2.6. RNA- binding proteins and non-coding RNAs

MicroRNAs (miRNAs) are small RNAs that negatively regulate gene expression post-transcriptionally. It has been demonstrated that miRNAs regulate stemness, cell division, differentiation, and homeostasis of multiple Drosophila stem cells (Hatfield et al., 2005; Park et al., 2007; Shcherbata, 2019). Primary miRNAs (pri-miRNAs) are expressed in the nucleus by RNA polymerase II. The hairpin structure of pri-miRNAs is recognized by the RNase II enzyme, Drosha, which along with its partner, Pasha/DGCR8, cleaves the pri-miRNAs into precursor miRNAs (pre-miRNAs). Subsequently, another RNase II enzyme, Dicer-1 (Dcr-1), which along with its partner, Loquacious, cleaves pre-miRNAs into a 22-nucleotide RNA duplex followed by interacting with Argonaute 1 (Ago1). With Ago1, miRNAs form the RNA-induced silencing complex (miRISC), which cleave and degrade target mRNAs, as well as repress their translation (Shcherbata, 2019).

Mutations in genes encoding components for miRNA biogenesis, such as dcr-1, ago1, loquacious, and mei-P26, all lead to GSC loss phenotypes (Azzam et al., 2012; Forstemann et al., 2005; Jin and Xie, 2007; Li et al., 2012; Yang et al., 2007). In mei-P26 mutant ovaries, GSC loss can be attributed to disrupted BMP signal transduction, which results in the impaired Dad and pMad expression in GSCs. In addition, ectopic Bam and Brain tumor (Brat) expression is detected in the mei-P26 mutant germaria. In wild-type GSCs, brat mRNA is the target of the Nanos-Pumilio complex: when Nanos expression decreases in the late-stage germline, Brat expression increases and in turn, represses the translation of Mad. When pMad is not present, bam expression is de-repressed, which initiates germline differentiation (Figure 2B) (Harris et al., 2011). These results suggest that Mei-P26 cooperates with Nanos and miRISC to repress the translation of specific mRNA targets such as brat, with which pMad is upregulated and bam is repressed, a condition for promoting GSC self-renewal (Li et al., 2012; Li et al., 2013).

In addition, miRNAs themselves regulate GSC maintenance in both male and female gonads. In the testis, malfunction of the miRNA-310/313 cluster results in abnormal germline and somatic gonadal cell differentiation, which can be suppressed by reducing Armadillo (Arm, or β-catenin) levels. Indeed, miR-310/313 recognizes the 3’ UTR of arm mRNA and targets it for degradation. Arm is an important effector of the Wnt signaling pathway and a key cell-cell adhesion molecule contributing to attachment of GSCs to the niche cells (Song et al., 2002) (Figure 2). These results indicate that miR-310/313 controls Wnt signaling and regulates GSC-hub cell adhesion in testes (Pancratov et al., 2013).

Piwi interacting RNA (piRNAs) are the most abundant small non-coding RNA in the Drosophila male and female gonads. The main role of piRNAs is to protect genomes of both germline and somatic gonadal cells by repressing transposons (Brennecke et al., 2008; Hermant et al., 2015). The piRNAs act in a complex with the PIWI family proteins, which includes Piwi, Aubergine (Aub), and Argonaute 3 (Ago3) (Brennecke et al., 2007; Saito et al., 2006). Together, this complex silences transposons at both the transcriptional and post-transcriptional levels (Le Thomas et al., 2013; Lim et al., 2009; Vagin et al., 2006; Yang and Xi, 2017). Piwi is the founding member of the PIWI family and is present in both germline and somatic gonadal cells, while Aub and Ago3 are only required for piRNA production in the germline (Brennecke et al., 2007). In the ovaries, Piwi protein has been shown to interact with PcG components in the terminal filament cells, cap cells and GSCs to regulate female GSC maintenance and differentiation (Peng et al., 2016). Loss of Piwi in the early germline results in GSC loss, but Pc or E(z) mutations suppress this phenotype in piwi mutant ovaries. Piwi also physically interacts with PcG components Su(z)12 and Esc; E(z) has a higher enrichment at its target genes in piwi mutant ovaries. As Su(z)12, Esc and E(z) are all PRC2 components, Piwi may interact with Su(z)12 and Esc to sequester E(z) away from the chromatin, which may lead to reduced H3K27me3 but increased RNA Polymerase II at target genes to activate transcription. Therefore, the negative regulation of PRC2 and H3K27me3 by Piwi may be necessary for transposon suppression, proper female GSC activity, and oogenesis (Peng et al., 2016).

3. Epigenetic Mechanisms Regulating GSC Differentiation

3.1. Canonical Histones

Canonical linker histone H1 regulates proper female GSC differentiation. In the ovary, H1 is required for escort cell maintenance and germ cell differentiation. Knockdown of H1 in the escort cells leads to accumulation of GSC-like cells with upregulated BMP signaling, ectopic expression of cap cell-specific genes, as well as elevated transposon activity and DNA damage, resulting in escort cell death. Overall, these results demonstrate that H1 acts cell-autonomously in escort cells to repress cap cell identity and transposon activity, which are necessary to maintain escort cell identity and regulate proper germ cell differentiation (Yang et al., 2017).

3.2. Histone Modifying Enzymes

Polycomb Group (PcG) and Trithorax Group (TrxG):

In addition to Su(z)2 and Psc, Pc has been shown to regulate Abd-B expression in the CySCs. Knockdown of Pc in the CySCs leads to Zfh1-positive CySC-like tumor cells, overproliferation of early-stage germ cells at the expense of differentiation. The overproliferative CySCs phenotype can be partially rescued by compromising Abd-B, suggesting mis-expressed Abd-B in the CySCs underlies these cellular defects (Zhang et al., 2017).

In the Drosophila ovary, knockdown of PRC1 components Sce, Ph, or Psc/Su(z) 2 in the escort cells results in a GSC-like tumor cells, which express the BMP pathway components pMad and Dad, but lack Bam expression. Upregulated BMP signaling is responsible for this tumor phenotype, as compromising Dpp results in partial rescue. Finally, the TrxG component Brm activates Dpp expression in the escort cells, as compromising Brm also rescues this tumor phenotype (Dingwall et al., 1995). Therefore, PRC1 acts in the escort cell to repress Dpp to allow for the proper differentiation of GSCs. Either inactivation of PRC1 or hyperactivation of Brm induces ectopic Dpp expression, which prevents germline differentiation with accumulation of GSC-like tumor cells in the Drosophila ovary (Li et al., 2016). Therefore, PRC1 acts non-cell autonomously in the escort cells to restrict active BMP signaling in GSCs.

Eggless/ dSETDB1:

In addition to maintaining GSCs, Egg is required for GSC differentiation. Loss of egg results in escort cell death, increased BMP signaling in the germaria, and GSC differentiation defects (Wang et al., 2011). Egg also acts in escort cells to regulate GSC differentiation non-cell autonomously, likely by repressing BMP regulators, such as Dally.

Su(var)3-3/ lysine -specific demethylase 1 (Lsd1):

Lsd1 is a demethylase for H3K4 mono- and di methylation and is a regulator for heterochromatin spreading and GSC niche size in the Drosophila ovary (Table 1) (Di Stefano et al., 2007; Eliazer et al., 2011; Holowatyj et al., 2015; Rudolph et al., 2007). Lsd1 also acts non-cell-autonomously to regulate female GSC differentiation. Inactivation of lsd1 by mutation or RNAi in escort cells induces escort cell death and formation of GSC-like tumors, due to ectopic Dad expression and loss of Bam expression. ChIP experiments suggest that Engrailed is a direct target of Lsd1, suggesting that BMP signaling is indirectly repressed by Lsd1. In wild-type germaria, Engrailed, a homeobox transcription factor, acts in the terminal filament and cap cells to maintain female GSCs, but in lsd1 mutant germaria, Engrailed is detected in the escort cells (Bolivar et al., 2006; Desplan et al., 1985; Kornberg, 1981; Morata and Lawrence, 1975; Rojas-Rios et al., 2012). Moreover, compromising Engrailed in lsd1 mutant ovaries rescues the GSC-like tumor phenotype and overexpression of Engrailed phenocopies lsd1 mutant phenotype. Knockdown of lsd1 in the escort cells also causes cell fate changes, shown by ectopic expression of Hedgehog (Hh), which is normally expressed exclusively in the terminal filament and cap cells to promote expression of Dpp for the neighboring GSCs (Rojas-Rios et al., 2012). In conclusion, Lsd1 acts in the escort cells to inhibit Engrailed, which prevents hyperactive Hh signaling from activating Dpp expression. These functions restrict Dpp expression solely in the cap cells and terminal filament cells, which in turn ensures BMP signaling is only received by the female GSCs (Eliazer et al., 2014; Eliazer et al., 2011).

Tat interactive protein 60kDa (Tip60):

The Drosophila Tip60 complex acetylates lysines 5, 8, 12, and 16 on histone H4 to regulate gene expression, chromatin packaging, and DNA repair (Kusch et al., 2004; Schirling et al., 2010). It is comprised of the histone acetyltransferase Tip60, the ATPase Domino, Nipped-A, Enhancer of Pc [E(Pc)] and a few other components (Table 1) (Clapier and Cairns, 2009; McCarthy et al., 2018). Among them, E(Pc) and Nipped-A are shown to regulate male and female GSC differentiation, respectively. Knockdown of E(Pc) in the CySC and early cyst cells in testes causes overproliferation of CySC-like cells and GSC-like cells, and ectopic expression of somatic cell markers in germline tumors. Cell-type-specific ChIP experiments show that E(Pc) directly binds to genes encoding components of multiple signaling pathways, including JAK-STAT and EGF. The EGF signaling pathway has been previously shown to control encapsulation, proper differentiation and mitotic divisions of early-stage germ cells by the cyst cells (Kiger et al., 2000; Parrott et al., 2012; Schulz et al., 2002). Inactivation of Tip60 acetyltransferase function in the CySC lineage results in similar phenotypes as seen in E(Pc) mutant testis. Therefore, E(Pc) and Tip60 act synergistically to promote CySC differentiation cell-autonomously and to regulate proper germ cell differentiation and maintain germline identity non-cell-autonomously (Feng et al., 2017).

In the male germline, E(Pc) is required for proper germ cell activity, as the germline-specific knockdown of E(Pc) results in germline tumor and GSC cell death phenotypes. In addition, compromising E(Pc) in the germline results in increased CycB expression in the late-stage germ cells and enhanced γH2Av, a marker for DNA double-strand break (DSB) and cell death (Lake et al., 2013). Finally, knocking down E(Pc) in the germline reduces Bam expression, responsible for the germline tumor phenotype. In summary, in the male germline E(Pc) regulates transcription of cycB through H4 acetylation, accumulation of Bam protein, and DNA repair for proper GSC differentiation and the mitosis-to-meiosis transition (Feng et al., 2018).

In the female germline, knockdown of Nipped-A results in reduced Bam expression, leading to the accumulation of GSC-like tumor cells. Inactivation of Tip60 complex components, such as Bap55, Domino, E(Pc), YL-1 and Tip60, also results in GSC-like tumors in the ovaries. Through ChIP experiments, bgcn is detected as a direct target gene of Nipped-A. Since Bgcn regulates GSC differentiation, overexpressing Bgcn could rescue differentiation defects of Nipped-A knockdown germ cells. In conclusion, the Tip60 complex is required cell autonomously to ensure Bgcn expression in the pre-cystoblast to promote female GSC differentiation (McCarthy et al., 2018).

Ovaries absent (Ova):

A recent study identified Ova and its bridging role between HP1 and Lsd1 to silence heterochromatic genes in the Drosophila ovary. The ova mutant ovaries show either germline tumor or germ cell loss phenotypes. Specific knockdown of ova in the somatic gonadal cells results in GSC-like tumor cells, accompanied with ectopic pMad and Dad expression but decreased Bam levels. Biochemical assays show that Ova directly binds to Lsd1 and HP1α in vitro and expression of the Ova protein fragment that interacts with both HP1α and Lsd1 is sufficient to rescue the ova mutant defects in ovaries. Moreover, ChIP experiments show increased H3K4me2 at telomeric transposons in ova mutant ovaries compared to the control ovaries. Given that Lsd1 is an H3K4me2 demethylase, these results suggest that Ova is indispensable for the H3K4 demethylation activity of Lsd1 in heterochromatic gene silencing. Therefore, Ova is necessary for heterochromatin formation as well as HP1- and Lsd1-mediated gene silencing of GSC self–renewal genes to ensure proper differentiation of female GSCs (Yang et al., 2019).

3.3. RNA- binding proteins and noncoding RNAs

miRNAs and piRNAs have also been shown to regulate GSC differentiation. Mutations of miR-9a result in decreased GSC division and failure in spermatogenesis. RNA-seq experiments have shown increased N-cadherin expression in miR-9a mutant testes. Consistently, overexpression of N-cadherin phenocopies while compromising N-cadherin levels suppresses miR-9a mutant phenotypes. Since N-cadherin is a cadherin family member for adherens junction between GSCs and hub cells (Figure 2), these results suggest that normal functions of miR-9a downregulate N-cadherin to allow for detachment of GSCs from the niche for differentiation (Epstein et al., 2017).

In the ovary, Vret, a Tutor domain-containing protein, is associated with Piwi and Aub. Knocking down Vret in the germline or the somatic gonadal cells results in mis-localized Piwi to the nucleus in both cell types and loss of Aub in the germline nuage structure. In addition, vret mutant ovaries show reduced piRNA levels and enhanced transposons mobilization, as well as somatic cell death and GSC differentiation defects. These result in germaria filled with GSC-like cells, but these phenotypes can be suppressed when wild-type Vret is expressed in the somatic gonadal cells. Based on these data, Vret is likely an essential component of the Piwi- and Aub-containing RISC complexes in the germline, as well as Piwi-containing RISC complexes in the somatic gonadal cells, to regulate transposon levels and biogenesis of primary piRNAs in the ovary (Zamparini et al., 2011).

4. Conclusions and Perspective

In this review, we focus on distinct epigenetic regulatory mechanisms in both female and male GSC maintenance and differentiation using Drosophila as the model organism. The commonalities and differences between these two GSC systems provide insight into some general scenario as well as sexual dimorphism. Here, we summarize these highlights and comment on existing caveats as well as future perspectives:

  1. Both GSC systems achieve a balanced ratio between stem cells and differentiating cells through ACD, which greatly facilitate studying epigenetic mechanisms in endogenous stem cells at single-cell resolution. This feature, combined with cell-type-specific labeling and cutting-edge imaging methods, such as live cell imaging and superresolution imaging, will provide more important insight on how epigenetic regulators play biological functions in vivo.

  2. Both GSC systems have well characterized niche structure, critical signaling pathways emanated from the niche, cellular features including cell cycle progression and markers for inter- and intra-cellular components. Interestingly, studies exploring the roles of epigenetic factors in regulating GSC self-renewal and differentiation mainly reveal their control on the activity of two signaling pathways: JAK/STAT and BMP pathways. These could be due to two reasons: First, it is likely because of the key roles these two signaling pathways play in determining the balance between stem cell fate and differentiation cell fate. For example, epigenetic regulators tend to change the expression of stemness genes, such as dad and stat92E, or the differentiation genes, such as bam and bgcn. Second, the convolution of the results on a few downstream targets could be due to detection methods. Given the abundant reagents available to analyze abnormal expression or localization of these known key factors, it is not surprising that they serve as the top candidates when designing experiments to explore the “read-outs” of abnormal epigenetic regulation. However, rapid development on single-cell genomics and epigenomics techniques will now or soon allow characterization of the direct targets of these epigenetic regulators in pure cells with distinct identities, leading to a comprehensive understanding of their roles in adult stem cell systems. On the other hand, due to their intricate regulation on signaling pathways, it is intriguing to see that many of these epigenetic regulations have both cell-autonomous and non-cell-autonomous roles. Co-existence of GSCs with somatic gonadal cells in both systems provides a unique opportunity to dissect these relationships in vivo, given their reliable anatomy and morphological distinctions, as well as the relative less complicated tissue architecture compared to the mammalian tissue/organ.

  3. Interestingly, disruption of epigenetic regulation often results in multiple cellular defects. Characterization of these pleiotropic phenotypes often reveal gene misexpression (i.e. expression levels are too high, too low, or are ectopic in the wrong cell and/or at the wrong timing), suggesting a rather confused cell fate. Given that it is very plausible that each epigenetic regulator has many targets and even the same epigenetic regulator has different targets in different cells, the nature of the cell fate state in their loss-of-function mutants could be quite complicated. Furthermore, most of the “rescuing” results when genetic suppression has been detected are mainly based on morphological recovery but not on functional recovery, the later requires more rigorous assay including the ability to regain stem cell activity for both successful self-renew and full differentiation to become functional egg or sperm. Therefore, to completely understand their molecular and cellular defects, new technology such as single-cell RNA-seq could help elucidate the genome-wide transcriptome change; and lineage tracing could reveal their differentiation footage. Finally, given our current knowledge that certain chromatin changes could precede actual transcription change, such as the phenomena of “poised” RNA Pol II and bivalent histone modifications, the downstream effects could comprise both actual gene expression changes and alterations only at the chromatin level. The later will require chromatin structure profiling using small number of cell or even at the single-cell level, which is becoming more and more feasible given the quick advancement of single-cell epigenome techniques.

  4. Finally, even though results using both female and male GSC systems demonstrate many common themes, distinct feature have also been identified, as discussed throughoutin this review. Many of these distinctions could reflect the intrinsic differences between these two adult stem cell lineages. These include findings from many fly germ cell laboratories, such as the different Bam expression patterns, the distinct roles of the JAK-STAT pathway, position and function differences of certain organelles (e.g. spectrosome), germline-soma interactions at the niche, etc. As their abnormalities are often found upon compromising certain epigenetic regulator using genetic mutations or RNAi knockdown, which often takes some time for the phenotypes to manifest. It is still challenging to know the dynamics of the functions of epigenetic regulators. More acute disruption methods especially the reversible ones, such as the Auxin-inducible degron (AID) technology, combined with high-temporal resolution analyses of cellular defects, could greatly facilitate understanding of their primary roles.

In summary, our understandings of epigenetic regulation in early-stage germ cells at single-gene resolution have greatly improved in recent years. Moreover, the advanced techniques will pave the way for better understanding of the dynamic epigenetic regulation in the Drosophila gonads, at both genome-wide level and individual gene locus, as well as in real-time with single cell resolution.

Figure 3: Epigenetic Regulation of GSC maintenance and differentiation in the male and female Drosophila germline.

Figure 3:

Chromatin regulators and histone modifying enzymes are placed where they regulate GSC maintenance or differentiation in the male (A) and female (B) early germline. Schematic drawings of niche and somatic cells regulating GSC maintenance, GSC self-renewal, and GSC differentiation.

Highlights:

Note—Since this is a review but not a research article, we do not think this requirement applies. Please let us know if you think otherwise. Thanks!

Acknowledgements:

We apologize to people whose work cannot be discussed in this review due to limited space. We thank X.C. lab members for insightful suggestions. Supported by NIH F31GM134641 (V.V.), NIGMS/NIH R35GM127075, NICHD/NIH R01HD102474, the Howard Hughes Medical Institute, the David and Lucile Packard Foundation, and Johns Hopkins University startup funds (X.C.).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest: The authors declare no conflict of interest.

References

  1. Ables ET, Drummond-Barbosa D, 2010. The steroid hormone ecdysone functions with intrinsic chromatin remodeling factors to control female germline stem cells in Drosophila. Cell Stem Cell 7, 581–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Akhtar A, Becker PB, 2000. Activation of transcription through histone H4 acetylation by MOF, an acetyltransferase essential for dosage compensation in Drosophila. Mol Cell 5, 367–375. [DOI] [PubMed] [Google Scholar]
  3. Akiyama T, Kamimura K, Firkus C, Takeo S, Shimmi O, Nakato H, 2008. Dally regulates Dpp morphogen gradient formation by stabilizing Dpp on the cell surface. Dev Biol 313, 408–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Allfrey VG, Faulkner R, Mirsky AE, 1964. Acetylation and Methylation of Histones and Their Possible Role in the Regulation of Rna Synthesis. Proc Natl Acad Sci U S A 51, 786–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Anderson AE, Karandikar UC, Pepple KL, Chen Z, Bergmann A, Mardon G, 2011. The enhancer of trithorax and polycomb gene Caf1/p55 is essential for cell survival and patterning in Drosophila development. Development 138, 1957–1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Angulo B, Srinivasan S, Bolival BJ, Olivares GH, Spence AC, Fuller MT, 2019. DREF Genetically Counteracts Mi-2 and Caf1 to Regulate Adult Stem Cell Maintenance. PLoS Genet 15, e1008187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Ardehali MB, Mei A, Zobeck KL, Caron M, Lis JT, Kusch T, 2011. Drosophila Set1 is the major histone H3 lysine 4 trimethyltransferase with role in transcription. The EMBO Journal 30, 2817–2828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Azzam G, Smibert P, Lai EC, Liu JL, 2012. Drosophila Argonaute 1 and its miRNA biogenesis partners are required for oocyte formation and germline cell division. Dev Biol 365, 384–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Badenhorst P, Voas M, Rebay I, Wu C, 2002. Biological functions of the ISWI chromatin remodeling complex NURF. Genes Dev 16, 3186–3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Baldi S, Becker PB, 2013. The variant histone H2A.V of Drosophila--three roles, two guises. Chromosoma 122, 245–258. [DOI] [PubMed] [Google Scholar]
  11. Bannister AJ, Kouzarides T, 2011. Regulation of chromatin by histone modifications. Cell Res 21, 381–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bannister AJ, Zegerman P, Partridge JF, Miska EA, Thomas JO, Allshire RC, Kouzarides T, 2001. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. Nature 410, 120–124. [DOI] [PubMed] [Google Scholar]
  13. Bao Y, Shen X, 2007. INO80 subfamily of chromatin remodeling complexes. Mutat Res 618, 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bartke T, Vermeulen M, Xhemalce B, Robson SC, Mann M, Kouzarides T, 2010. Nucleosome-interacting proteins regulated by DNA and histone methylation. Cell 143, 470–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bolivar J, Pearson J, Lopez-Onieva L, Gonzalez-Reyes A, 2006. Genetic dissection of a stem cell niche: the case of the Drosophila ovary. Dev Dyn 235, 2969–2979. [DOI] [PubMed] [Google Scholar]
  16. Boros IM, 2012. Histone modification in Drosophila. Brief Funct Genomics 11, 319–331. [DOI] [PubMed] [Google Scholar]
  17. Brawley C, Matunis E, 2004. Regeneration of male germline stem cells by spermatogonial dedifferentiation in vivo. Science 304, 1331–1334. [DOI] [PubMed] [Google Scholar]
  18. Bray S, Musisi H, Bienz M, 2005. Bre1 is required for Notch signaling and histone modification. Dev Cell 8, 279–286. [DOI] [PubMed] [Google Scholar]
  19. Brennecke J, Aravin AA, Stark A, Dus M, Kellis M, Sachidanandam R, Hannon GJ, 2007. Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila. Cell 128, 1089–1103. [DOI] [PubMed] [Google Scholar]
  20. Brennecke J, Malone CD, Aravin AA, Sachidanandam R, Stark A, Hannon GJ, 2008. An epigenetic role for maternally inherited piRNAs in transposon silencing. Science 322, 1387–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Buszczak M, Paterno S, Spradling AC, 2009. Drosophila stem cells share a common requirement for the histone H2B ubiquitin protease scrawny. Science 323, 248–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y, 2002. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 298, 1039–1043. [DOI] [PubMed] [Google Scholar]
  23. Capuano F, Mulleder M, Kok R, Blom HJ, Ralser M, 2014. Cytosine DNA methylation is found in Drosophila melanogaster but absent in Saccharomyces cerevisiae, Schizosaccharomyces pombe, and other yeast species. Anal Chem 86, 3697–3702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Casale AM, Cappucci U, Fanti L, Piacentini L, 2019. Heterochromatin protein 1 (HP1) is intrinsically required for post-transcriptional regulation of Drosophila Germline Stem Cell (GSC) maintenance. Sci Rep 9, 4372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Casanueva MO, Ferguson EL, 2004. Germline stem cell number in the Drosophila ovary is regulated by redundant mechanisms that control Dpp signaling. Development 131, 1881–1890. [DOI] [PubMed] [Google Scholar]
  26. Casper AL, Baxter K, Van Doren M, 2011. no child left behind encodes a novel chromatin factor required for germline stem cell maintenance in males but not females. Development 138, 3357–3366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chen D, McKearin D, 2003a. Dpp signaling silences bam transcription directly to establish asymmetric divisions of germline stem cells. Curr Biol 13, 1786–1791. [DOI] [PubMed] [Google Scholar]
  28. Chen D, McKearin DM, 2003b. A discrete transcriptional silencer in the bam gene determines asymmetric division of the Drosophila germline stem cell. Development 130, 1159–1170. [DOI] [PubMed] [Google Scholar]
  29. Cheng J, Turkel N, Hemati N, Fuller MT, Hunt AJ, Yamashita YM, 2008. Centrosome misorientation reduces stem cell division during ageing. Nature 456, 599–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Cherry CM, Matunis EL, 2010. Epigenetic regulation of stem cell maintenance in the Drosophila testis via the nucleosome-remodeling factor NURF. Cell Stem Cell 6, 557–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Clapier CR, Cairns BR, 2009. The biology of chromatin remodeling complexes. Annu Rev Biochem 78, 273–304. [DOI] [PubMed] [Google Scholar]
  32. Clough E, Moon W, Wang S, Smith K, Hazelrigg T, 2007. Histone methylation is required for oogenesis in Drosophila. Development 134, 157–165. [DOI] [PubMed] [Google Scholar]
  33. Clough E, Tedeschi T, Hazelrigg T, 2014. Epigenetic regulation of oogenesis and germ stem cell maintenance by the Drosophila histone methyltransferase Eggless/dSetDB1. Dev Biol 388, 181–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Corona DF, Clapier CR, Becker PB, Tamkun JW, 2002. Modulation of ISWI function by site-specific histone acetylation. EMBO Rep 3, 242–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Corona DF, Tamkun JW, 2004. Multiple roles for ISWI in transcription, chromosome organization and DNA replication. Biochim Biophys Acta 1677, 113–119. [DOI] [PubMed] [Google Scholar]
  36. Czermin B, Melfi R, McCabe D, Seitz V, Imhof A, Pirrotta V, 2002. Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell 111, 185–196. [DOI] [PubMed] [Google Scholar]
  37. Davies EL, Fuller MT, 2008. Regulation of self-renewal and differentiation in adult stem cell lineages: lessons from the Drosophila male germ line. Cold Spring Harb Symp Quant Biol 73, 137–145. [DOI] [PubMed] [Google Scholar]
  38. Davies EL, Lim JG, Joo WJ, Tam CH, Fuller MT, 2013. The transcriptional regulator lola is required for stem cell maintenance and germ cell differentiation in the Drosophila testis. Dev Biol 373, 310–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Dawson MA, Bannister AJ, Gottgens B, Foster SD, Bartke T, Green AR, Kouzarides T, 2009. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature 461, 819–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Decotto E, Spradling AC, 2005. The Drosophila ovarian and testis stem cell niches: similar somatic stem cells and signals. Dev Cell 9, 501–510. [DOI] [PubMed] [Google Scholar]
  41. DeFalco T, Le Bras S, Van Doren M, 2004. Abdominal-B is essential for proper sexually dimorphic development of the Drosophila gonad. Mech Dev 121, 1323–1333. [DOI] [PubMed] [Google Scholar]
  42. Desplan C, Theis J, O’Farrell PH, 1985. The Drosophila developmental gene, engrailed, encodes a sequence-specific DNA binding activity. Nature 318, 630–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Deuring R, Fanti L, Armstrong JA, Sarte M, Papoulas O, Prestel M, Daubresse G, Verardo M, Moseley SL, Berloco M, Tsukiyama T, Wu C, Pimpinelli S, Tamkun JW, 2000. The ISWI chromatin-remodeling protein is required for gene expression and the maintenance of higher order chromatin structure in vivo. Mol Cell 5, 355–365. [DOI] [PubMed] [Google Scholar]
  44. Di Stefano L, Ji JY, Moon NS, Herr A, Dyson N, 2007. Mutation of Drosophila Lsd1 disrupts H3-K4 methylation, resulting in tissue-specific defects during development. Curr Biol 17, 808–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Dingwall AK, Beek SJ, McCallum CM, Tamkun JW, Kalpana GV, Goff SP, Scott MP, 1995. The Drosophila snr1 and brm proteins are related to yeast SWI/SNF proteins and are components of a large protein complex. Mol Biol Cell 6, 777–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Dou Y, Milne TA, Ruthenburg AJ, Lee S, Lee JW, Verdine GL, Allis CD, Roeder RG, 2006. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat Struct Mol Biol 13, 713–719. [DOI] [PubMed] [Google Scholar]
  47. Eisen JA, Sweder KS, Hanawalt PC, 1995. Evolution of the SNF2 family of proteins: subfamilies with distinct sequences and functions. Nucleic Acids Res 23, 2715–2723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Eissenberg JC, James TC, Foster-Hartnett DM, Hartnett T, Ngan V, Elgin SC, 1990. Mutation in a heterochromatin-specific chromosomal protein is associated with suppression of position-effect variegation in Drosophila melanogaster. Proc Natl Acad Sci U S A 87, 9923–9927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Eissenberg JC, Lee MG, Schneider J, Ilvarsonn A, Shiekhattar R, Shilatifard A, 2007. The trithorax-group gene in Drosophila little imaginal discs encodes a trimethylated histone H3 Lys4 demethylase. Nat Struct Mol Biol 14, 344–346. [DOI] [PubMed] [Google Scholar]
  50. Eliazer S, Buszczak M, 2011. Finding a niche: studies from the Drosophila ovary. Stem Cell Res Ther 2, 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Eliazer S, Palacios V, Wang Z, Kollipara RK, Kittler R, Buszczak M, 2014. Lsd1 restricts the number of germline stem cells by regulating multiple targets in escort cells. PLoS Genet 10, e1004200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Eliazer S, Shalaby NA, Buszczak M, 2011. Loss of lysine-specific demethylase 1 nonautonomously causes stem cell tumors in the Drosophila ovary. Proc Natl Acad Sci U S A 108, 7064–7069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Epstein Y, Perry N, Volin M, Zohar-Fux M, Braun R, Porat-Kuperstein L, Toledano H, 2017. miR-9a modulates maintenance and ageing of Drosophila germline stem cells by limiting N-cadherin expression. Nat Commun 8, 600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Eun SH, Feng L, Cedeno-Rosario L, Gan Q, Wei G, Cui K, Zhao K, Chen X, 2017. Polycomb Group Gene E(z) Is Required for Spermatogonial Dedifferentiation in Drosophila Adult Testis. J Mol Biol 429, 2030–2041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Fanti L, Giovinazzo G, Berloco M, Pimpinelli S, 1998. The heterochromatin protein 1 prevents telomere fusions in Drosophila. Mol Cell 2, 527–538. [DOI] [PubMed] [Google Scholar]
  56. Feng L, Shi Z, Chen X, 2017. Enhancer of polycomb coordinates multiple signaling pathways to promote both cyst and germline stem cell differentiation in the Drosophila adult testis. PLoS Genet 13, e1006571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Feng L, Shi Z, Xie J, Ma B, Chen X, 2018. Enhancer of polycomb maintains germline activity and genome integrity in Drosophila testis. Cell Death Differ 25, 1486–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Flaherty MS, Salis P, Evans CJ, Ekas LA, Marouf A, Zavadil J, Banerjee U, Bach EA, 2010. chinmo is a functional effector of the JAK/STAT pathway that regulates eye development, tumor formation, and stem cell self-renewal in Drosophila. Dev Cell 18, 556–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Forbes A, Lehmann R, 1998. Nanos and Pumilio have critical roles in the development and function of Drosophila germline stem cells. Development 125, 679–690. [DOI] [PubMed] [Google Scholar]
  60. Forstemann K, Tomari Y, Du T, Vagin VV, Denli AM, Bratu DP, Klattenhoff C, Theurkauf WE, Zamore PD, 2005. Normal microRNA maturation and germ-line stem cell maintenance requires Loquacious, a double-stranded RNA-binding domain protein. PLoS Biol 3, e236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Francis NJ, Kingston RE, Woodcock CL, 2004. Chromatin compaction by a polycomb group protein complex. Science 306, 1574–1577. [DOI] [PubMed] [Google Scholar]
  62. Francis NJ, Saurin AJ, Shao Z, Kingston RE, 2001. Reconstitution of a functional core polycomb repressive complex. Mol Cell 8, 545–556. [DOI] [PubMed] [Google Scholar]
  63. Fuller MT, Spradling AC, 2007. Male and female Drosophila germline stem cells: two versions of immortality. Science 316, 402–404. [DOI] [PubMed] [Google Scholar]
  64. Gambetta MC, Muller J, 2014. O-GlcNAcylation prevents aggregation of the Polycomb group repressor polyhomeotic. Dev Cell 31, 629–639. [DOI] [PubMed] [Google Scholar]
  65. Gleason RJ, Anand A, Kai T, Chen X, 2018. Protecting and Diversifying the Germline. Genetics 208, 435–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Goll MG, Kirpekar F, Maggert KA, Yoder JA, Hsieh CL, Zhang X, Golic KG, Jacobsen SE, Bestor TH, 2006. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science 311, 395–398. [DOI] [PubMed] [Google Scholar]
  67. Gonczy P, DiNardo S, 1996. The germ line regulates somatic cyst cell proliferation and fate during Drosophila spermatogenesis. Development 122, 2437–2447. [DOI] [PubMed] [Google Scholar]
  68. Gonzalez J, Qi H, Liu N, Lin H, 2015. Piwi Is a Key Regulator of Both Somatic and Germline Stem Cells in the Drosophila Testis. Cell Rep 12, 150–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Grewal SI, Jia S, 2007. Heterochromatin revisited. Nat Rev Genet 8, 35–46. [DOI] [PubMed] [Google Scholar]
  70. Guo Z, Wang Z, 2009. The glypican Dally is required in the niche for the maintenance of germline stem cells and short-range BMP signaling in the Drosophila ovary. Development 136, 3627–3635. [DOI] [PubMed] [Google Scholar]
  71. Hallson G, Hollebakken RE, Li T, Syrzycka M, Kim I, Cotsworth S, Fitzpatrick KA, Sinclair DA, Honda BM, 2012. dSet1 is the main H3K4 di- and tri-methyltransferase throughout Drosophila development. Genetics 190, 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hardy RW, Tokuyasu KT, Lindsley DL, Garavito M, 1979. The germinal proliferation center in the testis of Drosophila melanogaster. J Ultrastruct Res 69, 180–190. [DOI] [PubMed] [Google Scholar]
  73. Harris RE, Pargett M, Sutcliffe C, Umulis D, Ashe HL, 2011. Brat promotes stem cell differentiation via control of a bistable switch that restricts BMP signaling. Dev Cell 20, 72–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Harrison DA, McCoon PE, Binari R, Gilman M, Perrimon N, 1998. Drosophila unpaired encodes a secreted protein that activates the JAK signaling pathway. Genes Dev 12, 3252–3263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Hassan AH, Prochasson P, Neely KE, Galasinski SC, Chandy M, Carrozza MJ, Workman JL, 2002. Function and selectivity of bromodomains in anchoring chromatin-modifying complexes to promoter nucleosomes. Cell 111, 369–379. [DOI] [PubMed] [Google Scholar]
  76. Hatfield SD, Shcherbata HR, Fischer KA, Nakahara K, Carthew RW, Ruohola-Baker H, 2005. Stem cell division is regulated by the microRNA pathway. Nature 435, 974–978. [DOI] [PubMed] [Google Scholar]
  77. He J, Xuan T, Xin T, An H, Wang J, Zhao G, Li M, 2014. Evidence for chromatin-remodeling complex PBAP-controlled maintenance of the Drosophila ovarian germline stem cells. PLoS One 9, e103473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Hennig W, Weyrich A, 2013. Histone modifications in the male germ line of Drosophila. BMC Dev Biol 13, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Hermant C, Boivin A, Teysset L, Delmarre V, Asif-Laidin A, van den Beek M, Antoniewski C, Ronsseray S, 2015. Paramutation in Drosophila Requires Both Nuclear and Cytoplasmic Actors of the piRNA Pathway and Induces Cis-spreading of piRNA Production. Genetics 201, 1381–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Herrera SC, Bach EA, 2018. JNK signaling triggers spermatogonial dedifferentiation during chronic stress to maintain the germline stem cell pool in the Drosophila testis. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Herrera SC, Bach EA, 2019. JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates. Development 146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Hirose F, Ohshima N, Kwon EJ, Yoshida H, Yamaguchi M, 2002. Drosophila Mi-2 negatively regulates dDREF by inhibiting its DNA-binding activity. Mol Cell Biol 22, 5182–5193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Hirose F, Yamaguchi M, Kuroda K, Omori A, Hachiya T, Ikeda M, Nishimoto Y, Matsukage A, 1996. Isolation and characterization of cDNA for DREF, a promoter-activating factor for Drosophila DNA replication-related genes. J Biol Chem 271, 3930–3937. [DOI] [PubMed] [Google Scholar]
  84. Hodawadekar SC, Marmorstein R, 2007. Chemistry of acetyl transfer by histone modifying enzymes: structure, mechanism and implications for effector design. Oncogene 26, 5528–5540. [DOI] [PubMed] [Google Scholar]
  85. Holowatyj A, Yang ZQ, Pile LA, 2015. Histone lysine demethylases in Drosophila melanogaster. Fly (Austin) 9, 36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Huang F, Paulson A, Dutta A, Venkatesh S, Smolle M, Abmayr SM, Workman JL, 2014. Histone acetyltransferase Enok regulates oocyte polarization by promoting expression of the actin nucleation factor spire. Genes Dev 28, 2750–2763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Hudson JB, Podos SD, Keith K, Simpson SL, Ferguson EL, 1998. The Drosophila Medea gene is required downstream of dpp and encodes a functional homolog of human Smad4. Development 125, 1407–1420. [DOI] [PubMed] [Google Scholar]
  88. Issigonis M, Tulina N, de Cuevas M, Brawley C, Sandler L, Matunis E, 2009. JAK-STAT signal inhibition regulates competition in the Drosophila testis stem cell niche. Science 326, 153–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. James TC, Eissenberg JC, Craig C, Dietrich V, Hobson A, Elgin SC, 1989. Distribution patterns of HP1, a heterochromatin-associated nonhistone chromosomal protein of Drosophila. Eur J Cell Biol 50, 170–180. [PubMed] [Google Scholar]
  90. James TC, Elgin SC, 1986. Identification of a nonhistone chromosomal protein associated with heterochromatin in Drosophila melanogaster and its gene. Mol Cell Biol 6, 3862–3872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Jin Z, Xie T, 2007. Dcr-1 maintains Drosophila ovarian stem cells. Curr Biol 17, 539–544. [DOI] [PubMed] [Google Scholar]
  92. Kahney EW, Snedeker JC, Chen X, 2019. Regulation of Drosophila germline stem cells. Curr Opin Cell Biol 60, 27–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kai T, Spradling A, 2004. Differentiating germ cells can revert into functional stem cells in Drosophila melanogaster ovaries. Nature 428, 564–569. [DOI] [PubMed] [Google Scholar]
  94. Kalb R, Latwiel S, Baymaz HI, Jansen PW, Muller CW, Vermeulen M, Muller J, 2014. Histone H2A monoubiquitination promotes histone H3 methylation in Polycomb repression. Nat Struct Mol Biol 21, 569–571. [DOI] [PubMed] [Google Scholar]
  95. Kassis JA, Kennison JA, Tamkun JW, 2017. Polycomb and Trithorax Group Genes in Drosophila. Genetics 206, 1699–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Kawase E, Wong MD, Ding BC, Xie T, 2004. Gbb/Bmp signaling is essential for maintaining germline stem cells and for repressing bam transcription in the Drosophila testis. Development 131, 1365–1375. [DOI] [PubMed] [Google Scholar]
  97. Ketel CS, Andersen EF, Vargas ML, Suh J, Strome S, Simon JA, 2005. Subunit contributions to histone methyltransferase activities of fly and worm polycomb group complexes. Mol Cell Biol 25, 6857–6868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Kiger AA, Jones DL, Schulz C, Rogers MB, Fuller MT, 2001. Stem cell self-renewal specified by JAK-STAT activation in response to a support cell cue. Science 294, 2542–2545. [DOI] [PubMed] [Google Scholar]
  99. Kiger AA, White-Cooper H, Fuller MT, 2000. Somatic support cells restrict germline stem cell self-renewal and promote differentiation. Nature 407, 750–754. [DOI] [PubMed] [Google Scholar]
  100. King IF, Emmons RB, Francis NJ, Wild B, Muller J, Kingston RE, Wu CT, 2005. Analysis of a polycomb group protein defines regions that link repressive activity on nucleosomal templates to in vivo function. Mol Cell Biol 25, 6578–6591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Kobor MS, Venkatasubrahmanyam S, Meneghini MD, Gin JW, Jennings JL, Link AJ, Madhani HD, Rine J, 2004. A protein complex containing the conserved Swi2/Snf2-related ATPase Swr1p deposits histone variant H2A.Z into euchromatin. PLoS Biol 2, E131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Kornberg T, 1981. Engrailed: a gene controlling compartment and segment formation in Drosophila. Proc Natl Acad Sci U S A 78, 1095–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Kusch T, Florens L, Macdonald WH, Swanson SK, Glaser RL, Yates JR 3rd, Abmayr SM, Washburn MP, Workman JL, 2004. Acetylation by Tip60 is required for selective histone variant exchange at DNA lesions. Science 306, 2084–2087. [DOI] [PubMed] [Google Scholar]
  104. Lachner M, O’Carroll D, Rea S, Mechtler K, Jenuwein T, 2001. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116–120. [DOI] [PubMed] [Google Scholar]
  105. Lake CM, Holsclaw JK, Bellendir SP, Sekelsky J, Hawley RS, 2013. The development of a monoclonal antibody recognizing the Drosophila melanogaster phosphorylated histone H2A variant (gamma-H2AV). G3 (Bethesda) 3, 1539–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Law JA, Jacobsen SE, 2010. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat Rev Genet 11, 204–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Le Thomas A, Rogers AK, Webster A, Marinov GK, Liao SE, Perkins EM, Hur JK, Aravin AA, Toth KF, 2013. Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state. Genes Dev 27, 390–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Leatherman JL, Dinardo S, 2008. Zfh-1 controls somatic stem cell self-renewal in the Drosophila testis and nonautonomously influences germline stem cell self-renewal. Cell Stem Cell 3, 44–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Leatherman JL, Dinardo S, 2010. Germline self-renewal requires cyst stem cells and stat regulates niche adhesion in Drosophila testes. Nat Cell Biol 12, 806–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Lee N, Zhang J, Klose RJ, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y, 2007. The trithorax-group protein Lid is a histone H3 trimethyl-Lys4 demethylase. Nat Struct Mol Biol 14, 341–343. [DOI] [PubMed] [Google Scholar]
  111. Li X, Yang F, Chen H, Deng B, Li X, Xi R, 2016. Control of germline stem cell differentiation by Polycomb and Trithorax group genes in the niche microenvironment. Development 143, 3449–3458. [DOI] [PubMed] [Google Scholar]
  112. Li Y, Ma Q, Cherry CM, Matunis EL, 2014. Steroid signaling promotes stem cell maintenance in the Drosophila testis. Dev Biol 394, 129–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Li Y, Maines JZ, Tastan OY, McKearin DM, Buszczak M, 2012. Mei-P26 regulates the maintenance of ovarian germline stem cells by promoting BMP signaling. Development 139, 1547–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Li Y, Minor NT, Park JK, McKearin DM, Maines JZ, 2009. Bam and Bgcn antagonize Nanos-dependent germ-line stem cell maintenance. Proc Natl Acad Sci U S A 106, 9304–9309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Li Y, Zhang Q, Carreira-Rosario A, Maines JZ, McKearin DM, Buszczak M, 2013. Mei-p26 cooperates with Bam, Bgcn and Sxl to promote early germline development in the Drosophila ovary. PLoS One 8, e58301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Lim AK, Tao L, Kai T, 2009. piRNAs mediate posttranscriptional retroelement silencing and localization to pi-bodies in the Drosophila germline. J Cell Biol 186, 333–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Lim C, Gandhi S, Biniossek ML, Feng L, Schilling O, Urban S, Chen X, 2015. An Aminopeptidase in the Drosophila Testicular Niche Acts in Germline Stem Cell Maintenance and Spermatogonial Dedifferentiation. Cell Rep 13, 315–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Liu M, Lim TM, Cai Y, 2010. The Drosophila female germline stem cell lineage acts to spatially restrict DPP function within the niche. Sci Signal 3, ra57. [DOI] [PubMed] [Google Scholar]
  119. Liu Z, Zhong G, Chai PC, Luo L, Liu S, Yang Y, Baeg GH, Cai Y, 2015. Coordinated niche-associated signals promote germline homeostasis in the Drosophila ovary. J Cell Biol 211, 469–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Lo SM, Ahuja NK, Francis NJ, 2009. Polycomb group protein Suppressor 2 of zeste is a functional homolog of Posterior Sex Combs. Mol Cell Biol 29, 515–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Loza-Coll MA, Petrossian CC, Boyle ML, Jones DL, 2019. Heterochromatin Protein 1 (HP1) inhibits stem cell proliferation induced by ectopic activation of the Jak/STAT pathway in the Drosophila testis. Exp Cell Res 377, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Luo GZ, Blanco MA, Greer EL, He C, Shi Y, 2015. DNA N(6)-methyladenine: a new epigenetic mark in eukaryotes? Nat Rev Mol Cell Biol 16, 705–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Maines JZ, Park JK, Williams M, McKearin DM, 2007. Stonewalling Drosophila stem cell differentiation by epigenetic controls. Development 134, 1471–1479. [DOI] [PubMed] [Google Scholar]
  124. Marfella CG, Imbalzano AN, 2007. The Chd family of chromatin remodelers. Mutat Res 618, 30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Margueron R, Justin N, Ohno K, Sharpe ML, Son J, Drury WJ 3rd, Voigt P, Martin SR, Taylor WR, De Marco V, Pirrotta V, Reinberg D, Gamblin SJ, 2009. Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461, 762–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Matunis EL, Stine RR, de Cuevas M, 2012. Recent advances in Drosophila male germline stem cell biology. Spermatogenesis 2, 137–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. McCarthy A, Deiulio A, Martin ET, Upadhyay M, Rangan P, 2018. Tip60 complex promotes expression of a differentiation factor to regulate germline differentiation in female Drosophila. Mol Biol Cell 29, 2933–2945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. McKearin D, Ohlstein B, 1995. A role for the Drosophila bag-of-marbles protein in the differentiation of cystoblasts from germline stem cells. Development 121, 2937–2947. [DOI] [PubMed] [Google Scholar]
  129. McKearin DM, Spradling AC, 1990. bag-of-marbles: a Drosophila gene required to initiate both male and female gametogenesis. Genes Dev 4, 2242–2251. [DOI] [PubMed] [Google Scholar]
  130. Messmer S, Franke A, Paro R, 1992. Analysis of the functional role of the Polycomb chromo domain in Drosophila melanogaster. Genes Dev 6, 1241–1254. [DOI] [PubMed] [Google Scholar]
  131. Mizuguchi G, Shen X, Landry J, Wu WH, Sen S, Wu C, 2004. ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex. Science 303, 343–348. [DOI] [PubMed] [Google Scholar]
  132. Mohan M, Herz HM, Smith ER, Zhang Y, Jackson J, Washburn MP, Florens L, Eissenberg JC, Shilatifard A, 2011. The COMPASS family of H3K4 methylases in Drosophila. Mol Cell Biol 31, 4310–4318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Mohrmann L, Langenberg K, Krijgsveld J, Kal AJ, Heck AJ, Verrijzer CP, 2004. Differential targeting of two distinct SWI/SNF-related Drosophila chromatin-remodeling complexes. Mol Cell Biol 24, 3077–3088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Morata G, Lawrence PA, 1975. Control of compartment development by the engrailed gene in Drosophila. Nature 255, 614–617. [DOI] [PubMed] [Google Scholar]
  135. Morillo Prado JR, Chen X, Fuller MT, 2012. Polycomb group genes Psc and Su(z)2 maintain somatic stem cell identity and activity in Drosophila. PLoS One 7, e52892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Morillo Prado JR, Srinivasan S, Fuller MT, 2013. The histone variant His2Av is required for adult stem cell maintenance in the Drosophila testis. PLoS Genet 9, e1003903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Moshkin YM, Mohrmann L, van Ijcken WF, Verrijzer CP, 2007. Functional differentiation of SWI/SNF remodelers in transcription and cell cycle control. Mol Cell Biol 27, 651–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Muller J, Hart CM, Francis NJ, Vargas ML, Sengupta A, Wild B, Miller EL, O’Connor MB, Kingston RE, Simon JA, 2002. Histone methyltransferase activity of a Drosophila Polycomb group repressor complex. Cell 111, 197–208. [DOI] [PubMed] [Google Scholar]
  139. Narlikar GJ, Sundaramoorthy R, Owen-Hughes T, 2013. Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes. Cell 154, 490–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Nystul T, Spradling A, 2007. An epithelial niche in the Drosophila ovary undergoes long-range stem cell replacement. Cell Stem Cell 1, 277–285. [DOI] [PubMed] [Google Scholar]
  141. Pancratov R, Peng F, Smibert P, Yang S Jr., Olson ER, Guha-Gilford C, Kapoor AJ, Liang FX, Lai EC, Flaherty MS, DasgGupta R, 2013. The miR-310/13 cluster antagonizes beta-catenin function in the regulation of germ and somatic cell differentiation in the Drosophila testis. Development 140, 2904–2916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Park JK, Liu X, Strauss TJ, McKearin DM, Liu Q, 2007. The miRNA pathway intrinsically controls self-renewal of Drosophila germline stem cells. Curr Biol 17, 533–538. [DOI] [PubMed] [Google Scholar]
  143. Parrott BB, Hudson A, Brady R, Schulz C, 2012. Control of germline stem cell division frequency--a novel, developmentally regulated role for epidermal growth factor signaling. PLoS One 7, e36460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Peng JC, Valouev A, Liu N, Lin H, 2016. Piwi maintains germline stem cells and oogenesis in Drosophila through negative regulation of Polycomb group proteins. Nat Genet 48, 283–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Perrini B, Piacentini L, Fanti L, Altieri F, Chichiarelli S, Berloco M, Turano C, Ferraro A, Pimpinelli S, 2004. HP1 controls telomere capping, telomere elongation, and telomere silencing by two different mechanisms in Drosophila. Mol Cell 15, 467–476. [DOI] [PubMed] [Google Scholar]
  146. Phalke S, Nickel O, Walluscheck D, Hortig F, Onorati MC, Reuter G, 2009. Retrotransposon silencing and telomere integrity in somatic cells of Drosophila depends on the cytosine-5 methyltransferase DNMT2. Nat Genet 41, 696–702. [DOI] [PubMed] [Google Scholar]
  147. Raddatz G, Guzzardo PM, Olova N, Fantappie MR, Rampp M, Schaefer M, Reik W, Hannon GJ, Lyko F, 2013. Dnmt2-dependent methylomes lack defined DNA methylation patterns. Proc Natl Acad Sci U S A 110, 8627–8631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Ranjan R, Snedeker J, Chen X, 2019. Asymmetric Centromeres Differentially Coordinate with Mitotic Machinery to Ensure Biased Sister Chromatid Segregation in Germline Stem Cells. Cell Stem Cell 25, 666–681 e665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Rojas-Rios P, Guerrero I, Gonzalez-Reyes A, 2012. Cytoneme-mediated delivery of hedgehog regulates the expression of bone morphogenetic proteins to maintain germline stem cells in Drosophila. PLoS Biol 10, e1001298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Rudolph T, Yonezawa M, Lein S, Heidrich K, Kubicek S, Schafer C, Phalke S, Walther M, Schmidt A, Jenuwein T, Reuter G, 2007. Heterochromatin formation in Drosophila is initiated through active removal of H3K4 methylation by the LSD1 homolog SU(VAR)3–3. Mol Cell 26, 103–115. [DOI] [PubMed] [Google Scholar]
  151. Ruhf ML, Braun A, Papoulas O, Tamkun JW, Randsholt N, Meister M, 2001. The domino gene of Drosophila encodes novel members of the SWI2/SNF2 family of DNA-dependent ATPases, which contribute to the silencing of homeotic genes. Development 128, 1429–1441. [DOI] [PubMed] [Google Scholar]
  152. Saito K, Nishida KM, Mori T, Kawamura Y, Miyoshi K, Nagami T, Siomi H, Siomi MC, 2006. Specific association of Piwi with rasiRNAs derived from retrotransposon and heterochromatic regions in the Drosophila genome. Genes Dev 20, 2214–2222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Salzmann V, Inaba M, Cheng J, Yamashita YM, 2013. Lineage tracing quantification reveals symmetric stem cell division in Drosophila male germline stem cells. Cell Mol Bioeng 6, 441–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Schaefer M, Pollex T, Hanna K, Tuorto F, Meusburger M, Helm M, Lyko F, 2010. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev 24, 1590–1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Schirling C, Heseding C, Heise F, Kesper D, Klebes A, Klein-Hitpass L, Vortkamp A, Hoffmann D, Saumweber H, Ehrenhofer-Murray AE, 2010. Widespread regulation of gene expression in the Drosophila genome by the histone acetyltransferase dTip60. Chromosoma 119, 99–113. [DOI] [PubMed] [Google Scholar]
  156. Schotta G, Ebert A, Krauss V, Fischer A, Hoffmann J, Rea S, Jenuwein T, Dorn R, Reuter G, 2002. Central role of Drosophila SU(VAR)3–9 in histone H3-K9 methylation and heterochromatic gene silencing. EMBO J 21, 1121–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Schotta G, Ebert A, Reuter G, 2003. SU(VAR)3–9 is a conserved key function in heterochromatic gene silencing. Genetica 117, 149–158. [DOI] [PubMed] [Google Scholar]
  158. Schulz C, Wood CG, Jones DL, Tazuke SI, Fuller MT, 2002. Signaling from germ cells mediated by the rhomboid homolog stet organizes encapsulation by somatic support cells. Development 129, 4523–4534. [DOI] [PubMed] [Google Scholar]
  159. Schwedes CC, Carney GE, 2012. Ecdysone signaling in adult Drosophila melanogaster. J Insect Physiol 58, 293–302. [DOI] [PubMed] [Google Scholar]
  160. Shcherbata HR, 2019. miRNA functions in stem cells and their niches: lessons from the Drosophila ovary. Curr Opin Insect Sci 31, 29–36. [DOI] [PubMed] [Google Scholar]
  161. Shen X, Mizuguchi G, Hamiche A, Wu C, 2000. A chromatin remodelling complex involved in transcription and DNA processing. Nature 406, 541–544. [DOI] [PubMed] [Google Scholar]
  162. Sheng XR, Brawley CM, Matunis EL, 2009. Dedifferentiating spermatogonia outcompete somatic stem cells for niche occupancy in the Drosophila testis. Cell Stem Cell 5, 191–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Sheng XR, Matunis E, 2011. Live imaging of the Drosophila spermatogonial stem cell niche reveals novel mechanisms regulating germline stem cell output. Development 138, 3367–3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Smith ER, Lee MG, Winter B, Droz NM, Eissenberg JC, Shiekhattar R, Shilatifard A, 2008. Drosophila UTX is a histone H3 Lys27 demethylase that colocalizes with the elongating form of RNA polymerase II. Mol Cell Biol 28, 1041–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Song X, Wong MD, Kawase E, Xi R, Ding BC, McCarthy JJ, Xie T, 2004. Bmp signals from niche cells directly repress transcription of a differentiation-promoting gene, bag of marbles, in germline stem cells in the Drosophila ovary. Development 131, 1353–1364. [DOI] [PubMed] [Google Scholar]
  166. Song X, Xie T, 2002. DE-cadherin-mediated cell adhesion is essential for maintaining somatic stem cells in the Drosophila ovary. Proc Natl Acad Sci U S A 99, 14813–14818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Song X, Zhu CH, Doan C, Xie T, 2002. Germline stem cells anchored by adherens junctions in the Drosophila ovary niches. Science 296, 1855–1857. [DOI] [PubMed] [Google Scholar]
  168. Spradling A, Fuller MT, Braun RE, Yoshida S, 2011. Germline stem cells. Cold Spring Harb Perspect Biol 3, a002642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Streit A, Bernasconi L, Sergeev P, Cruz A, Steinmann-Zwicky M, 2002. mgm 1, the earliest sex-specific germline marker in Drosophila, reflects expression of the gene esg in male stem cells. Int J Dev Biol 46, 159–166. [PubMed] [Google Scholar]
  170. Sun J, Wei HM, Xu J, Chang JF, Yang Z, Ren X, Lv WW, Liu LP, Pan LX, Wang X, Qiao HH, Zhu B, Ji JY, Yan D, Xie T, Sun FL, Ni JQ, 2015. Histone H1-mediated epigenetic regulation controls germline stem cell self-renewal by modulating H4K16 acetylation. Nat Commun 6, 8856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Suzuki MM, Bird A, 2008. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet 9, 465–476. [DOI] [PubMed] [Google Scholar]
  172. Swaminathan J, Baxter EM, Corces VG, 2005. The role of histone H2Av variant replacement and histone H4 acetylation in the establishment of Drosophila heterochromatin. Genes Dev 19, 65–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Takayama S, Dhahbi J, Roberts A, Mao G, Heo SJ, Pachter L, Martin DI, Boffelli D, 2014. Genome methylation in D. melanogaster is found at specific short motifs and is independent of DNMT2 activity. Genome Res 24, 821–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Talbert PB, Henikoff S, 2017. Histone variants on the move: substrates for chromatin dynamics. Nat Rev Mol Cell Biol 18, 115–126. [DOI] [PubMed] [Google Scholar]
  175. Tanabe M, Kouzmenko AP, Ito S, Sawatsubashi S, Suzuki E, Fujiyama S, Yamagata K, Zhao Y, Kimura S, Ueda T, Murata T, Matsukawa H, Takeyama K, Kato S, 2008. Activation of facultatively silenced Drosophila loci associates with increased acetylation of histone H2AvD. Genes Cells 13, 1279–1288. [DOI] [PubMed] [Google Scholar]
  176. Tarayrah L, Herz HM, Shilatifard A, Chen X, 2013. Histone demethylase dUTX antagonizes JAK-STAT signaling to maintain proper gene expression and architecture of the Drosophila testis niche. Development 140, 1014–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Tarayrah L, Li Y, Gan Q, Chen X, 2015. Epigenetic regulator Lid maintains germline stem cells through regulating JAK-STAT signaling pathway activity. Biol Open 4, 1518–1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Terry NA, Tulina N, Matunis E, DiNardo S, 2006. Novel regulators revealed by profiling Drosophila testis stem cells within their niche. Dev Biol 294, 246–257. [DOI] [PubMed] [Google Scholar]
  179. Tie F, Banerjee R, Conrad PA, Scacheri PC, Harte PJ, 2012. Histone demethylase UTX and chromatin remodeler BRM bind directly to CBP and modulate acetylation of histone H3 lysine 27. Mol Cell Biol 32, 2323–2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Tie F, Stratton CA, Kurzhals RL, Harte PJ, 2007. The N terminus of Drosophila ESC binds directly to histone H3 and is required for E(Z)-dependent trimethylation of H3 lysine 27. Mol Cell Biol 27, 2014–2026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Tran V, Lim C, Xie J, Chen X, 2012. Asymmetric division of Drosophila male germline stem cell shows asymmetric histone distribution. Science 338, 679–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Tulina N, Matunis E, 2001. Control of stem cell self-renewal in Drosophila spermatogenesis by JAK-STAT signaling. Science 294, 2546–2549. [DOI] [PubMed] [Google Scholar]
  183. Vagin VV, Sigova A, Li C, Seitz H, Gvozdev V, Zamore PD, 2006. A distinct small RNA pathway silences selfish genetic elements in the germline. Science 313, 320–324. [DOI] [PubMed] [Google Scholar]
  184. van Daal A, White EM, Gorovsky MA, Elgin SC, 1988. Drosophila has a single copy of the gene encoding a highly conserved histone H2A variant of the H2A.F/Z type. Nucleic Acids Res 16, 7487–7497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Vermaak D, Malik HS, 2009. Multiple roles for heterochromatin protein 1 genes in Drosophila. Annu Rev Genet 43, 467–492. [DOI] [PubMed] [Google Scholar]
  186. Vermeulen M, Eberl HC, Matarese F, Marks H, Denissov S, Butter F, Lee KK, Olsen JV, Hyman AA, Stunnenberg HG, Mann M, 2010. Quantitative interaction proteomics and genome-wide profiling of epigenetic histone marks and their readers. Cell 142, 967–980. [DOI] [PubMed] [Google Scholar]
  187. Voog J, Sandall SL, Hime GR, Resende LP, Loza-Coll M, Aslanian A, Yates JR 3rd, Hunter T, Fuller MT, Jones DL, 2014. Escargot restricts niche cell to stem cell conversion in the Drosophila testis. Cell Rep 7, 722–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, Zhang Y, 2004. Role of histone H2A ubiquitination in Polycomb silencing. Nature 431, 873–878. [DOI] [PubMed] [Google Scholar]
  189. Wang X, Pan L, Wang S, Zhou J, McDowell W, Park J, Haug J, Staehling K, Tang H, Xie T, 2011. Histone H3K9 trimethylase Eggless controls germline stem cell maintenance and differentiation. PLoS Genet 7, e1002426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Wang Z, Lin H, 2004. Nanos maintains germline stem cell self-renewal by preventing differentiation. Science 303, 2016–2019. [DOI] [PubMed] [Google Scholar]
  191. Wang Z, Lin H, 2007. Sex-lethal is a target of Bruno-mediated translational repression in promoting the differentiation of stem cell progeny during Drosophila oogenesis. Dev Biol 302, 160–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Wen P, Quan Z, Xi R, 2012. The biological function of the WD40 repeat-containing protein p55/Caf1 in Drosophila. Dev Dyn 241, 455–464. [DOI] [PubMed] [Google Scholar]
  193. Woodage T, Basrai MA, Baxevanis AD, Hieter P, Collins FS, 1997. Characterization of the CHD family of proteins. Proc Natl Acad Sci U S A 94, 11472–11477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Wooten M, Snedeker J, Nizami ZF, Yang X, Ranjan R, Urban E, Kim JM, Gall J, Xiao J, Chen X, 2019. Asymmetric histone inheritance via strand-specific incorporation and biased replication fork movement. Nat Struct Mol Biol 26, 732–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Wu H, Zhang Y, 2014. Reversing DNA methylation: mechanisms, genomics, and biological functions. Cell 156, 45–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Xi R, Xie T, 2005. Stem cell self-renewal controlled by chromatin remodeling factors. Science 310, 1487–1489. [DOI] [PubMed] [Google Scholar]
  197. Xie J, Wooten M, Tran V, Chen BC, Pozmanter C, Simbolon C, Betzig E, Chen X, 2015. Histone H3 Threonine Phosphorylation Regulates Asymmetric Histone Inheritance in the Drosophila Male Germline. Cell 163, 920–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Xie T, Spradling AC, 1998. decapentaplegic is essential for the maintenance and division of germline stem cells in the Drosophila ovary. Cell 94, 251–260. [DOI] [PubMed] [Google Scholar]
  199. Xie T, Spradling AC, 2000. A niche maintaining germ line stem cells in the Drosophila ovary. Science 290, 328–330. [DOI] [PubMed] [Google Scholar]
  200. Xin T, Xuan T, Tan J, Li M, Zhao G, Li M, 2013. The Drosophila putative histone acetyltransferase Enok maintains female germline stem cells through regulating Bruno and the niche. Dev Biol 384, 1–12. [DOI] [PubMed] [Google Scholar]
  201. Xing Y, Li WX, 2015. Heterochromatin components in germline stem cell maintenance. Sci Rep 5, 17463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Xu C, Bian C, Yang W, Galka M, Ouyang H, Chen C, Qiu W, Liu H, Jones AE, MacKenzie F, Pan P, Li SS, Wang H, Min J, 2010. Binding of different histone marks differentially regulates the activity and specificity of polycomb repressive complex 2 (PRC2). Proc Natl Acad Sci U S A 107, 19266–19271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Xuan T, Xin T, He J, Tan J, Gao Y, Feng S, He L, Zhao G, Li M, 2013. dBre1/dSet1-dependent pathway for histone H3K4 trimethylation has essential roles in controlling germline stem cell maintenance and germ cell differentiation in the Drosophila ovary. Dev Biol 379, 167–181. [DOI] [PubMed] [Google Scholar]
  204. Yadlapalli S, Yamashita YM, 2013. Chromosome-specific nonrandom sister chromatid segregation during stem-cell division. Nature 498, 251–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Yamashita YM, Fuller MT, 2005. Asymmetric stem cell division and function of the niche in the Drosophila male germ line. Int J Hematol 82, 377–380. [DOI] [PubMed] [Google Scholar]
  206. Yan D, Neumuller RA, Buckner M, Ayers K, Li H, Hu Y, Yang-Zhou D, Pan L, Wang X, Kelley C, Vinayagam A, Binari R, Randklev S, Perkins LA, Xie T, Cooley L, Perrimon N, 2014. A regulatory network of Drosophila germline stem cell self-renewal. Dev Cell 28, 459–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Yang F, Quan Z, Huang H, He M, Liu X, Cai T, Xi R, 2019. Ovaries absent links dLsd1 to HP1a for local H3K4 demethylation required for heterochromatic gene silencing. Elife 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Yang F, Xi R, 2017. Silencing transposable elements in the Drosophila germline. Cell Mol Life Sci 74, 435–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Yang L, Chen D, Duan R, Xia L, Wang J, Qurashi A, Jin P, Chen D, 2007. Argonaute 1 regulates the fate of germline stem cells in Drosophila. Development 134, 4265–4272. [DOI] [PubMed] [Google Scholar]
  210. Yang Z, Sun J, Hu Y, Wang F, Wang X, Qiao HH, Xu J, Mao D, Ren X, Pan LX, Xu RG, Xu BW, Zhang Y, Li H, Miao W, Hu Y, Chang Z, Wang D, Li H, Chang Z, Liu LP, Liu Q, Ni JQ, 2017. Histone H1 defect in escort cells triggers germline tumor in Drosophila ovary. Dev Biol 424, 40–49. [DOI] [PubMed] [Google Scholar]
  211. Zamparini AL, Davis MY, Malone CD, Vieira E, Zavadil J, Sachidanandam R, Hannon GJ, Lehmann R, 2011. Vreteno, a gonad-specific protein, is essential for germline development and primary piRNA biogenesis in Drosophila. Development 138, 4039–4050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Zhang S, Pan C, Lv X, Wu W, Chen H, Wu W, Wu H, Zhang L, Zhao Y, 2017. Repression of Abd-B by Polycomb is critical for cell identity maintenance in adult Drosophila testis. Sci Rep 7, 5101. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES